TSH and Thyrotropic Agonists: Key Actors in Thyroid Homeostasis

Johannes W Dietrich, Gabi Landgrafe, Elisavet H Fotiadou, Johannes W Dietrich, Gabi Landgrafe, Elisavet H Fotiadou

Abstract

This paper provides the reader with an overview of our current knowledge of hypothalamic-pituitary-thyroid feedback from a cybernetic standpoint. Over the past decades we have gained a plethora of information from biochemical, clinical, and epidemiological investigation, especially on the role of TSH and other thyrotropic agonists as critical components of this complex relationship. Integrating these data into a systems perspective delivers new insights into static and dynamic behaviour of thyroid homeostasis. Explicit usage of this information with mathematical methods promises to deliver a better understanding of thyrotropic feedback control and new options for personalised diagnosis of thyroid dysfunction and targeted therapy, also by permitting a new perspective on the conundrum of the TSH reference range.

Figures

Figure 1
Figure 1
Information processing structure of the logarithmic standard model of thyroid homeostasis [105, 121].
Figure 2
Figure 2
Information processing structure of a nonlinear parametrically isomorphic model based on Michaelis-Menten kinetics, noncompetitive divisive inhibition, and pharmacokinetic data [11]. Modified with permission from [49].
Figure 3
Figure 3
SimThyr, a continuous simulation program for thyrotropic feedback control [11].
Figure 4
Figure 4
Characteristic curves of pituitary and thyroid. The area shaded in green denotes univariate reference ranges for TSH and FT4. The dashed red line denotes the pituitary's response in form of TSH incretion to varying FT4 levels; the continuous blue line represents the thyroid's response to TSH. Note that for the response curve of the thyroid—contrary to convention—the ordinate (TSH) is the independent axis, while the dependent axis is the abscissa (FT4). This uncommon notation facilitates superposition of both characteristic curves. Marked is a normal equilibrium point (also referred to as setpoint) defined by the intersection of both 50% percentiles. Response curves were calculated from percentiles for secretory capacities of pituitary (GH) and thyroid (GT) using the mathematical model displayed in Figure 2. Structure parameters were derived from a subgroup of subjects included in the NOMOTHETICOS trial [153].
Figure 5
Figure 5
Successive development of hypothyroidism as a consequence of decreasing GT. Beginning with a hypothetical “sublatent” form defined by reduced GT and still normal levels of TSH and FT4 (panel b), further steps are subclinical hypothyroidism with increased TSH levels and FT4 still in the lowest fraction of the reference region (panel c) and overt hypothyroidism where both parameters have left their reference region (panel d). See text for additional information.
Figure 6
Figure 6
Partial and complete thyrotropic insufficiency as results of nonlinear interaction of pituitary and thyroid. TSH axis is logarithmically scaled in order to zoom small values. GH values are given in percent from normal values. See text for additional information.
Figure 7
Figure 7
Computer simulation of thyrotropic adaptation in critical illness. A gradual increase of central type 2 deiodinase activity over several days with subsequent restoration to normal values has been simulated with SimThyr using the mathematical model shown in Figure 2. Note the temporarily increased TSH values after day 17 that are occasionally observed also in vivo in patients recovering from nonthyroidal illness syndrome.
Figure 8
Figure 8
Interaction of TSH, thyrotropic agonists, and thyrotropic antagonists with TSH receptor. SMLs bind to a pocket within the heptahelical transmembrane domain, while TSH, HCG, and TRAbs bind primarily to the TSHr amino-terminal ectodomain.
Figure 9
Figure 9
Comparison of conventional univariate reference ranges for TSH and FT4 (grey box) and a bihormonal reference region (green kite-like area) from nonlinear modelling of thyroid homeostasis. For more information see text and legend of Figure 4.

References

    1. Aron M. Action de la préhypophyse sur la thyroïde chez le cobaye. Comptes Rendus des Séances de la Société de Biologie. 1929;102:682–684.
    1. Crew FA, Wiesner BP. On the existence of a fourth hormone, thyreotropic in nature, of the anterior pituitary. British Medical Journal. 1930;1:777–778.
    1. Salter W. The Endocrine Function of Iodine. Cambridge, Mass, USA: Harvard University Press; 1940.
    1. Hohlweg W, Junkmann K. Die hormonal-nervöse Regulierung des Hypophysenvorderlappens [The hormonal and neurogenic regulation of the function of the anterior pituitary] Klinische Wochenschrift. 1932;11:321–323.
    1. McCullagh DR. Dual endocrine activity of the testes. Science. 1932;76(1957):19–20.
    1. Moore CR, Price D. The question of sex hormone antagonism. Proceedings of the Society For Experimental Biology and Medicine. 1930;28:38–40.
    1. Moore CR, Price D. Gonadal hormone functions, and the reciprocal influence between gonads and hypophysis with its bearing on the problem of sex hormone antagonism. American Journal of Anatomy. 1932;50:13–67.
    1. Astwood EB. Mechanism of action of antithyroid compounds. Brookhaven Symposia in Biology. 1955;7:61–73.
    1. Hoskins R. The thyroid-pituitary apparatus as a servo (feed-back) mechanism. The Journal of Clinical Endocrinology & Metabolism. 1949;9:1429–1431.
    1. Stanley MM, Astwood EB. The response of the thyroid gland in normal human subjects to the administration of thyrotropin, as shown by studies with I131. Endocrinology. 1949;44:49–60.
    1. Dietrich JW. Entwicklung Und Klinische Anwendung Eines Nichtlinearen Modells. Berlin, Germany: Logos; 2002. Der Hypophysen-Schilddrüsen-Regelkreis. Entwicklung und klinische Anwendung eines nichtlinearen Modells.
    1. Sam S, Frohman LA. Normal physiology of hypothalamic pituitary regulation. Endocrinology and Metabolism Clinics of North America. 2008;37(1):1–22.
    1. Zoeller RT, Tan SW, Tyl RW. General background on the hypothalamic-pituitary-thyroid (HPT) axis. Critical Reviews in Toxicology. 2007;37(1-2):11–53.
    1. Friesema ECH, Ganguly S, Abdalla A, Manning Fox JE, Halestrap AP, Visser TJ. Identification of monocarboxylate transporter 8 as a specific thyroid hormone transporter. The Journal of Biological Chemistry. 2003;278(41):40128–40135.
    1. Friesema ECH, Jansen J, Visser TJ. Thyroid hormone transporters. Biochemical Society Transactions. 2005;33(1):228–232.
    1. Dietrich JW, Brisseau K, Boehm BO. Resorption, Transport und Bioverfügbarkeit von Schilddrüsenhormonen [Absorption, transport and bio-availability of iodothyronines] Deutsche Medizinische Wochenschrift. 2008;133:1644–1648.
    1. Feldt-Rasmussen U, Rasmussen ÅK. Thyroid hormone transport and actions. In: KG E, Rivkees SA, Basel WK, editors. Diseases of the Thyroid in Childhood and Adolescence. Vol. 11. Karger; 2007. pp. 80–103.
    1. Refetoff S, Murata Y, Mori Y, Janssen OE, Takeda K, Hayashi Y. Thyroxine-binding globulin: organization of the gene and variants. Hormone Research. 1996;45(3–5):128–138.
    1. Gereben B, Zavacki AM, Ribich S, et al. Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling. Endocrine Reviews. 2008;29(7):898–938.
    1. Silv JE, Larsen PR. Contribution of plasma triiodothyronine and local thyroxine monodeiodination to triiodothyronine to nuclear triiodothyronine receptor saturation in pituitary, liver, and kidney of hypothyroid rats. The Journal of Clinical Investigation. 1978;61(5):1247–1259.
    1. Boelen A, Kwakkel J, Thijssen-Timmer DC, Alkemade A, Fliers E, Wiersinga WM. Simultaneous changes in central and peripheral components of the hypothalamus-pituitary-thyroid axis in lipopolysaccharide-induced acute illness in mice. Journal of Endocrinology. 2004;182(2):315–323.
    1. De Groot LJ. Non-thyroidal illness syndrome is a manifestation of hypothalamic-pituitary dysfunction, and in view of current evidence, should be treated with appropriate replacement therapies. Critical Care Clinics. 2006;22(1):57–86.
    1. Dietrich JW, Stachon A, Antic B, Klein HH, Hering S. The AQUA-FONTIS study: protocol of a multidisciplinary, cross-sectional and prospective longitudinal study for developing standardized diagnostics and classification of non-thyroidal illness syndrome. BMC Endocrine Disorders. 2008;8, article 13
    1. Van den Berghe G. Dynamic neuroendocrine responses to critical illness. Frontiers in Neuroendocrinology. 2002;23(4):370–391.
    1. Arrojo EDR, Bianco AC. Type 2 deiodinase at the crossroads of thyroid hormone action. International Journal of Biochemistry and Cell Biology. 2011;43(10):1432–1441.
    1. Bianco AC, Maia AL, da Silva WS, Christoffolete MA. Adaptive activation of thyroid hormone and energy expenditure. Bioscience Reports. 2005;25(3-4):191–208.
    1. Martinez-deMena R, Obregón MJ. Insulin increases the adrenergic stimulation of 5′ deiodinase activity and mRNA expression in rat brown adipocytes; role of MAPK and PI3K. Journal of Molecular Endocrinology. 2005;34(1):139–151.
    1. Garcia GC, Lopez-Bojorquez L, Nunez J, Valverde RC, Orozco A. 3,5-Diiodothyronine in vivo maintains euthyroidal expression of type 2 iodothyronine deiodinase, growth hormone, and thyroid hormone receptor β1 in the killifish. American Journal of Physiology. 2007;293(2):R877–R883.
    1. Thomas C, Auwerx J, Schoonjans K. Bile acids and the membrane bile acid receptor TGR5—connecting nutrition and metabolism. Thyroid. 2008;18(2):167–174.
    1. Watanabe M, Houten SM, Mataki C, et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature. 2006;439(7075):484–489.
    1. Christoffolete MA, Ribeiro R, Singru P, et al. Atypical expression of type 2 iodothyronine deiodinase in thyrotrophs explains the thyroxine-mediated pituitary thyrotropin feedback mechanism. Endocrinology. 2006;147(4):1735–1743.
    1. Brabant G, Ranft U, Ocran K, Hesch RD, von zur Mühlen A. Thyrotropin—an episodically secreted hormone. Acta Endocrinologica. 1986;112(3):315–322.
    1. Greenspan SL, Klibanski A, Schoenfeld D, Ridgway EC. Pulsatile secretion of thyrotropin in man. The Journal of Clinical Endocrinology & Metabolism. 1986;63(3):661–668.
    1. Samuels MH, Henry P, Luther M, Ridgway EC. Pulsatile TSH secretion during 48-hour continuous TRH infusions. Thyroid. 1993;3(3):201–206.
    1. Brabant G, Prank K, Ranft U, et al. Physiological regulation of circadian and pulsatile thyrotropin secretion in normal man and woman. The Journal of Clinical Endocrinology & Metabolism. 1990;70(2):403–409.
    1. Covarrubias L, Redondo JL, Vargas MA, et al. In vitro TRH release from hypothalamus slices varies during the diurnal cycle. Neurochemical Research. 1994;19(7):845–850.
    1. Franklyn JA. Drug therapy: the management of hyperthyroidism. The New England Journal of Medicine. 1994;330(24):1731–1738.
    1. Brownlie BEW, Legge HM. Thyrotropin results in euthyroid patients with a past history of hyperthyroidism. Acta Endocrinologica. 1990;122(5):623–627.
    1. Jaeschke H, Eszlinger M, Lueblinghoff J, Coslovsky R, Paschke R. Prolonged inappropriate TSH suppression during hypothyroidism after thyroid ablation in a patient with nonautoimmune familial hyperthyroidism. Hormone and Metabolic Research. 2011;43(7):500–504.
    1. Gelwane G, de Roux N, Chevenne D, Carel JC, Léger J. Pituitary-thyroid feedback in a patient with a sporadic activating thyrotropin (TSH) receptor mutation: implication that thyroid-secreted factors other than thyroid hormones contribute to serum TSH levels. The Journal of Clinical Endocrinology & Metabolism. 2009;94(8):2787–2791.
    1. Zwaveling-Soonawala N, van Trotsenburg P, Vulsma T. Central hypothyroidism in an infant born to an adequately treated mother with Graves’ disease: an effect of maternally derived thyrotrophin receptor antibodies? Thyroid. 2009;19(6):661–662.
    1. Kakita T, Laborde NP, Odell WD. Autoregulatory control of thyrotropin in rabbits. Endocrinology. 1984;114(6):2301–2305.
    1. Kakita T, Odell WD. Pituitary gland: one site of ultrashort-feedback regulation for control of thyrotropin. American Journal of Physiology. 1986;250(2):E121–E124.
