Journal List > Hanyang Med Rev > v.32(4) > 1044124

Park: Thyroid and Hydrogen Peroxide

Abstract

The thyroid gland produces high concentrations of hydrogen peroxide (H2O2) which is used for the biosynthesis of the thyroid hormones, triiodothyronine and thyroxine. The most important step in thyroid hormone synthesis is oxidation and organification of iodide, which itself is a very potent oxidant. To oxidize iodide, thyrocytes generate high intracellular concentrations of H2O2 using the NADPH-dependent dual oxidase enzymes (DUOX) and remove used H2O2 by various well-characterized antioxidant systems, such as those involving glutathione peroxidases and peroxiredoxins. Although thyrocytes have very intricate and efficient systems to regulate H2O2 generation and removal, leakage of H2O2 may be inevitable because of the high intracellular concentrations of H2O2 in thyrocytes. Leakage of the highly reactive oxidant H2O2 is likely to cause the damage to various intracellular proteins and DNA that has been demonstrated to be a causative factor in the induction of various diseases, such as thyroid cancer, chronic thyroiditis and hyperthyroidism. Therefore, ongoing investigation of the systems regulating H2O2 production and elimination in thyrocytesis essential for understanding the regulation and process of thyroid hormone synthesis and also for elucidating pathogenetic mechanisms underlying various autoimmune thyroid diseases and the initiation and promotion of thyroid cancers.

Figures and Tables

Fig. 1
Thyroid hormone synthesis. Adopted from the Wikipedia Commons.
hmr-32-187-g001