    1. Prummel MF, Méduri G, Loosfeldt H. Human anterior pituitary contains a TSH-receptor. Journal of Endocrinological Investigation. 1997;20, article 27
    1. Prummel MF, Brokken LJS, Meduri G, Misrahi M, Bakker O, Wiersinga WM. Expression of the thyroid-stimulating hormone receptor in the folliculo-stellate cells of the human anterior pituitary. The Journal of Clinical Endocrinology & Metabolism. 2000;85(11):4347–4353.
    1. Brokken LJS, Bakker O, Wiersinga WM, Prummel MF. Functional thyrotropin receptor expression in the pituitary folliculo-stellate cell line TtT/GF. Experimental and Clinical Endocrinology and Diabetes. 2005;113(1):13–20.
    1. Brokken LJS, Scheenhart JWC, Wiersinga WM, Prummel MF. Suppression of serum TSH by Graves’ Ig: evidence for a functional pituitary TSH receptor. The Journal of Clinical Endocrinology & Metabolism. 2001;86(10):4814–4817.
    1. Prummel MF, Brokken LJS, Wiersinga WM. Ultra short-loop feedback control of thyrotropin secretion. Thyroid. 2004;14(10):825–829.
    1. Dietrich JW, Tesche A, Pickardt CR, Mitzdorf U. Thyrotropic feedback control: evidence for an additional ultrashort feedback loop from fractal analysis. Cybernetics and Systems. 2004;35(4):315–331.
    1. Brokken LJS, Wiersinga WM, Prummel MF. Thyrotropin receptor autoantibodies are associated with continued thyrotropin suppression in treated euthyroid Graves’ disease patients. The Journal of Clinical Endocrinology & Metabolism. 2003;88(9):4135–4138.
    1. DiStefano JJ., III Hypothalamic and rate feedback in the thyroid hormone regulation system: an hypothesis. The Bulletin of Mathematical Biophysics. 1969;31(2):233–246.
    1. Campbell HJ, George R, Harris GW. The acute effects of injection of thyrotrophic hormone or of electrical stimulation of the hypothalamus on thyroid activity. The Journal of Physiology. 1960;152:527–544.
    1. Harris GW, Woods JW. The effect of electrical stimulation of the hypothalamus or pituitary gland on thyroid activity. The Journal of Physiology. 1958;143(2):246–274.
    1. Shizume K, Matsuda K, Irie M, et al. Effect of electrical stimulation of the hypothalamus on thyroid function. Endocrinology. 1962;70:298–302.
    1. Lechan RM, Fekete C. Feedback regulation of thyrotropin-releasing hormone (TRH): mechanisms for the non-thyroidal illness syndrome. Journal of Endocrinological Investigation. 2004;27(6):105–119.
    1. Dahl GE, Evans NP, Thrun LA, Karsch FJ. A central negative feedback action of thyroid hormones on thyrotropin- releasing hormone secretion. Endocrinology. 1994;135(6):2392–2397.
    1. Rondeel JMM, de Greef WJ, van der Schoot P, Karels B, Klootwijk W, Visser TJ. Effect of thyroid status and paraventricular area lesions on the release of thyrotropin-releasing hormone and catecholamines into hypophysial portal blood. Endocrinology. 1988;123(1):523–527.
    1. Von Euler C, Holmgren B. The role of hypothalamo-hypophysial connexions in thyroid secretion. The Journal of Physiology. 1956;131(1):137–146.
    1. Von Euler C, Holmgren B. The thyroxine receptor of the thyroid-pituitary system. The Journal of Physiology. 1956;131(1):125–136.
    1. Bogdanove EM, Crabill EV. Thyroid-pituitary feedback: direct or indirect? A comparison of the effects of intrahypothalamic and intrapituitary thyroid autotransplants on piuitary thyroidectomy reactions in the rat. Endocrinology. 1961;69:581–595.
    1. Fekete C, Gereben B, Doleschall M, et al. Lipopolysaccharide induces type 2 iodothyronine deiodinase in the mediobasal hypothalamus: implications for the nonthyroidal illness syndrome. Endocrinology. 2004;145(4):1649–1655.
    1. Lechan RM, Fekete C. The TRH neuron: a hypothalamic integrator of energy metabolism. Progress in Brain Research. 2006;153:209–235.
    1. Chiamolera MI, Wondisford FE. Minireview: thyrotropin-releasing hormone and the thyroid hormone feedback mechanism. Endocrinology. 2009;150(3):1091–1096.
    1. Bauman TR, Anderson RR, Turner CW. Effects of induced mild hyperthyroidism on serum protein-bound iodine, thyroxine distribution volume, and biological half-life of thyroxine-131-I in dairy cattle. Journal of Dairy Science. 1969;52(2):245–249.
    1. Cullen MJ, Doherty GF, Ingbar SH. The effect of hypothyroidism and thyrotoxicosis on thyroxine metabolism in the rat. Endocrinology. 1973;92(4):1028–1033.
    1. Nicoloff JT, Low JC, Dussault JH, Fisher DA. Simultaneous measurement of thyroxine and triiodothyronine peripheral turnover kinetics in man. The Journal of Clinical Investigation. 1972;51(3):473–483.
    1. Odell WD, Utiger RD, Wilber JF, Condliffe PG. Estimation of the secretion rate of thyrotropin in man. The Journal of Clinical Investigation. 1967;46(6):953–959.
    1. Bruhn TO, Rondeel JMM, Jackson IMD. Thyrotropin-releasing hormone gene expression in the anterior pituitary. IV. Evidence for paracrine and autocrine regulation. Endocrinology. 1998;139(8):3416–3422.
    1. Hennemann G. Notes on the history of cellular uptake and deiodination of thyroid hormone. Thyroid. 2005;15(8):753–756.
    1. Hennemann G, Docter R, Friesema ECH, de Jong M, Krenning EP, Visser TJ. Plasma membrane transport of thyroid hormones and its role in thyroid hormone metabolism and bioavailability. Endocrine Reviews. 2001;22(4):451–476.
    1. Peeters RP, van Geyten SD, Wouters PJ, et al. Tissue thyroid hormone levels in critical illness. The Journal of Clinical Endocrinology & Metabolism. 2005;90(12):6498–6507.
    1. Thijssen-Timmer DC, Peelers RP, Wouters P, et al. Thyroid hormone receptor isoforni expression in livers of critically ill patients. Thyroid. 2007;17(2):105–112.
    1. Warner MH, Beckett GJ. Mechanisms behind the non-thyroidal illness syndrome: an update. Journal of Endocrinology. 2010;205(1):1–13.
    1. Hays MT. Thyroid hormone and the gut. Endocrine Research. 1988;14(2-3):203–224.
    1. Ciobanu L, Dumitrascu DL. Gastrointestinal motility disorders in endocrine diseases. Polskie Archiwum Medycyny Wewnetrznej. 2011;121(4):129–136.
    1. Yaylali O, Kirac S, Yilmaz M, et al. Does hypothyroidism affect gastrointestinal motility? Gastroenterology Research and Practice. 2009;2009:7 pages.529802
    1. Friedman Y, Lang M, Burke G. Inhibition of thyroid adenylate cyclase by thyroid hormone: a possible locus for the ’short-loop’ negative feedback phenomenon. Endocrinology. 1977;101(3):858–868.
    1. Goldenheim PD, Amir SM, Uchimura H, Ingbar SH. Studies of the binding of thyroid hormones and their short loop feedback effects in bovine thyroid tissue. Endocrinology. 1979;105(6):1308–1313.
    1. Rotella CM, Tanini A, Zonefrati R, Toccafondi R. Interaction between iodothyronines and thyrotropin receptor in human cultured thyroid cells. Journal of Receptor Research. 1981;2(4):367–382.
    1. Yu S, Friedman Y, Richman R, Burke G. Altered thyroidal responsivity to thyrotropin induced by circulating thyroid hormones. A “short loop” regulatory mechanism? The Journal of Clinical Investigation. 1976;57(3):745–755.
    1. Baenziger JU, Green ED. Pituitary glucoprotein hormone oligosaccharides: structure, synthesis and function of the asparagine-linked oligosaccharides on lutropin, follitropin and thyrotropin. Biochimica et Biophysica Acta. 1988;947(2):287–306.
    1. Donadio S, Pascual A, Thijssen JHH, Ronin C. Feasibility study of new calibrators for thyroid-stimulating hormone (TSH) immunoprocedures based on remodeling of recombinant TSH to mimic glycoforms circulating in patients with thyroid disorders. Clinical Chemistry. 2006;52(2):286–297.
    1. Gesundheit N, Magner JA, Chen T, Weintraub BD. Differential sulfation and sialylation of secreted mouse thyrotropin (TSH) subunits: regulation by TSH-releasing hormone. Endocrinology. 1986;119(2):455–463.
    1. Persani L. Hypothalamic thyrotropin-releasing hormone and thyrotropin biological activity. Thyroid. 1998;8(10):941–946.
    1. Persani L, Borgato S, Romoli R, Asteria C, Pizzocaro A, Beck-Peccoz P. Changes in the degree of sialylation of carbohydrate chains modify the biological properties of circulating thyrotropin isoforms in various physiological and pathological states. The Journal of Clinical Endocrinology & Metabolism. 1998;83(7):2486–2492.
    1. Saji M, Ikuyama S, Akamizu T, Kohn LD. Increases in cytosolic Ca++ down regulate thyrotropin receptor gene expression by a mechanism different from the cAMP signal. Biochemical and Biophysical Research Communications. 1991;176(1):94–101.
    1. Gyves PW, Gesundheit N, Thotakura NR, Stannard BS, DeCherney GS, Weintraub BD. Changes in the sialylation and sulfation of secreted thyrotropin in congenital hypothyroidism. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(10):3792–3796.
    1. Trojan J, Theodoropoulou M, Usadel KH, Stalla GK, Schaaf L. Modulation of human thyrotropin oligosaccharide structures—enhanced proportion of sialylated and terminally galactosylated serum thyrotropin isoforms in subclinical and overt primary hypothyroidism. Journal of Endocrinology. 1998;158(3):359–365.
    1. Magner J, Roy P, Fainter L, Barnard V, Fletcher P., Jr. Transiently decreased sialylation of thyrotropin (TSH) in a patient with the euthyroid sick syndrome. Thyroid. 1997;7(1):55–61.
    1. Persani L, Ferretti E, Borgato S, Faglia G, Beck-Peccoz P. Circulating thyrotropin bioactivity in sporadic central hypothyroidism. The Journal of Clinical Endocrinology & Metabolism. 2000;85(10):3631–3635.
    1. Morell AG, Gregoriadis G, Scheinberg IH, Hickman J, Ashwell G. The role of sialic acid in determining the survival of glycoproteins in the circulation. The Journal of Biological Chemistry. 1971;246(5):1461–1467.
    1. Frenzel R, Krohn K, Eszlinger M, Tönjes A, Paschke R. Sialylation of human thyrotropin receptor improves and prolongs its cell-surface expression. Molecular Pharmacology. 2005;68(4):1106–1113.
    1. Danziger L, Elmergreen GL. The thyroid-pituitary homeostatic mechanism. The Bulletin of Mathematical Biophysics. 1956;18(1):1–13.
    1. Roston S. Mathematical representation of some endocrinological systems. The Bulletin of Mathematical Biophysics. 1959;21(3):271–282.
    1. Norwich KH, Reiter R. Homeostatic control of thyroxin concentration expressed by a set of linear differential equations. The Bulletin of Mathematical Biophysics. 1965;27(2):133–144.
    1. DiStefano JJ, Stear EB. Neuroendocrine control of thyroid secretion in living systems: a feedback control system model. The Bulletin of Mathematical Biophysics. 1968;30(1):3–26.
    1. DiStefano JJ. A model of the normal thyroid hormone glandular secretion mechanism. Journal of Theoretical Biology. 1969;22(3):412–417.
    1. DiStefano JJ, III, Chang RF. Computer simulation of thyroid hormone binding, distribution, and disposal dynamics in man. The American Journal of Physiology. 1971;221(5):1529–1544.
    1. DiStefano JJ, Stear EB. Neuroendocrine control of thyroid secretion in living systems: a feedback control system model. The Bulletin of Mathematical Biophysics. 1968;30(1):3–26.
    1. Sudova K, Langer P. Iodine kinetics and hypothalamo pituitary regulation of thyroid function following its short term inhibition: simulation with the aid of compartmental system modelling (COSMO) Computers and Biomedical Research. 1975;8(1):88–96.
    1. Saratchandran P, Carson ER, Reeve J. An improved mathematical model of human thyroid hormone regulation. Clinical Endocrinology. 1976;5(5):473–483.
    1. Seif FJ. Mathematical model of pituitary thyrotropic function. Experientia. 1977;33(9):1243–1245.
    1. Wilkin TJ, Storey BE, Isles TE, Crooks J, Swanson Beck J. High TSH concentrations in “euthyroidism”: explanation based on control loop theory. British Medical Journal. 1977;1(6067):993–996.
    1. Hatakeyama T, Yagi H. Computer simulation for hormones related to primary thyropathy. Biological Cybernetics. 1985;52(4):259–266.
    1. Cohen JL. Thyroid-stimulation hormone and its disorders. In: Becker KL, editor. Principles and Practice of Endocrinology and Metabolism. Philadelphia, Pa, USA: J. B. Lippincott Company; 1990. pp. 144–152.
    1. Li G, Liu B, Liu Y. A dynamical model of the pulsatile secretion of the hypothalamo-pituitary-thyroid axis. BioSystems. 1995;35(1):83–92.
    1. Liu Y, Liu B, Xie J, Liu YX. A new mathematical model of hypothalamo-pituitary-thyroid axis. Mathematical and Computer Modelling. 1994;19(9):81–90.
    1. Dietrich JW, Mitzdorf U, Weitkunat R, Pickardt CR. The pituitary-thyroid feedback control: stability and oscillations in a new nonlinear model. Journal of Endocrinological Investigation. 1997;20(5)
    1. Falaschi P, Martocchia A, Proietti A, et al. The hypothalamic-pituitary-thyroid axis in subjects with subclinical thyroid diseases: the impact of the negative feedback mechanism. Neuroendocrinology Letters. 2004;25(4):292–296.
    1. Degon M, Chipkin SR, Hollot CV, Zoeller RT, Chait Y. A computational model of the human thyroid. Mathematical Biosciences. 2008;212(1):22–53.
    1. Leow MKS. A mathematical model of pituitary-thyroid interaction to provide an insight into the nature of the thyrotropin-thyroid hormone relationship. Journal of Theoretical Biology. 2007;248(2):275–287.
    1. Mclanahan ED, Andersen ME, Fisher JW. A biologically based dose-response model for dietary iodide and the hypothalamic-pituitary-thyroid axis in the adult rat: evaluation of iodide deficiency. Toxicological Sciences. 2008;102(2):241–253.
    1. Eisenberg M, Samuels M, DiStefano JJ., III Extensions, validation, and clinical applications of a feedback control system simulator of the hypothalamo-pituitary-thyroid axis. Thyroid. 2008;18(10):1071–1085.
    1. Eisenberg MC, Santini F, Marsili A, Pinchera A, DiStefano JJ., III TSH regulation dynamics in central and extreme primary hypothyroidism. Thyroid. 2010;20(11):1215–1228.
    1. Panda JN, Turner CW. Thyroxine feed-back on the regulation of thyrotropin [TSH] secretion. The Journal of Physiology. 1968;195(1):29–37.
    1. Dietrich JW, Fischer MR, Jauch J, Pantke E, Gärtner R, Pickardt CR. SPINA-THYR: a novel systems theoretic approach to determine the secretion capacity of the thyroid gland. European Journal of Internal Medicine. 1999;10(supplement 1):p. S34.
    1. Eisenberg M, DiStefano JJ. TSH-based protocol, tablet instability, and absorption effects on L-T 4 bioequivalence. Thyroid. 2009;19(2):103–110.
    1. Eisenberg M, Samuels M, DiStefano JJ., III L-T4 bioequivalence and hormone replacement studies via feedback control simulations. Thyroid. 2006;16(12):1279–1292.
    1. Ben-Shachar R, Eisenberg M, Huang SA, DiStefano JJ., III Simulation of post-thyroidectomy treatment alternatives for triiodothyronine or thyroxine replacement in pediatric cancer patients. Thyroid. 2012;22:595–603.
    1. Jostel A, Ryder WDJ, Shalet SM. The use of thyroid function tests in the diagnosis of hypopituitarism: definition and evaluation of the TSH Index. Clinical Endocrinology. 2009;71(4):529–534.
    1. Reichlin S, Utiger RD. Regulation of the pituitary-thyroid axis in man: relationship of TSH concentration to concentration of free and total thyroxine in plasma. The Journal of Clinical Endocrinology & Metabolism. 1967;27(2):251–255.
    1. Baloch Z, Carayon P, Conte-Devolx B, et al. Laboratory medicine practice guidelines. Laboratory support for the diagnosis and monitoring of thyroid disease. Thyroid. 2003;13(1):3–126.
    1. Jonklaas J, Soldin SJ. Tandem mass spectrometry as a novel tool for elucidating pituitary-thyroid relationships. Thyroid. 2008;18(12):1303–1311.
    1. Ito M, Miyauchi A, Morita S, et al. TSH-suppressive doses of levothyroxine are required to achieve preoperative native serum triiodothyronine levels in patients who have undergone total thyroidectomy. European Journal of Endocrinology. 2012;167:373–378.
    1. Benhadi N, Fliers E, Visser TJ, Reitsma JB, Wiersinga WM. Pilot study on the assessment of the setpoint of the hypothalamus- pituitary-thyroid axis in healthy volunteers. European Journal of Endocrinology. 2010;162(2):323–329.
    1. Ercan-Fang S, Schwartz HL, Mariash CN, Oppenheimer JH. Quantitative assessment of pituitary resistance to thyroid hormone from plots of the logarithm of thyrotropin versus serum free thyroxine index. The Journal of Clinical Endocrinology & Metabolism. 2000;85(6):2299–2303.
    1. Hoermann R, Eckl W, Hoermann C, Larisch R. Complex relationship between free thyroxine and TSH in the regulation of thyroid function. European Journal of Endocrinology. 2010;162(6):1123–1129.
    1. Clark PM, Holder RL, Haque SM, Hobbs FD, Roberts LM, Franklyn JA. The relationship between serum TSH and free T4 in older people. Journal of Clinical Pathology. 2012;65:463–465.
    1. Rothacker K, Hadlow NC, Wardrop R, Walsh JP. The relationship between TSH and free T4 is not log-linear and differs between genders and age groups. Proceedings of the ASN Events: The Annual Scientific Meeting of the Endocrine Society of Australia and the Society for Reproductive Biology; August 2012; Queensland, Australia. Gold Coast;
    1. Dietrich JW. SimThyr. 3.2 edition, Fairfax, VA: sourceforge. 2012,
    1. Degroot LJ, Decostre P, Phair R. A mathematical model of human iodine metabolism. The Journal of Clinical Endocrinology & Metabolism. 1971;32(6):757–765.
    1. Blincoe C. Computer simulation of iodine metabolism by mammals. Journal of Animal Science. 1975;40(2):342–350.
    1. Blincoe C. Thyroid homeostasis as studied by computer simulation. International Journal of Bio-Medical Computing. 1981;12(2):101–108.
    1. Berkovski V. New iodine models family for simulation of short-term biokinetics processes, pregnancy and lactation. Food and Nutrition Bulletin. 2002;23(3):87–94.
    1. Mak PH, DiStefano JJ. Optimal control policies for the prescription of thyroid hormones. Mathematical Biosciences. 1978;42(3-4):159–186.
    1. Brown-Grant K, Brennan RD, Yates FE. Simulation of the thyroid hormone-binding protein interactions in human plasma. The Journal of Clinical Endocrinology & Metabolism. 1970;30(6):733–751.
    1. Endo S, Nitta Y, Ohtaki M, et al. Estimation of dose absorbed fraction for 131I-beta rays in rat thyroid. Journal of Radiation Research. 1998;39:223–230.
    1. Hermanská J, Vosmiková K, Jirsa L, Kárný M, Sámal M. Biophysical inputs into the software ‘MIRDose’. Sbornik Lekarsky. 1998;99(4):521–527.
    1. Kolbert KS, Pentlow KS, Pearson JR, et al. Prediction of absorbed dose to normal organs in thyroid cancer patients treated with 131I by use of 124I PET and 3-dimensional internal dosimetry software. Journal of Nuclear Medicine. 2007;48(1):143–149.
    1. Kramer GH, Chamberlain MJ, Yiu S. A study of thyroid radioiodine monitoring by Monte Carlo simulations: implications for equipment design. Physics in Medicine and Biology. 1997;42(11):2175–2182.
    1. Kramer GH, Crowley P. The assessment of the effect of thyroid size and shape on the activity estimate using Monte Carlo simulation. Health Physics. 2000;78(6):727–738.
    1. Merle Y, Mentre F, Mallet A, Aurengo A. Computer-assisted individual estimation of radioiodine thyroid uptake in Grave’s disease. Computer Methods and Programs in Biomedicine. 1993;40(1):33–41.
    1. Noteboom JL, Hummel WA, Jansen JTM, van Bekkum DW, Broerse JJ. Simulation of measurements of uptake of 123I-iodide in the thyroid of fetal chimpanzees. Radiation Research. 1997;147(6):686–690.
    1. Traino AC, Di Martino F, Grosso M, et al. A predictive mathematical model for the calculation of the final mass of Graves' disease thyroids treated with 131I. Physics in Medicine and Biology. 2005;50:2181–2191.
    1. Bhati S, Patni HK. Monte-Carlo simulation of uncertainty in the estimation of 125 I in the thyroid. Radiation Protection Dosimetry. 2009;136(1):23–29.
    1. Diaz Londono G, Lallena AM. Specific absorbed fractions in thyroid diagnostics and treatment: Monte Carlo calculation with PENELOPE. Radiation Protection Dosimetry. 2012;150:41–49.
    1. Hammes J, Pietrzyk U, Schmidt M, Schicha H, Eschner W. GATE based Monte Carlo simulation of planar scintigraphy to estimate the nodular dose in radioiodine therapy for autonomous thyroid adenoma. Zeitschrift für Medizinische Physik. 2012;21:290–300.
    1. Khrutchinsky A, Drozdovitch V, Kutsen S, et al. Mathematical modeling of a survey-meter used to measure radioactivity in human thyroids: Monte Carlo calculations of the device response and uncertainties. Applied Radiation and Isotopes. 2012;70:743–751.
    1. Mowlavi AA, Fornasier MR, de Denaro M. Thyroid volume’s influence on energy deposition from 131I calculated by Monte Carlo (MC) simulation. Radiology and Oncology. 2011;45(2):143–146.
    1. Saeedzadeh E, Sarkar S, Abbaspour Tehrani-Fard A, Ay MR, Khosravi HR, Loudos G. 3D calculation of absorbed dose for 131I-targeted radiotherapy: a Monte Carlo study. Radiation Protection Dosimetry. 2012;150:298–305.
    1. Martínez L, Webb P, Polikarpov I, Skaf MS. Molecular dynamics simulations of ligand dissociation from thyroid hormone receptors: evidence of the likeliest escape pathway and its implications for the design of novel ligands. Journal of Medicinal Chemistry. 2006;49(1):23–26.
    1. Vedani A, Zumstein M, Lill MA, Ernst B. Simulating α/β selectivity at the human thyroid hormone receptor: consensus scoring using multidimensional QSAR. ChemMedChem. 2007;2(1):78–87.
    1. NOMOTHETICOS: Nonlinear Modelling of Thyroid Hormones' Effect on Thyrotropin Incretion in Confirmed Open-loop Situation. .
    1. Dayan CM, Panicker V. Novel insights into thyroid hormones from the study of common genetic variation. Nature Reviews Endocrinology. 2009;5(4):211–218.
    1. Boccone L, Mariotti S, Dessì V, Pruna D, Meloni A, Loudianos G. Allan-Herndon-Dudley syndrome (AHDS) caused by a novel SLC16A2 gene mutation showing severe neurologic features and unexpectedly low TRH-stimulated serum TSH. European Journal of Medical Genetics. 2010;53(6):392–395.
    1. Crushell E, Reardon W. Elevated TSH levels in a mentally retarded boy. European Journal of Pediatrics. 2010;169(5):573–575.
    1. Fuchs O, Pearr N, Pohlensz J, Schmidt H. Elevated serum triiodothyronine and intellectual and motor disability with paroxysmal dyskinesia caused by a monocarboxylate transporter 8 gene mutation. Developmental Medicine and Child Neurology. 2009;51(3):240–244.
    1. Schwartz CE, Stevenson RE. The MCT8 thyroid hormone transporter and Allan-Herndon-Dudley syndrome. Best Practice and Research. 2007;21(2):307–321.
    1. van der Deure WM, Peeters RP, Visser TJ. Molecular aspects of thyroid hormone transporters, including MCT8, MCT10, and OATPs, and the effects of genetic variation in these transporters. Journal of Molecular Endocrinology. 2010;44(1):1–11.