References

1. Song Y, Driessens N, Costa M, De Deken X, Detours V, Corvilain B, et al. Roles of hydrogen peroxide in thyroid physiology and disease. J Clin Endocrinol Metab. 2007. 92:3764–3773.
crossref
2. Ahn CS, Rosenberg IN. Fellinger K, Höfer R, editors. Oxidation of 14C-formate in thyroid slices: effects of TSH, dibutyric cyclic 3',5'-AMP (dbc-AMP) and prostaglandin E1 (PGE1). Futher advances in thyroid research. 1971. Viena: Verlag der Wiener Medizinischen Akademle;825–837.
3. Bénard B, Brault J. Production of peroxide in the thyroid. Union Med Can. 1971. 100:701–705.
4. Stone JR. An assessment of proposed mechanisms for sensing hydrogen peroxide in mammalian systems. Arch Biochem Biophys. 2004. 422:119–124.
crossref
5. Quinn MT, Ammons MC, Deleo FR. The expanding role of NADPH oxidases in health and disease: no longer just agents of death and destruction. Clin Sci (Lond). 2006. 111:1–20.
crossref
6. Rhee SG, Kang SW, Jeong W, Chang TS, Yang KS, Woo HA. Intracellular messenger function of hydrogen peroxide and its regulation by peroxiredoxins. Curr Opin Cell Biol. 2005. 17:183–189.
crossref
7. Groemping Y, Rittinger K. Activation and assembly of the NADPH oxidase: a structural perspective. Biochem J. 2005. 386(Pt 3):401–406.
crossref
8. Gorzalczany Y, Sigal N, Itan M, Lotan O, Pick E. Targeting of Rac1 to the phagocyte membrane is sufficient for the induction of NADPH oxidase assembly. J Biol Chem. 2000. 275:40073–40081.
crossref
9. Carrasco C, Kopp P. Braverman LE, Utiger RD, editors. Thyroid hormone synthesis. Werner and Ingbar's the thyroid: a fundamental and clinical text. 2005. 9th ed. Philadelphia: Lippincott Williams & Wilkins;37–76.
10. De Deken X, Wang D, Many MC, Costagliola S, Libert F, Vassart G, et al. Cloning of two human thyroid cDNAs encoding new members of the NADPH oxidase family. J Biol Chem. 2000. 275:23227–23233.
crossref
11. Dupuy C, Ohayon R, Valent A, Noel-Hudson MS, Deme D, Virion A. Purification of a novel flavoprotein involved in the thyroid NADPH oxidase. Cloning of the porcine and human cDNAs. J Biol Chem. 1999. 274:37265–37269.
crossref
12. Pfarr N, Korsch E, Kaspers S, Herbst A, Stach A, Zimmer C, et al. Congenital hypothyroidism caused by new mutations in the thyroid oxidase 2 (THOX2) gene. Clin Endocrinol (Oxf). 2006. 65:810–815.
crossref
13. Moreno JC, Bikker H, Kempers MJ, van Trotsenburg AS, Baas F, de Vijlder JJ, et al. Inactivating mutations in the gene for thyroid oxidase 2 (THOX2) and congenital hypothyroidism. N Engl J Med. 2002. 347:95–102.
crossref
14. Ris-Stalpers C. Physiology and pathophysiology of the DUOXes. Antioxid Redox Signal. 2006. 8:1563–1572.
crossref
15. Rigutto S, Hoste C, Grasberger H, Milenkovic M, Communi D, Dumont JE, et al. Activation of dual oxidases (duox1 and duox2): differential regulation mediated by PKA and PKC-dependent phosphorylation. J Biol Chem. 2009. 284:6725–6734.
16. Dupuy C, Kaniewski J, Deme D, Pommier J, Virion A. NADPH-dependent H2O2 generation catalyzed by thyroid plasma membranes. Studies with electron scavengers. Eur J Biochem. 1989. 185:597–603.
crossref
17. Wang D, De Deken X, Milenkovic M, Song Y, Pirson I, Dumont JE, et al. Identification of a novel partner of duox: EFP1, a thioredoxin-related protein. J Biol Chem. 2005. 280:3096–3103.
18. Flohé L, Ursini F. Peroxidase: a term of many meanings. Antioxid Redox Signal. 2008. 10:1485–1490.
crossref
19. Kim K, Kim IH, Lee KY, Rhee SG, Stadtman ER. The isolation and purification of a specific "protector" protein which inhibits enzyme inactivation by a thiol/Fe(III)/O2 mixed-function oxidation system. J Biol Chem. 1988. 263:4704–4711.
crossref
20. Rhee SG, Chae HZ, Kim K. Peroxiredoxins: a historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling. Free Radic Biol Med. 2005. 38:1543–1552.
crossref
21. Park JH. Cancer Facts & Figures. 2012. Seoul: National Cancer Center, Minestry of Health & Welfare;6–15.
22. Detours V, Delys L, Libert F, Weiss Solís D, Bogdanova T, Dumont JE, et al. Genome-wide gene expression profiling suggests distinct radiation susceptibilities in sporadic and post-Chernobyl papillary thyroid cancers. Br J Cancer. 2007. 97:818–825.
crossref
23. Xing M. Oxidative stress: a new risk factor for thyroid cancer. Endocr Relat Cancer. 2012. 19:C7–C11.
crossref
24. Sharma R, Traore K, Trush MA, Rose NR, Burek CL. Intracellular adhesion molecule-1 up-regulation on thyrocytes by iodine of non-obese diabetic H2(h4) mice is reactive oxygen species-dependent. Clin Exp Immunol. 2008. 152:13–20.
crossref
25. Burek CL, Rose NR. Autoimmune thyroiditis and ROS. Autoimmun Rev. 2008. 7:530–537.
crossref
26. Duntas LH, Mantzou E, Koutras DA. Effects of a six month treatment with selenomethionine in patients with autoimmune thyroiditis. Eur J Endocrinol. 2003. 148:389–393.
crossref
27. Gartner R, Gasnier BC, Dietrich JW, Krebs B, Angstwurm MW. Selenium supplementation in patients with autoimmune thyroiditis decreases thyroid peroxidase antibodies concentrations. J Clin Endocrinol Metab. 2002. 87:1687–1691.
crossref
28. Turker O, Kumanlioglu K, Karapolat I, Dogan I. Selenium treatment in autoimmune thyroiditis: 9-month followup with variable doses. J Endocrinol. 2006. 190:151–156.
crossref
29. Bjorkman U, Ekholm R. Hydrogen peroxide degradation and glutathione peroxidase activity in cultures of thyroid cells. Mol Cell Endocrinol. 1995. 111:99–107.
crossref
30. Kim H, Park S, Suh JM, Chung HK, Shong M, Kwon OY. Thyroid-stimulating hormone transcriptionally regulates the thiol-specific antioxidant gene. Cell Physiol Biochem. 2001. 11:247–252.
crossref
31. Mano T, Shinohara R, Iwase K, Kotake M, Hamada M, Uchimuro K, et al. Changes in free radical scavengers andlipid peroxide in thyroid glands of various thyroid disorders. Horm Metab Res. 1997. 29:351–354.
crossref
32. Bacic Vrca V, Skreb F, Cepelak I, Mayer L. Supplementation with antioxidants in the treatment of Graves' disease: the effect on the extracellular antioxidative parameters. Acta Pharm. 2004. 54:79–89.
TOOLS
Similar articles