    1. Dumitrescu AM, Liao XH, Best TB, Brockmann K, Refetoff S. A novel syndrome combining thyroid and neurological abnormalities is associated with mutations in a monocarboxylate transporter gene. American Journal of Human Genetics. 2004;74(1):168–175.
    1. Maranduba CMC, Friesema ECH, Kok F, et al. Decreased cellular uptake and metabolism in Allan-Herndon-Dudley syndrome (AHDS) due to a novel mutation in the MCT8 thyroid hormone transporter. Journal of Medical Genetics. 2006;43(5):457–460.
    1. de Jong FJ, Peeters RP, den Heijer T, et al. The association of polymorphisms in the type 1 and 2 deiodinase genes with circulating thyroid hormone parameters and atrophy of the medial temporal lobe. The Journal of Clinical Endocrinology & Metabolism. 2007;92(2):636–640.
    1. Hoftijzer HC, Heemstra KA, Visser TJ, et al. The type 2 deiodinase ORFa-Gly3Asp polymorphism (rs12885300) influences the set point of the hypothalamus-pituitary-thyroid axis in patients treated for differentiated thyroid carcinoma. The Journal of Clinical Endocrinology & Metabolism. 2011;96:E1527–E1533.
    1. Panicker V, Cluett C, Shields B, et al. A common variation in deiodinase 1 gene DIO1 is associated with the relative levels of free thyroxine and triiodothyronine. The Journal of Clinical Endocrinology & Metabolism. 2008;93(8):3075–3081.
    1. Peeters RP, van den Beld AW, Attalki H, et al. A new polymorphism in the type II deiodinase gene is associated with circulating thyroid hormone parameters. American Journal of Physiology. 2005;289(1):E75–E81.
    1. Peeters RP, van Toor H, Klootwijk W, et al. Polymorphisms in thyroid hormone pathway genes are associated with plasma TSH and iodothyronine levels in healthy subjects. The Journal of Clinical Endocrinology & Metabolism. 2003;88(6):2880–2888.
    1. van der Deure WM, Hansen PS, Peeters RP, et al. The effect of genetic variation in the type 1 deiodinase gene on the interindividual variation in serum thyroid hormone levels: an investigation in healthy Danish twins. Clinical Endocrinology. 2009;70(6):954–960.
    1. Alberobello AT, Congedo V, Liu H, et al. An intronic SNP in the thyroid hormone receptor beta gene is associated with pituitary cell-specific over-expression of a mutant thyroid hormone receptor beta2 (R338W) in the index case of pituitary-selective resistance to thyroid hormone. Journal of Translational Medicine. 2011;9, article 144
    1. Sørensen HG, van der Deure WM, Hansen PS, et al. Identification and consequences of polymorphisms in the thyroid hormone receptor alpha and beta genes. Thyroid. 2008;18(10):1087–1094.
    1. Lee S, Young BM, Wan W, Chan IH, Privalsky ML. A mechanism for pituitary-resistance to thyroid hormone (PRTH) syndrome: a loss in cooperative coactivator contacts by thyroid hormone receptor (TR)β 2. Molecular Endocrinology. 2011;25(7):1111–1125.
    1. Tian H, Mahajan MA, Chun TW, Habeos I, Samuels HH. The N-terminal A/B domain of the thyroid hormone receptor-β2 isoform influences ligand-dependent recruitment of coactivators to the ligand-binding domain. Molecular Endocrinology. 2006;20(9):2036–2051.
    1. Malaguti A, Della Casa C, Castorina S, et al. Molecular mechanisms for pituitary thyrotroph cell growth. Journal of Endocrinological Investigation. 2004;27(6):151–167.
    1. Pals K, Vankelecom H, Denef C. Triiodothyronine expands the lactotroph and maintains the lactosomatotroph population, whereas thyrotrophin-releasing hormone augments thyrotroph abundance in aggregate cell cultures of postnatal rat pituitary gland. Journal of Neuroendocrinology. 2006;18(3):203–216.
    1. Szarek E, Farrand K, Mcmillen IC, Young IR, Houghton D, Schwartz J. Hypothalamic input is required for development of normal numbers of thyrotrophs and gonadotrophs, but not other anterior pituitary cells in late gestation sheep. The Journal of Physiology. 2008;586(4):1185–1194.
    1. Alberti L, Proverbio MC, Costagliola S, et al. Germline mutations of TSH receptor gene as cause of nonautoimmune subclinical hypothyroidism. The Journal of Clinical Endocrinology & Metabolism. 2002;87(6):2549–2555.
    1. Calebiro D, Gelmini G, Cordella D, et al. Frequent TSH receptor genetic alterations with variable signaling impairment in a large series of children with nonautoimmune isolated hyperthyrotropinemia. The Journal of Clinical Endocrinology & Metabolism. 2012;97:156–160.
    1. Tonacchera M, Perri A, De Marco G, et al. Low prevalence of thyrotropin receptor mutations in a large series of subjects with sporadic and familial nonautoimmune subclinical hypothyroidism. The Journal of Clinical Endocrinology & Metabolism. 2004;89(11):5787–5793.
    1. Persani L, Calebiro D, Cordella D, et al. Genetics and phenomics of hypothyroidism due to TSH resistance. Molecular and Cellular Endocrinology Journal. 2010;322:72–82.
    1. Clifton-Bligh RJ, Gregory JW, Ludgate M, et al. Two novel mutations in the thyrotropin (TSH) receptor gene in a child with resistance to TSH. The Journal of Clinical Endocrinology & Metabolism. 1997;82(4):1094–1100.
    1. de Roux N, Misrahi M, Brauner R, et al. Four families with loss of function mutations of the thyrotropin receptor. The Journal of Clinical Endocrinology & Metabolism. 1996;81(12):4229–4235.
    1. Duprez L, Parma J, Van Sande J, et al. TSH receptor mutations and thyroid disease. Trends in Endocrinology and Metabolism. 1998;9(4):133–140.
    1. Sunthornthepvarakul T, Gottschalk ME, Hayashi Y, Refetoff S. Brief report: resistance to thyrotropin caused by mutations in the thyrotropin-receptor gene. The New England Journal of Medicine. 1995;332(3):155–160.
    1. Vassart G, Costagliola S. G protein-coupled receptors: mutations and endocrine diseases. Nature Reviews Endocrinology. 2011;7(6):362–372.
    1. Radetti G, Maselli M, Buzi F, et al. The natural history of the normal/mild elevated TSH serum levels in children and adolescents with Hashimoto's thyroiditis and isolated hyperthyrotropinaemia: a 3-year follow-up. Clinical Endocrinology. 2012;76:394–398.
    1. Abramowicz MJ, Duprez L, Parma J, Vassart G, Heinrichs C. Familial congenital hypothyroidism due to inactivating mutation of the thyrotropin receptor causing profound hypoplasia of the thyroid gland. The Journal of Clinical Investigation. 1997;99(12):3018–3024.
    1. Biebermann H, Schöneberg T, Krude H, Schultz G, Gudermann T, Grüters A. Mutations of the human thyrotropin receptor gene causing thyroid hypoplasia and persistent congenital hypothyroidism. The Journal of Clinical Endocrinology & Metabolism. 1997;82(10):3471–3480.
    1. Gagné N, Parma J, Deal C, Vassart G, Van Vliet G. Apparent congenital athyreosis contrasting with normal plasma thyroglobulin levels and associated with inactivating mutations in the thyrotropin receptor gene: are athyreosis and ectopic thyroid distinct entities? The Journal of Clinical Endocrinology & Metabolism. 1998;83(5):1771–1775.
    1. Martino E, Pinchera A. Central hypothyroidism. In: Braverman LE, Utiger RD, editors. Werner and Ingbar's the Thyroid. 9th edition. Philadelphia, Pa, USA: Lippincott Williams & Wilkins; 2005. pp. 754–768.
    1. Beck-Peccoz P, Amr S, Menezes-Ferreira M, Faglia G, Weintraub BD. Decreased receptor binding of biologically inactive thyrotropin in central hypothyroidism. Effect of treatment with thyrotropin-releasing hormone. The New England Journal of Medicine. 1985;312(17):1085–1090.
    1. Collu R. Genetic aspects of central hypothyroidism. Journal of Endocrinological Investigation. 2000;23(2):125–134.
    1. Beck-Peccoz P, Brucker-Davis F, Persani L, Smallridge RC, Weintraub BD. Thyrotropin-secreting pituitary tumors. Endocrine Reviews. 1996;17(6):610–638.
    1. Refetoff S. Resistance to thyroid hormone. In: Braverman LE, Uller RP, editors. Werner and Ingbar's the Thyroid. Philadelphia, Pa, USA: Lippincott Williams & Wilkins; 2005. pp. 1109–1129.
    1. Li QL, Feng P, Satoh T, et al. Regulation of the human TRH (hTRH) gene by human thyroid hormone receptor β1 (hTRβ1) mutants. Endocrine Research. 1997;23(4):297–309.
    1. Rothenbuchner G, Loos U, Kiessling WR, Birk J, Pfeiffer EF. The influence of total starvation on the pituitary-thyroid-axis in obese individuals. Acta Endocrinologica, Supplement. 1973;173:p. 144.
    1. Portnay GI, O’Brien JT, Bush J, et al. The effect of starvation on the concentration and binding of thyroxine and triiodothyronine in serum and on the response to TRH. The Journal of Clinical Endocrinology & Metabolism. 1974;39(1):191–194.
    1. Chopra IJ. Nonthyroidal illness syndrome or euthyroid sick syndrome? Endocrine Practice. 1996;2:45–52.
    1. De Groot LJ. Dangerous dogmas in medicine: the nonthyroidal illness syndrome. The Journal of Clinical Endocrinology & Metabolism. 1999;84(1):151–164.
    1. Iervasi G, Pingitore A, Landi P, et al. Low-T3 syndrome: a strong prognostic predictor of death in patients with heart disease. Circulation. 2003;107(5):708–713.
    1. Scoscia E, Baglioni S, Eslami A, Iervasi G, Monti S, Todisco T. Low triiodothyronine (T3) state: a predictor of outcome in respiratory failure? Results of a clinical pilot study. European Journal of Endocrinology. 2004;151(5):557–560.
    1. Angelousi AG, Karageorgopoulos DE, Kapaskelis AM, Falagas ME. Association between thyroid function tests at baseline and the outcome of patients with sepsis or septic shock: a systematic review. European Journal of Endocrinology. 2011;164(2):147–155.
    1. Costa-e-Sousa RH, Hollenberg AN. Minireview: the neural regulation of the hypothalamic-pituitary-thyroid axis. Endocrinology. 2012;153:4128–4135.
    1. Schifferdecker E, Hering S, Bohm BO, et al. Thyroid hormone binding inhibition in critically ill patients—who is the inhibitor? Experimental and Clinical Endocrinology. 1990;95(2):267–270.
    1. Schifferdecker E, Hering S, Forster H, et al. Freie Fettsäuren im Serum bei krisenhaften Erkrankungen: Spielen sie eine Rolle bei der Proteinbindung von Schilddrüsenhormonen? [Free fatty acids in the serum in critical diseases: do they play a role in the protein binding of thyroid hormones?] Klin Wochenschr. 1988;66:308–313.
    1. Burger A, Nicod P, Suter P, Vallotton MB, Vagenakis P, Braverman L. Reduced active thyroid hormone levels in acute illness. The Lancet. 1976;1(7961):653–655.
    1. Carlin K, Carlin S. Possible etiology for euthyroid sick syndrome. Medical Hypotheses. 1993;40(1):38–43.
    1. Pittman CS, Shimizu T, Burger A, Chambers B., Jr. The nondeiodinative pathways of thyroxine metabolism: 3,5,3’,5’-tetraiodothyroacetic acid turnover in normal and fasting human subjects. The Journal of Clinical Endocrinology & Metabolism. 1980;50(4):712–716.
    1. Scanlan TS. Minireview: 3-iodothyronamine (T1AM): a new player on the thyroid endocrine team? Endocrinology. 2009;150(3):1108–1111.
    1. Doyle KP, Suchland KL, Ciesielski TMP, et al. Novel thyroxine derivatives, thyronamine and 3-iodothyronamine, induce transient hypothermia and marked neuroprotection against stroke injury. Stroke. 2007;38(9):2569–2576.
    1. Wood WJL, Geraci T, Nilsen A, DeBarber AE, Scanlan TS. Iodothyronamines are oxidatively deaminated to iodothyroacetic acids in vivo. ChemBioChem. 2009;10(2):361–365.
    1. Boelen A, Wiersinga WM, Fliers E. Fasting-induced changes in the hypothalamus-pituitary-thyroid axis. Thyroid. 2008;18(2):123–129.
    1. Kohel KA, MacKenzie DS, Rostal DC, Grumbles JS, Lance VA. Seasonality in plasma thyroxine in the desert tortoise, Gopherus agassizii. General and Comparative Endocrinology. 2001;121(2):214–222.
    1. Nicol SC, Andersen NA, Tomasi TE. Seasonal variations in thyroid hormone levels in free-living echidnas (Tachyglossus aculeatus) General and Comparative Endocrinology. 2000;117(1):1–7.
    1. Tomasi TE, Hellgren EC, Tucker TJ. Thyroid hormone concentrations in black bears (Ursus americanus): hibernation and pregnancy effects. General and Comparative Endocrinology. 1998;109(2):192–199.
    1. Mebis L, Eerdekens A, Guiza F, et al. Contribution of nutritional deficit to the pathogenesis of the nonthyroidal illness syndrome in critical illness: a rabbit model study. Endocrinology. 2012;153:973–984.
    1. Klemperer JD, Klein I, Gomez M, et al. Thyroid hormone treatment after coronary-artery bypass surgery. The New England Journal of Medicine. 1995;333(23):1522–1527.
    1. Klemperer JD, Klein IL, Ojamaa K, et al. Triiodothyronine therapy lowers the incidence of atrial fibrillation after cardiac operations. Annals of Thoracic Surgery. 1996;61(5):1323–1329.
    1. Ranasinghe AM, Quinn DW, Pagano D, et al. Glucose-insulin-potassium and tri-iodothyronine individually improve hemodynamic performance and are associated with reduced troponin I release after on-pump coronary artery bypass grafting. Circulation. 2006;114(1):I245–I250.
    1. Wyne KL. The role of thyroid hormone therapy in acutely ill cardiac patients. Critical Care. 2005;9(4):333–334.
    1. Güden M, Akpinar B, Sağbaş E, Sanisoğlu I, Cakali E, Bayindir O. Effects of intravenous triiodothyronine during coronary artery bypass surgery. Asian Cardiovascular and Thoracic Annals. 2002;10(3):219–222.
    1. Magalhäes APA, Gus M, Silva LB, Schaan BD. Oral triiodothyronine for the prevention of thyroid hormone reduction in adult valvular cardiac surgery. Brazilian Journal of Medical and Biological Research. 2006;39(7):969–978.
    1. Ronald A, Dunning J. Best evidence topic—cardiac general: does perioperative thyroxine have a role during adult cardiac surgery? Interactive Cardiovascular and Thoracic Surgery. 2006;5(2):166–178.
    1. Acker CG, Singh AR, Flick RP, Bernardini J, Greenberg A, Johnson JP. A trial of thyroxine in acute renal failure. Kidney International. 2000;57(1):293–298.
    1. Brent GA, Hershman JM. Thyroxine therapy in patients with severe nonthyroidal illnesses and low serum thyroxine concentration. The Journal of Clinical Endocrinology & Metabolism. 1986;63(1):1–8.
    1. Caplan RH. Comment on dangerous dogmas in medicine—the nonthyroidal illness syndrome. The Journal of Clinical Endocrinology & Metabolism. 1999;84(6):2262–2263.
    1. Utiger RD. Altered thyroid function in nonthyroidal illness and surgery—to treat or not to treat? The New England Journal of Medicine. 1995;333(23):1562–1563.
    1. Mebis L, van den Berghe G. The hypothalamus-pituitary-thyroid axis in critical illness. Netherlands Journal of Medicine. 2009;67(10):332–340.
    1. Larsen PR, Berry MJ. Nutritional and hormonal regulation of thyroid hormone deiodinases. Annual Review of Nutrition. 1995;15:323–352.
    1. Maia AL, Goemann IM, Meyer ELS, Wajner SM. Deiodinases: the balance of thyroid hormone: type 1 iodothyronine deiodinase in human physiology and disease. Journal of Endocrinology. 2011;209(3):283–297.
    1. Murakami M, Kamiya Y, Morimura T, et al. Thyrotropin receptors in brown adipose tissue: thyrotropin stimulates type II iodothyronine deiodinase and uncoupling protein-1 in brown adipocytes. Endocrinology. 2001;142(3):1195–1201.
    1. Nakao N, Ono H, Yamamura T, et al. Thyrotrophin in the pars tuberalis triggers photoperiodic response. Nature. 2008;452(7185):317–322.
    1. Boelen A, Kwakkel J, Wiersinga WM, Fliers E. Chronic local inflammation in mice results in decreased TRH and type 3 deiodinase mRNA expression in the hypothalamic paraventricular nucleus independently of diminished food intake. Journal of Endocrinology. 2006;191(3):707–714.
    1. Pantalone KM, Nasr C. Approach to a low tsh level: patience is a virtue. Cleveland Clinic Journal of Medicine. 2010;77(11):803–811.
    1. Figge J, Leinung M, Goodman AD, et al. The clinical evaluation of patients with subclinical hyperthyroidism and free triiodothyronine (free T3) toxicosis. American Journal of Medicine. 1994;96(3):229–234.
    1. Takamatsu J, Sugawara M, Kuma K, et al. Ratio of serum triiodothyronine to thyroxine and the prognosis of triiodothyronine-predominant Graves’ disease. Annals of Internal Medicine. 1984;100(3):372–375.
    1. Kim BW, Daniels GH, Harrison BJ, et al. Overexpression of type 2 iodothyronine deiodinase in follicular carcinoma as a cause of low circulating free thyroxine levels. The Journal of Clinical Endocrinology & Metabolism. 2003;88(2):594–598.
    1. Miyauchi A, Takamura Y, Ito Y, et al. 3,5,3′-Triiodothyronine thyrotoxicosis due to increased conversion of administered levothyroxine in patients with massive metastatic follicular thyroid carcinoma. The Journal of Clinical Endocrinology & Metabolism. 2008;93(6):2239–2242.
    1. Shahin AA, Abdoh S, Abdelrazik M. Prolactin and thyroid hormones in patients with systemic sclerosis: correlations with disease manifestations and activity. Zeitschrift fur Rheumatologie. 2002;61(6):703–709.
    1. Murakami M, Araki O, Hosoi Y, et al. Expression and regulation of type II iodothyronine deiodinase in human thyroid gland. Endocrinology. 2001;142(7):2961–2967.
    1. Suzuki S, Shigematsu S, Inaba H, Takei M, Takeda T, Komatsu M. Pituitary resistance to thyroid hormones: pathophysiology and therapeutic options. Endocrine. 2011;40:366–371.
    1. Bochukova E, Schoenmakers N, Agostini M, et al. A mutation in the thyroid hormone receptor alpha gene. The New England Journal of Medicine. 2012;366:243–249.
    1. Kaplan MM, Swartz SL, Larsen PR. Partial peripheral resistance to thyroid hormone. American Journal of Medicine. 1981;70(5):1115–1121.
    1. Tjørve E, Tjørve KMC, Olsen JO, Senum R, Oftebro H. On commonness and rarity of thyroid hormone resistance: a discussion based on mechanisms of reduced sensitivity in peripheral tissues. Medical Hypotheses. 2007;69(4):913–921.
    1. Movius EG, Phyillaier MM, Robbins J. Phloretin inhibits cellular uptake and nuclear receptor binding of triiodothyronine in human Hep G2 hepatocarcinoma cells. Endocrinology. 1989;124(4):1988–1997.
    1. Lisbôa PC, Curty FH, Moreira RM, Pazos-Moura CC. Effects of estradiol benzoate on 5′-iodothyronine deiodinase activities in female rat anterior pituitary gland, liver and thyroid gland. Brazilian Journal of Medical and Biological Research. 1997;30(12):1479–1484.
    1. Benvenga S, Ruggeri RM, Russo A, Lapa D, Campenni A, Trimarchi F. Usefulness of L-carnitine, a naturally occurring peripheral antagonist of thyroid hormone action, in iatrogenic hyperthyroidism: a randomized, double-blind, placebo-controlled clinical trial. The Journal of Clinical Endocrinology & Metabolism. 2001;86(8):3579–3594.
    1. Benvenga S, Lakshmanan M, Trimarchi F. Carnitine is a naturally occurring inhibitor of thyroid hormone nuclear uptake. Thyroid. 2000;10(12):1043–1050.
    1. Santos GM, Pantoja CJA, Costa ESA, et al. Thyroid hormone receptor binding to DNA and T3-dependent transcriptional activation are inhibited by uremic toxins. Nuclear Receptor. 2005;3, article 1
    1. DeGroot LJ, Rue PA. Roentgenographic contrast agents inhibit triiodothyronine binding to nuclear receptors in vitro. The Journal of Clinical Endocrinology & Metabolism. 1979;49(4):538–542.
    1. Vorperian VR, Havighurst TC, Miller S, January CT. Adverse effects of low dose amiodarone: a meta-analysis. Journal of the American College of Cardiology. 1997;30(3):791–798.
    1. Hahn CG, Pawlyk AC, Whybrow PC, Gyulai L, Tejani-Butt SM. Lithium administration affects gene expression of thyroid hormone receptors in rat brain. Life Sciences. 1999;64(20):1793–1802.
    1. Englebienne P, Verhas M, Herst CV, De Meirleir K. Type I interferons induce proteins susceptible to act as thyroid receptor (TR) corepressors and to signal the TR for destruction by the proteasome: possible etiology for unexplained chronic fatigue. Medical Hypotheses. 2003;60(2):175–180.
    1. DeGroot LJ, Coleoni AH, Rue PA, Seo H, Martino E, Refetoff S. Reduced nuclear triiodothyronine receptors in starvation induced hypothyroidism. Biochemical and Biophysical Research Communications. 1977;79(1):173–178.
    1. Tagami T, Nakamura H, Sasaki S, Miyoshi Y, Nakao K. Starvation-induced decrease in the maximal binding capacity for triiodothyronine of the thyroid hormone receptor is due to a decrease in the receptor protein. Metabolism. 1996;45(8):970–973.
    1. Sheng ZG, Tang Y, Liu YX, et al. Low concentrations of bisphenol a suppress thyroid hormone receptor transcription through a nongenomic mechanism. Toxicology and Applied Pharmacology Journal. 2012;259:133–142.
    1. Davis PJ, Lin HY, Mousa SA, et al. Overlapping nongenomic and genomic actions of thyroid hormone and steroids. Steroids. 2011;76(9):829–833.
    1. Davis PJ, Davis FB, Mousa SA, Luidens MK, Lin HY. Membrane receptor for thyroid hormone: physiologic and pharmacologic implications. Annual Review of Pharmacology and Toxicology. 2011;51:99–115.
    1. Cheng SY, Leonard JL, Davis PJ. Molecular aspects of thyroid hormone actions. Endocrine Reviews. 2010;31(2):139–170.
    1. Axelband F, Dias J, Ferrão FM, Einicker-Lamas M. Nongenomic signaling pathways triggered by thyroid hormones and their metabolite 3-iodothyronamine on the cardiovascular system. Journal of Cellular Physiology. 2011;226(1):21–28.
    1. Yehuda-Shnaidman E, Kalderon B, Azazmeh N, Bar-Tana J. Gating of the mitochondrial permeability transition pore by thyroid hormone. FASEB Journal. 2010;24(1):93–104.
    1. Cao HJ, Lin HY, Luidens MK, Davis FB, Davis PJ. Cytoplasm-to-nucleus shuttling of thyroid hormone receptor-β1 (TRβ1) is directed from a plasma membrane integrin receptor by thyroid hormone. Endocrine Research. 2009;34(1-2):31–42.
    1. Davis PJ, Zhou M, Davis FB, Lansing L, Mousa SA, Lin HY. Mini-review: cell surface receptor for thyroid hormone and nongenomic regulation of ion fluxes in excitable cells. Physiology and Behavior. 2010;99(2):237–239.
    1. Davis PJ, Leonard JL, Davis FB. Mechanisms of nongenomic actions of thyroid hormone. Frontiers in Neuroendocrinology. 2008;29(2):211–218.
    1. Bhargava M, Lei J, Mariash CN, Ingbar DH. Thyroid hormone rapidly stimulates alveolar Na,K-ATPase by activation of phosphatidylinositol 3-kinase. Current Opinion in Endocrinology, Diabetes and Obesity. 2007;14(5):416–420.
    1. Davis PJ, Davis FB. Nongenomic actions of thyroid hormone. Thyroid. 1996;6(5):497–504.
    1. Vandenberg LN, Colborn T, Hayes TB, et al. Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses. Endocrine Reviews. 2012;33:378–455.
    1. Andra SS, Makris KC. Thyroid disrupting chemicals in plastic additives and thyroid health. Journal of Environmental Science and Health C. 2012;30:107–151.
    1. Boas M, Feldt-Rasmussen U, Main KM. Thyroid effects of endocrine disrupting chemicals. Molecular and Cellular Endocrinology Journal. 2012;355:240–248.
    1. Meeker JD, Ferguson KK. Relationship between urinary phthalate and bisphenol A concentrations and serum thyroid measures in U.S. adults and adolescents from the National Health and Nutrition Examination Survey (NHANES) 2007-2008. Environmental Health Perspectives. 2011;119:1396–1402.
    1. Leijs MM, ten Tusscher GW, Olie K, et al. Thyroid hormone metabolism and environmental chemical exposure. Environmental Health. 2012;11(supplement 1):p. S10.
    1. Ji K, Kim S, Kho Y, et al. Serum concentrations of major perfluorinated compounds among the general population in Korea: dietary sources and potential impact on thyroid hormones. Environment International. 2012;45:78–85.
    1. Iwasaki T, Miyazaki W, Takeshita A, Kuroda Y, Koibuchi N. Polychlorinated biphenyls suppress thyroid hormone-induced transactivation. Biochemical and Biophysical Research Communications. 2002;299(3):384–388.
    1. Miyazaki W, Iwasaki T, Takeshita A, Kuroda Y, Koibuchi N. Polychlorinated biphenyls suppress thyroid hormone receptor-mediated transcription through a novel mechanism. The Journal of Biological Chemistry. 2004;279(18):18195–18202.
    1. Rickenbacher U, McKinney JD, Oatley SJ, Blake CCF. Structurally specific binding of halogenated biphenyls to thyroxine transport protein. Journal of Medicinal Chemistry. 1986;29(5):641–648.
    1. Zoeller RT. Environmental chemicals impacting the thyroid: targets and consequences. Thyroid. 2007;17(9):811–817.
    1. Liu C, Wang C, Yan M, Quan C, Zhou J, Yang K. PCB153 disrupts thyroid hormone homeostasis by affecting its biosynthesis, biotransformation, feedback regulation, and metabolism. Hormone and Metabolic Research. 2012;44:662–669.
    1. Buckman AH, Fisk AT, Parrott JL, Solomon KR, Brown SB. PCBs can diminish the influence of temperature on thyroid indices in rainbow trout (Oncorhynchus mykiss) Aquatic Toxicology. 2007;84(3):366–378.
    1. Higashihara N, Shiraishi K, Miyata K, Oshima Y, Minobe Y, Yamasaki K. Subacute oral toxicity study of bisphenol F based on the draft protocol for the ‘Enhanced OECD Test Guideline no. 407’. Archives of Toxicology. 2007;81(12):825–832.
    1. Gauger KJ, Giera S, Sharlin DS, Bansal R, Iannacone E, Zoeller RT. Polychlorinated biphenyls 105 and 118 form thyroid hormone receptor agonists after cytochrome P4501A1 activation in rat pituitary GH3 cells. Environmental Health Perspectives. 2007;115(11):1623–1630.
    1. Giera S, Bansal R, Ortiz-Toro TM, Taub DG, Zoeller RT. Individual polychlorinated biphenyl (PCB) congeners produce tissue- and gene-specific effects on thyroid hormone signaling during development. Endocrinology. 2011;152(7):2909–2919.
    1. Koulouri O, Auldin MA, Agarwal R, et al. Diagnosis and treatment of hypothyroidism in TSH deficiency compared to primary thyroid disease: pituitary patients are at risk of under-replacement with levothyroxine. Clinical Endocrinology. 2011;74(6):744–749.
    1. Takamatsu J, Kuma K, Mozai T. Serum triiodothyronine to thyroxine ratio: a newly recognized predictor of the outcome of hyperthyroidism due to Graves’ disease. The Journal of Clinical Endocrinology & Metabolism. 1986;62(5):980–983.
    1. Amino N, Yabu Y, Miki T, et al. Serum ratio of triiodothyronine to thyroxine, and thyroxine-binding globulin and calcitonin concentrations in Grave’s disease and destruction-induced thyrotoxicosis. The Journal of Clinical Endocrinology & Metabolism. 1981;53(1):113–116.
    1. Horn K, Koeppen D, Pickardt CR, Scriba PC. Thyroidal autoregulation in nontoxic goiter patients: normalization of the T3/T4 ratio in serum by treatment with potassium iodide. Klinische Wochenschrift. 1975;53(2):94–95.
    1. Laurberg P. Mechanisms governing the relative proportions of thyroxine and 3,5,3’-triiodothyronine in thyroid secretion. Metabolism. 1984;33(4):379–392.
    1. Berger MM, Reymond MJ, Shenkin A, et al. Influence of selenium supplements on the post-traumatic alterations of the thyroid axis: a placebo-controlled trial. Intensive Care Medicine. 2001;27(1):91–100.
    1. Fisher DA. Physiological variations in thyroid hormones: physiological and pathophysiological considerations. Clinical Chemistry. 1996;42(1):135–139.
    1. Dong BJ. How medications affect thyroid function. Western Journal of Medicine. 2000;172(2):102–106.
    1. Mortoglou A, Candiloros H. The serum triiodothyronine to thyroxine (T3/T4) ratio in various thyroid disorders and after Levothyroxine replacement therapy. Hormones. 2004;3:120–126.
    1. Sesmilo G, Simó O, Choque L, Casamitjana R, Puig-Domingo M, Halperin I. Serum free triiodothyronine (T3) to free thyroxine (T4) ratio in treated central hypothyroidism compared with primary hypothyroidism and euthyroidism. Endocrinologia y Nutricion. 2011;58(1):9–15.
    1. Rosołowska-Huszcz D, Kozłowska L, Rydzewski A. Influence of low protein diet on nonthyroidal illness syndrome in chronic renal failure. Endocrine. 2005;27(3):283–288.
    1. Liu S, Ren J, Zhao Y, et al. Nonthyroidal illness syndrome: is it far away from Crohn's disease? Journal of Clinical Gastroenterology. In press.
    1. Grußendorf M. Metabolismus der Schilddrüsenhormone. 1st edition. Stuttgart, Germany: Georg Thieme; 1988.
    1. Dumont JE, Vassart G DeGroot. Endocrinology. WB Saunders; 1995. Thyroid regulation.
    1. Greenspan FS. The thyroid gland. In: Greenspan FS, Strewler GJ, Stamford CT, editors. Basic & Clinical Endocrinology. 5th edition. Appleton & Lange; 1997. pp. 192–262.
    1. Visser TJ, Kaplan MM, Leonard JL, Larsen PR. Evidence for two pathways of iodothyronine 5’-deiodination in rat pituitary that differ in kinetics, propylthiouracil sensitivity, and response to hypothyroidism. The Journal of Clinical Investigation. 1983;71(4):992–1002.
    1. den Brinker M, Joosten KFM, Visser TJ, et al. Euthyroid sick syndrome in meningococcal sepsis: the impact of peripheral thyroid hormone metabolism and binding proteins. The Journal of Clinical Endocrinology & Metabolism. 2005;90(10):5613–5620.
    1. Kano T, Kojima T, Takahashi T, Muto Y. Serum thyroid hormone levels in patients with fulminant hepatitis: usefulness of rT3 and the rT3/T3 ratio as prognostic indices. Gastroenterologia Japonica. 1987;22(3):344–353.
    1. Banovac K, Bzik L, Sekso M, Petek M. Decreased ratio of serum T3:rT3 in patients with hyperthyroidism. Endokrinologie. 1978;71(2):159–163.
    1. Ruhla S, Arafat AM, Weickert MO, et al. T3/rT3-ratio is associated with insulin resistance independent of TSH. Hormone and Metabolic Research. 2011;43(2):130–134.
    1. Lessels CM, Boag TP. Unrepeatable repeatabilities—a common mistake. The Auk. 1987;104:116–121.
    1. Jeyabalan A, Urban A, Ackermann A, et al. Kongress Der Deutschen Gesellschaft Für Innere Medizin. Vol. 117. Wiesbaden, Germany: 2011. Die berechnete Sekretionsleistung der Schilddrüse unterstützt die Differentialdiagnose zwischen latenter Hyperthyreose und Non-Thyroidal-Illness-Syndrom mit thyreotroper Adaptation; p. p. PS97.
    1. Pohlenz J, Weiss RE, Macchia PE, et al. Five new families with resistance to thyroid hormone not caused by mutations in the thyroid hormone receptor beta gene Resistance to thyroid hormone caused by two mutant thyroid hormone receptors beta, R243Q and R243W, with marked impairment of function that cannot be explained by altered in vitro 3, 5, 3′-triiodothyroinine binding affinity. The Journal of Clinical Endocrinology & Metabolism. 1999;84:3919–3928.
    1. Panicker V, Wilson SG, Spector TD, et al. Heritability of serum TSH, free T4 and free T3 concentrations: a study of a large UK twin cohort. Clinical Endocrinology. 2008;68(4):652–659.
    1. Christofides ND, Midgley JEM. Inaccuracies in free thyroid hormone measurement by ultrafiltration and tandem mass spectrometry. Clinical Chemistry. 2009;55(12):2228–2230.
    1. Jonklaas J, Kahric-Janicic N, Soldin OP, Soldin SJ. Correlations of free thyroid hormones measured by tandem mass spectrometry and immunoassay with thyroid-stimulating hormone across 4 patient populations. Clinical Chemistry. 2009;55(7):1380–1388.
    1. Lee RH, Spencer CA, Mestman JH, et al. Free T4 immunoassays are flawed during pregnancy. American Journal of Obstetrics and Gynecology. 2009;200(3):260.e1–260.e6.
    1. Midgley JEM, Christofides ND. Point—legitimate and illegitimate tests of free-analyte assay function. Clinical Chemistry. 2009;55(3):439–441.
    1. Midgley JE, Christofides ND. Free thyroxine assays: no going back! American Journal of Obstetrics and Gynecology. 2010;202(2):p. e6.
    1. Shivaraj G, Prakash BD, Sonal V, Shruthi K, Vinayak H, Avinash M. Thyroid function tests: a review. European Review for Medical and Pharmacological Sciences. 2009;13:341–349.
    1. Spencer CA. Clinical utility and cost-effectiveness of sensitive thyrotropin assays in ambulatory and hospitalized patients. Mayo Clinic Proceedings. 1988;63(12):1214–1222.
    1. Spencer CA, LoPresti JS, Patel A, et al. Applications of a new chemiluminometric thyrotropin assay to subnormal measurement. The Journal of Clinical Endocrinology & Metabolism. 1990;70(2):453–460.
    1. Wilcox RB, Nelson JC. Counterpoint: legitimate and illegitimate tests of free-analyte assay function: we need to identify the factors that influence free-analyte assay results. Clinical Chemistry. 2009;55(3):442–444.
    1. Loh TP, Kao SL, Halsall DJ, et al. Macro-thyrotropin: a case report and review of literature. The Journal of Clinical Endocrinology & Metabolism. 2012;97:1823–1828.
    1. Rix M, Laurberg P, Porzig C, Kristensen SR. Elevated thyroid-stimulating hormone level in a euthyroid neonate caused by macro thyrotropin-IgG complex. Acta Paediatrica. 2011;100:e135–e137.
    1. Sakai H, Fukuda G, Suzuki N, Watanabe C, Odawara M. Falsely elevated thyroid-stimulating hormone (TSH) level due to macro-TSH. Endocrine Journal. 2009;56(3):435–440.
    1. Cohen RN, Wondisford FE. Chemistry and biosynthesis of thyrotropin. In: Braverman LE, Uller RP, editors. Werner and Ingbar's the Thyroid. Philadelphia, Pa, USA: Lippincott Williams & Wilkins; 2005. pp. 159–175.
    1. Grossmann M, Weintraub BD, Szkudlinski MW. Novel insights into the molecular mechanisms of human thyrotropin action: structural, physiological, and therapeutic implications for the glycoprotein hormone family. Endocrine Reviews. 1997;18(4):476–501.
    1. Grossmann M, Szkudlinski MW, Wong R, Dias JA, Ji TH, Weintraub BD. Substitution of the seat-belt region of the thyroid-stimulating hormone (TSH) β-subunit with the corresponding regions of choriogonadotropin or follitropin confers luteotropic but not follitropic activity to chimeric TSH. The Journal of Biological Chemistry. 1997;272(24):15532–15540.
    1. Szkudlinski MW, Grossmann M, Leitolf H, Weintraub BD. Human thyroid-stimulating hormone: structure-function analysis. Methods. 2000;21(1):67–81.
    1. Nakabayashi K, Matsumi H, Bhalla A, et al. Thyrostimulin, a heterodimer of two new human glycoprotein hormone subunits, activates the thyroid-stimulating hormone receptor. The Journal of Clinical Investigation. 2002;109(11):1445–1452.
    1. Vassart G, Costagliola S. The thyrotropin receptor. In: Braverman LE, Uller RP, editors. Werner and Ingbar's the Thyroid. Philadelphia, Pa, USA: Lippincott Williams & Wilkins; 2005. pp. 176–183.
    1. Joshi LR, Weintraub BD. Naturally occurring forms of thyrotropin with low bioactivity and altered carbohydrate content act as competitive antagonists to more bioactive forms. Endocrinology. 1983;113(6):2145–2154.
    1. Sanders J, Miguel RN, Furmaniak J, Smith BR. TSH receptor monoclonal antibodies with agonist, antagonist, and inverse agonist activities. Methods in Enzymology. 2010;485:393–420.
    1. Gershengorn MC, Neumann S. Update in TSH receptor agonists and antagonists. The Journal of Clinical Endocrinology & Metabolism. 2012;97(12):4287–4292.
    1. Neumann S, Huang W, Titus S, et al. Small-molecule agonists for the thyrotropin receptor stimulate thyroid function in human thyrocytes and mice. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(30):12471–12476.
    1. Heinzeller T. Entwicklungsgeschichte, Ontogenese und Histologie. In: Stuttgart GR, editor. Schilddrüsenerkrankungen. Wissenschaftliche Verlagsgesellschaft; 2004. pp. 3–15.
    1. O’Rahilly R. The timing and sequence of events in the development of the human endocrine system during the embryonic period proper. Anatomy and Embryology. 1983;166(3):439–451.
    1. Glinoer D. The regulation of thyroid function in pregnancy: pathways of endocrine adaptation from physiology to pathology. Endocrine Reviews. 1997;18(3):404–433.
    1. Dietrich JW. Thyreotoxische Krise [thyroid storm] Med Klin Intensivmed Notfmed. 2012;107:448–453.
    1. Chiniwala NU, Woolf PD, Bruno CP, Kaur S, Spector H, Yacono K. Thyroid storm caused by a partial hydatidiform mole. Thyroid. 2008;18(4):479–481.
    1. Walkington L, Webster J, Hancock BW, Everard J, Coleman RE. Hyperthyroidism and human chorionic gonadotrophin production in gestational trophoblastic disease. British Journal of Cancer. 2011;104(11):1665–1669.
    1. Arrabal-Polo MA, Jimenez-Pacheco A, Arrabal-Martin M, et al. Hyperthyroidism as a clinical manifestation of a embryonal carcinoma of the testis. Acta Clinica Belgica. 2012;67:214–216.
    1. Giralt SA, Dexeus F, Amato R, Sella A, Logothetis C. Hyperthyroidism in men with germ cell tumors and high levels of beta-human chorionic gonadotropin. Cancer. 1992;69(5):1286–1290.
    1. Kato K, Mostafa MH, Mann K, Schindler AE, Hoermann R. The human chorionic gonadotropin molecule from patients with trophoblastic diseases has a high thyrotropic activity but is less active in the ovary. Gynecological Endocrinology. 2004;18(5):269–277.
    1. Hoermann R, Poertl S, Liss I, Amir SM, Mann K. Variation in the thyrotropic activity of human chorionic gonadotropin in Chinese hamster ovary cells arises from differential expression of the human thyrotropin receptor and microheterogeneity of the hormone. The Journal of Clinical Endocrinology & Metabolism. 1995;80(5):1605–1610.
    1. Rodien P, Brémont C, Sanson MLR, et al. Familial gestational hyperthyroidism caused by a mutant thyrotropin receptor hypersensitive to human chorionic gonadotropin. The New England Journal of Medicine. 1998;339(25):1823–1826.
    1. Okada SL, Ellsworth JL, Durnam DM, et al. A glycoprotein hormone expressed in corticotrophs exhibits unique binding properties on thyroid-stimulating hormone receptor. Molecular Endocrinology. 2006;20(2):414–425.
    1. van Zeijl CJJ, Surovtseva OV, Wiersinga WM, Fliers E, Boelen A. Acute inflammation increases pituitary and hypothalamic glycoprotein hormone subunit B5 mRNA expression in association with decreased thyrotrophin receptor mRNA expression in mice. Journal of Neuroendocrinology. 2011;23(4):310–319.
    1. Ikekubo K, Hino M, Saiki Y, et al. Immeasurably low and non-TRH-stimulatable TSH associated with normal I-123 uptake in two goitrous euthyroid patients: possible existence of other thyroid-hormone regulated thyroid stimulators other than TSH. Endocrine Journal. 2005;52(1):61–68.
    1. Li C, Hirooka Y, Habu S, Takagi J, Gotoh M, Nogimori T. Distribution of thyrostimulin in the rat: an immunohistochemical study. Endocrine Regulations. 2004;38(4):131–142.
    1. Suzuki C, Nagasaki H, Okajima Y, et al. Inflammatory cytokines regulate glycoprotein subunit β5 of thyrostimulin through nuclear factor-κB. Endocrinology. 2009;150(5):2237–2243.
    1. Takasu N, Matsushita M. Changes of TSH-stimulation blocking antibody (TSBAb) and thyroid stimulating antibody (TSAb) over 10 years in 34 TSBAb-positive patients with hypothyroidism and in 98 TSAb-positive Graves' patients with hyperthyroidism: reevaluation of TSBAb and TSAb in TSH-receptor-antibody (TRAb)-positive patients. Journal of Thyroid Research. 2012;2012:11 pages.182176
    1. Kremer JMH, Wilting J, Janssen LHM. Drug binding to human alpha-1-acid glycoprotein in health and disease. Pharmacological Reviews. 1988;40(1):1–47.
    1. Urien S, Bree F, Testa B, Tillement JP. pH-dependency of basic ligand binding to α1-acid glycoprotein (orosomucoid) Biochemical Journal. 1991;280, part 1:277–280.
    1. Zimmermann-Belsing T, Rasmussen AK, Feldt-Rasmussen U, Bog-Hansen TC. The influence of alpha1-acid glycoprotein (orosomucoid) and its glycoforms on the function of human thyrocytes and CHO cells transfected with the human TSH receptor. Molecular and Cellular Endocrinology. 2002;188(1-2):241–251.
    1. Akamizu T, Matsuda F, Okuda J, et al. Molecular analysis of stimulatory anti-thyrotropin receptor antibodies (TSAbs) involved in Graves' disease. Isolation and reconstruction of antibody genes, and production of monoclonal TSAbs. Journal of Immunology. 1996;157(7):3148–3152.
    1. Akamizu T, Moriyama K, Miura M, Saijo M, Matsuda F, Nakao K. Characterization of recombinant monoclonal antithyrotropin receptor antibodies (TSHRAbs) derived from lymphocytes of patients with Graves’ disease: epitope and binding study of two stimulatory TSHRAbs. Endocrinology. 1999;140(4):1594–1601.
    1. Morgenthaler NG, Kim MR, Tremble J, et al. Human immunoglobulin G autoantibodies to the thyrotropin receptor from Epstein-Barr virus-transformed B lymphocytes: characterization by immunoprecipitation with recombinant antigen and biological activity. The Journal of Clinical Endocrinology & Metabolism. 1996;81(9):3155–3161.
    1. Moriyama K, Okuda J, Saijo M, et al. Recombinant monoclonal thyrotropin-stimulation blocking antibody (TSBAb) established from peripheral lymphocytes of a hypothyroid patient with primary myxedema. Journal of Endocrinological Investigation. 2003;26(11):1076–1080.
    1. Okuda J, Akamizu T, Sugawa H, Matsuda F, Hua L, Mori T. Preparation and characterization of monoclonal antithyrotropin receptor antibodies obtained from peripheral lymphocytes of hypothyroid patients with primary myxedema. The Journal of Clinical Endocrinology & Metabolism. 1994;79(6):1600–1604.
    1. Yoshida T, Ichikawa Y, Ito K, Homma M. Monoclonal antibodies to the thyrotropin receptor bind to a 56-kDa subunit of the thyrotropin receptor and show heterogeneous bioactivities. The Journal of Biological Chemistry. 1988;263(31):16341–16347.
    1. Kohn LD, Suzuki K, Hoffman WH, et al. Characterization of monoclonal thyroid-stimulating and thyrotropin binding-inhibiting autoantibodies from a Hashimoto’s patient whose children had intrauterine and neonatal thyroid disease. The Journal of Clinical Endocrinology & Metabolism. 1997;82(12):3998–4009.
    1. Valente WA, Vitti P, Yavin Z, et al. Monoclonal antibodies to the thyrotropin receptor: stimulating and blocking antibodies derived from the lymphocytes of patients with Graves disease. Proceedings of the National Academy of Sciences of the United States of America. 1982;79(21 I):6680–6684.
    1. Valente WA, Yavin Z, Yavin E, et al. Monoclonal antibodies to the thyrotropin receptor. The identification of blocking and stimulating antibodies. Journal of Endocrinological Investigation. 1982;5(5):293–301.
    1. Sanders J, Jeffreys J, Depraetere H, et al. Thyroid-stimulating monoclonal antibodies. Thyroid. 2002;12(12):1043–1050.
    1. Ando T, Latif R, Pritsker A, Moran T, Nagayama Y, Davies TF. A monoclonal thyroid-stimulating antibody. The Journal of Clinical Investigation. 2002;110(11):1667–1674.
    1. Sanders J, Evans M, Premawardhana LDKE, et al. Human monoclonal thyroid stimulating autoantibody. The Lancet. 2003;362(9378):126–128.
    1. Evans M, Sanders J, Tagami T, et al. Monoclonal autoantibodies to the TSH receptor, one with stimulating activity and one with blocking activity, obtained from the same blood sample. Clinical Endocrinology. 2010;73(3):404–412.
    1. Sanders J, Allen F, Jeffreys J, et al. Characteristics of a monoclonal antibody to the thyrotropin receptor that acts as a powerful thyroid-stimulating autoantibody antagonist. Thyroid. 2005;15(7):672–682.
    1. Smith BR, Sanders J, Furmaniak J. TSH receptor antibodies. Thyroid. 2007;17(10):923–938.
    1. Sanders J, Evans M, Betterle C, et al. A human monoclonal autoantibody to the thyrotropin receptor with thyroid-stimulating blocking activity. Thyroid. 2008;18(7):735–746.
    1. Costagliola S, Bonomi M, Morgenthaler NG, et al. Delineation of the discontinuous-conformational epitope of a monoclonal antibody displaying full in vitro and in vivo thyrotropin activity. Molecular Endocrinology. 2004;18(12):3020–3034.
    1. Gilbert JA, Gianoukakis AG, Salehi S, et al. Monoclonal pathogenic antibodies to the thyroid-stimulating hormone receptor in Graves’ disease with potent thyroid-stimulating activity but differential blocking activity activate multiple signaling pathways. Journal of Immunology. 2006;176(8):5084–5092.
    1. Liu C, Hermsen D, Domberg J, et al. Comparison of M22-based ELISA and human-TSH-receptor-based luminescence assay for the measurement of thyrotropin receptor antibodies in patients with thyroid diseases. Hormone and Metabolic Research. 2008;40(7):479–483.
    1. Grossmann M, Leitolf H, Weintraub BD, Szkudlinski MW. A rational design strategy for protein hormone superagonists. Nature Biotechnology. 1998;16(9):871–875.
    1. Szkudlinski MW, Teh NG, Grossmann M, Tropea JE, Weintraub BD. Engineering human glycoprotein hormone superactive analogues. Nature Biotechnology. 1996;14(10):1257–1263.
    1. Azzam N, Bar-Shalom R, Fares F. Conversion of TSH heterodimer to a single polypeptide chain increases bioactivity and longevity. Endocrinology. 2012;153:954–960.
    1. Dayan CM, Saravanan P, Bayly G. Whose normal thyroid function is better—yours or mine? The Lancet. 2002;360(9330):353–354.
    1. Laurberg P, Andersen S, Carlé A, Karmisholt J, Knudsen N, Pedersen IB. The TSH upper reference limit: where are we at? Nature Reviews Endocrinology. 2011;7(4):232–239.
    1. Effraimidis G, Strieder TGA, Tijssen JGP, Wiersinga WM. Natural history of the transition from euthyroidism to overt autoimmune hypo- or hyperthyroidism: a prospective study. European Journal of Endocrinology. 2011;164(1):107–113.
    1. Gartner W, Weissel M. Do iodine-containing contrast media induce clinically relevant changes in thyroid function parameters of euthyroid patients within the first week? Thyroid. 2004;14(7):521–524.
    1. Stuckey BGA, Kent GN, Ward LC, Brown SJ, Walsh JP. Postpartum thyroid dysfunction and the long-term risk of hypothyroidism: results from a 12-year follow-up study of women with and without postpartum thyroid dysfunction. Clinical Endocrinology. 2010;73(3):389–395.
    1. Vanderpump MPJ, Tunbridge WMG, French JM, et al. The incidence of thyroid disorders in the community: a twenty-year follow-up of the Whickham Survey. Clinical Endocrinology. 1995;43(1):55–68.
    1. Walsh JP, Bremner AP, Feddema P, Leedman PJ, Brown SJ, O’Leary P. Thyrotropin and thyroid antibodies as predictors of hypothyroidism: a 13-year, longitudinal study of a community-based cohort using current immunoassay techniques. The Journal of Clinical Endocrinology & Metabolism. 2010;95(3):1095–1104.
    1. Hamilton TE, Davis S, Onstad L, Kopecky KJ. Thyrotropin levels in a population with no clinical, autoantibody, or ultrasonographic evidence of thyroid disease: implications for the diagnosis of subclinical hypothyroidism. The Journal of Clinical Endocrinology & Metabolism. 2008;93(4):1224–1230.
    1. Spencer CA, Hollowell JG, Kazarosyan M, Braverman LE. National Health and Nutrition Examination Survey III Thyroid-Stimulating Hormone (TSH)-thyroperoxidase antibody relationships demonstrate that TSH upper reference limits may be skewed by occult thyroid dysfunction. The Journal of Clinical Endocrinology & Metabolism. 2007;92(11):4236–4240.
    1. Vejbjerg P, Knudsen N, Perrild H, et al. The association between hypoechogenicity or irregular echo pattern at thyroid ultrasonography and thyroid function in the general population. European Journal of Endocrinology. 2006;155(4):547–552.
    1. Andersen S, Pedersen KM, Bruun NH, Laurberg P. Narrow individual variations in serum T4 and T3 in normal subjects: a clue to the understanding of subclinical thyroid disease. The Journal of Clinical Endocrinology & Metabolism. 2002;87(3):1068–1072.
    1. Hoermann R, Midgley JE. TSH measurement and its implications on personalised clinical decision making. Journal of Thyroid Research. 2012;2012:9 pages.438037
    1. Dickey RA, Wartofsky L, Feld S. Optimal thyrotropin level: Normal ranges and reference intervals are not equivalent. Thyroid. 2005;15(9):1035–1039.
    1. Wartofsky L, Dickey RA. The evidence for a narrower thyrotropin reference range is compelling. The Journal of Clinical Endocrinology & Metabolism. 2005;90(9):5483–5488.
    1. Wartofsky L, Van Nostrand D, Burman KD. Overt and ’subclinical’ hypothyroidism in women. Obstetrical and Gynecological Survey. 2006;61(8):535–542.
    1. Surks MI, Goswami G, Daniels GH. The thyrotropin reference range should remain unchanged. The Journal of Clinical Endocrinology & Metabolism. 2005;90(9):5489–5496.
    1. Amidi M, Leon DF, DeGroot WJ, Kroetz FW, Leonard JJ. Effect of the thyroid state on myocardial contractility and ventricular ejection rate in man. Circulation. 1968;38(2):229–239.
    1. Selvaraj S, Klein I, Danzi S, Akhter N, Bonow RO, Shah SJ. Association of serum triiodothyronine with B-type natriuretic peptide and severe left ventricular diastolic dysfunction in heart failure with preserved ejection fraction. American Journal of Cardiology. 2012;110:234–239.
    1. Belehradek J, Huxley JS. Changes in oxygen consumption during metamorphosis induced by thyroid administration in the axolotl. The Journal of Physiology. 1927;64:267–278.
    1. Dock W, Lewis JK. The effect of thyroid feeding on the oxygen consumption of the heart and of other tissues. The Journal of Physiology. 1932;74:401–406.
    1. Ortega Martinez De Victoria E, Pannacciulli N, Bogardus C, Krakoff J. Plasma concentrations of free triiodothyronine predict weight change in euthyroid persons. American Journal of Clinical Nutrition. 2007;85(2):440–445.
    1. Rawson RE, Concannon PW, Roberts PJ, Tennant BC. Seasonal differences in resting oxygen consumption, respiratory quotient, and free thyroxine in woodchucks. American Journal of Physiology. 1998;274(4):R963–R969.
    1. Carlson LD. Nonshivering thermogenesis and its endocrine control. Federation Proceedings. 1960;19(supplement 5):25–30.
    1. Edelman IS. Thyroid thermogenesis. The New England Journal of Medicine. 1974;290(23):1303–1308.
    1. Smith RE. Thermogenesis and thyroid action. Nature. 1964;204(4965):1311–1312.
    1. Burini R, Santidrian S, Moreyra M, Brown P, Munro HN, Young VR. Interaction of thyroid status and diet on muscle protein breakdown in the rat, as measured by N(τ)-methylhistidine excretion. Metabolism. 1981;30(7):679–687.
    1. Burman KD, Wartofsky L, Dinterman RE, Kesler P, Wannemacher RW., Jr. The effect of T3 and reverse T3 administration on muscle protein catabolism during fasting as measured by 3-methylhistidine excretion. Metabolism. 1979;28(8):805–813.
    1. Von Kazuyoshi H. Über das aus hühnerharn isolierte methylhistidin. Journal of Biochemistry. 1963;54(3):253–258.
    1. Munro HN, Young VR. Urinary excretion of N(tau) methylhistidine (3 methylhistidine): a tool to study metabolic responses in relation to nutrient and hormonal status in health and disease of man. American Journal of Clinical Nutrition. 1978;31(9):1608–1614.
    1. Rodier M, Richard JL, Bringer J, Cavalie G, Bellet H, Mirouze J. Thyroid status and muscle protein breakdown as assessed by urinary 3-methylhistidine excretion: study in thyrotoxic patients before and after treatment. Metabolism. 1984;33(1):97–100.
    1. Wang F, Tan Y, Wang C, et al. Thyroid-stimulating hormone levels within the reference range are associated with serum lipid profiles independent of thyroid hormones. The Journal of Clinical Endocrinology & Metabolism. 2012;97:2724–2731.
    1. Peters JP, Man EB. The interrelations of serum lipids in patients with thyroid disease. The Journal of Clinical Investigation. 1943;22:715–720.
    1. Atis G, Dalkilinc A, Altuntas Y, et al. Hyperthyroidism: a risk factor for female sexual dysfunction. Journal of Sexual Medicine. 2011;8(8):2327–2333.
    1. Pugeat M, Nader N, Hogeveen K, Raverot G, Déchaud H, Grenot C. Sex hormone-binding globulin gene expression in the liver: drugs and the metabolic syndrome. Molecular and Cellular Endocrinology. 2010;316(1):53–59.
    1. Selva DM, Hammond GL. Thyroid hormones act indirectly to increase sex hormone-binding globulin production by liver via hepatocyte nuclear factor-4α . Journal of Molecular Endocrinology. 2009;43(1):19–27.
    1. de Nayer P, Lambot MP, Desmons MC, Rennotte B, Malvaux P, Beckers C. Sex hormone-binding protein in hyperthyroxinemic patients: a discriminator for thyroid status in thyroid hormone resistance and familial dysalbuminemic hyperthyroxinemia. The Journal of Clinical Endocrinology & Metabolism. 1986;62(6):1309–1312.
    1. Sarne DH, Refetoff S, Rosenfield RL, Farriaux JP. Sex hormone-binding globulin in the diagnosis of peripheral tissue resistance to thyroid hormone: the value of changes after short term triiodothyronine administration. The Journal of Clinical Endocrinology & Metabolism. 1988;66(4):740–746.
    1. Meikle AW. The Interrelationships between thyroid dysfunction and hypogonadism in men and boys. Thyroid. 2004;14(supplement 1):S17–S25.
    1. Skowronska-Jozwiak EB, Krawczyk-Rusiecka K, Lewandowski KC, Adamczewski Z, Lewi Ski A. Successful treatment of thyrotoxicosis is accompanied by a decrease in serum sclerostin levels. Thyroid Research. 2012;5, article 14
    1. Mittag J, Behrends T, Nordstrom K, Anselmo J, Vennstrom B, Schomburg L. Serum copper as a novel biomarker for resistance to thyroid hormone. Biochemical Journal. 2012;443:103–109.
    1. Meeker JD, Rossano MG, Protas B, et al. Multiple metals predict prolactin and thyrotropin (TSH) levels in men. Environmental Research. 2009;109(7):869–873.
    1. Feher L, Hevizy HF. On the correlation of ceruloplasmin and the neuro-endocrine system. I. Correlation between the ceruloplasmin level and thyroid function. Magyar Belorvosi Archivum. 1962;15:210–213.
    1. Rechenberger J. Serumkupfer und Schilddrüsentatigkeit [serum copper and thyroid activity] Dtsch Z Verdau Stoffwechselkr. 1957;17:139–145.
    1. Koenig KF, Potlukova E, Mueller B, Christ-Crain M, Trendelenburg M. MBL serum levels in patients with sepsis correlate with thyroid function but not with outcome. Clinical Immunology. 2012;144:80–82.
    1. Potlukova E, Jiskra J, Freiberger T, et al. The production of mannan-binding lectin is dependent upon thyroid hormones regardless of the genotype: a cohort study of 95 patients with autoimmune thyroid disorders. Clinical Immunology. 2010;136(1):123–129.
    1. Biondi B, Wartofsky L. Combination treatment with T4 and T3: toward personalized replacement therapy in hypothyroidism? The Journal of Clinical Endocrinology & Metabolism. 2012;97:2256–2271.
    1. Gullo D, Latina A, Frasca F, Moli R, Pellegriti G, Vigneri R. Levothyroxine monotherapy cannot guarantee euthyroidism in all athyreotic patients. PLoS One. 2011;6(8)e22552
    1. Kaplan MM, Sarne DH, Schneider AB. In search of the impossible dream? Thyroid hormone replacement therapy that treats all symptoms in all hypothyroid patients. The Journal of Clinical Endocrinology & Metabolism. 2003;88(10):4540–4542.
    1. Sawka AM, Gerstein HC, Marriott MJ, MacQueen GM, Joffe RT. Does a combination regimen of thyroxine (T4) and 3,5,3'-triiodothyronine improve depressive symptoms better than T4 alone in patients with hypothyroidism? Results of a double-blind, randomized, controlled trial. The Journal of Clinical Endocrinology & Metabolism. 2003;88(10):4551–4555.
    1. Walsh JP, Shiels L, Lim EM, et al. Combined thyroxine/liothyronine treatment does not improve well-being, quality of life, or cognitive function compared to thyroxine alone: a randomized controlled trial in patients with primary hypothyroidism. The Journal of Clinical Endocrinology & Metabolism. 2003;88(10):4543–4550.
    1. Appelhof BC, Fliers E, Wekking EM, et al. Combined therapy with levothyroxine and liothyronine in two ratios, compared with levothyroxine monotherapy in primary hypothyroidism: a double-blind, randomized, controlled clinical trial. The Journal of Clinical Endocrinology & Metabolism. 2005;90(5):2666–2674.
    1. Grozinsky-Glasberg S, Fraser A, Nahshoni E, Weizman A, Leibovici L. Thyroxine-triiodothyronine combination therapy versus thyroxine monotherapy for clinical hypothyroidism: meta-analysis of randomized controlled trials. The Journal of Clinical Endocrinology & Metabolism. 2006;91(7):2592–2599.
    1. Nygaard B, Jensen EW, Kvetny J, Jarløv A, Faber J. Effect of combination therapy with thyroxine (T4) and 3,5,3′-triiodothyronine versus T4 monotherapy in patients with hypothyroidism, a double-blind, randomised cross-over study. European Journal of Endocrinology. 2009;161(6):895–902.
    1. Fadeyev VV, Morgunova TB, Sytch JP, Melnichenko GA. TSH and thyroid hormones concentrations in patients with hypothyroidism receiving replacement therapy with L-thyroxine alone or in combination with L-triiodothyronine. Hormones. 2005;4(2):101–107.
    1. Chao M, Jiawei X, Xia H, et al. Thyroxine alone or thyroxine plus triiodothyronine replacement therapy for hypothyroidism. Nuclear Medicine Communications. 2009;30(8):586–593.
    1. Panicker V, Saravanan P, Vaidya B, et al. Common variation in the DIO2 gene predicts baseline psychological well-being and response to combination thyroxine plus triiodothyronine therapy in hypothyroid patients. The Journal of Clinical Endocrinology & Metabolism. 2009;94(5):1623–1629.
    1. Appelhof BC, Peeters RP, Wiersinga WM, et al. Polymorphisms in type 2 deiodinase are not associated with well-being, neurocognitive functioning, and preference for combined thyroxine/3,5,3′- triiodothyronine therapy. The Journal of Clinical Endocrinology & Metabolism. 2005;90(11):6296–6299.

Source: PubMed

3
Abonnieren