Skip to main content
Endocrinology logoLink to Endocrinology
. 2021 Mar 15;162(8):bqab059. doi: 10.1210/endocr/bqab059

Deiodinases and the Metabolic Code for Thyroid Hormone Action

Samuel C Russo 1, Federico Salas-Lucia 1, Antonio C Bianco 1,
PMCID: PMC8237994  PMID: 33720335

Abstract

Deiodinases modify the biological activity of thyroid hormone (TH) molecules, ie, they may activate thyroxine (T4) to 3,5,3′-triiodothyronine (T3), or they may inactivate T3 to 3,3′-diiodo-L-thyronine (T2) or T4 to reverse triiodothyronine (rT3). Although evidence of deiodination of T4 to T3 has been available since the 1950s, objective evidence of TH metabolism was not established until the 1970s. The modern paradigm considers that the deiodinases not only play a role in the homeostasis of circulating T3, but they also provide dynamic control of TH signaling: cells that express the activating type 2 deiodinase (D2) have enhanced TH signaling due to intracellular build-up of T3; the opposite is seen in cells that express type 3 deiodinase (D3), the inactivating deiodinase. D2 and D3 are expressed in metabolically relevant tissues such as brown adipose tissue, skeletal muscle and liver, and their roles have been investigated using cell, animal, and human models. During development, D2 and D3 expression customize for each tissue/organ the timing and intensity of TH signaling. In adult cells, D2 is induced by cyclic adenosine monophosphate (cAMP), and its expression is invariably associated with enhanced T3 signaling, expression of PGC1 and accelerated energy expenditure. In contrast, D3 expression is induced by hypoxia-inducible factor 1α (HIF-1a), dampening T3 signaling and the metabolic rate. The coordinated expression of these enzymes adjusts TH signaling in a time- and tissue-specific fashion, affecting metabolic pathways in health and disease states.

Keywords: thyroid, deiodinase, thyroxine, triiodothyronine, energy expenditure, metabolic rate, oxygen consumption, thermogenesis


An unusual group of patients with “low basal metabolism without myxedema” or “euthyroid hypometabolism” was identified in 1917 as the application of indirect calorimetry to thyroid diseases was being developed (1, 2). After excluding other endocrinopathies associated with slower metabolism, several individuals with lower basal metabolic rates remained. These patients did not appear to have myxedema and did not improve with the administration of desiccated thyroid extract or levothyroxine (LT4). The syndrome was tentatively attributed to insensitivities to thyroid-stimulating hormone (TSH) or thyroid hormone (TH), accelerated TH catabolism, or poor intestinal absorption of TH. Many years later, in 1955, physicians in Boston reported a few patients with this syndrome (3, 4). Basal metabolic rate ranged from −20 to −25% (normal range, −5 to +5%), and the patients complained of lethargy, fatigue, irritability, and sensitivity to cold. Remarkably, the basal metabolic rate in these patients did not respond to oral administration of LT4, but treatment with L-triiodothyronine (LT3) and combinations of LT4+LT3 resulted in striking clinical improvement. Inspired by findings from Pitt-Rivers et al (5), who observed that athyreotic patients could activate thyroxine (T4) to the more potent triiodothyronine (T3), the authors postulated a peripheral defect in T4 metabolism. Unfortunately, double-blinded placebo-controlled studies were not performed, which added to the retraction of the in vivo deiodination findings (6) and dampened interest in the hypothesis of impaired activation of thyroxine. It took another 15 years before this issue was revisited when Braverman et al (7) demonstrated unequivocally that T4 is activated to T3 in athyreotic patients.

It is fascinating that the role played by TH deiodination in metabolism had such an interesting but often overlooked debut. Outer ring deiodination (ORD) of T4 to T3 and inner ring deiodination (IRD) of T4 and T3 adjust TH signaling ordinarily through modulating intracellular T3 levels. This model has been studied mostly in cellular and animal settings, yet it is anticipated that deiodinases play similar roles in humans and participate in disease mechanisms. Two dramatic examples crystalize the clinical role played by the ORD and IRD pathways in TH signaling. The first is the activation of ORD via type 2 deiodinase (D2) in the lungs of patients with acute lung injury (8) or idiopathic pulmonary fibrosis (9), which led to treatment of 2 patients with acute respiratory distress syndrome caused by COVID-19 infection with aerosolized T3, with encouraging results (10). The second is the syndrome of consumptive hypothyroidism caused by the ectopic IRD reactivation via type 3 deiodinase (D3) in newborns with large hemangiomas (11).

Two important signaling pathways place the genes encoding D2 and D3, respectively, Dio2 and Dio3, centerstage in the control of cellular metabolism. Dio2 is downstream of the cyclic adenosine monophosphate (cAMP) cascade (12, 13), the key pathway that accelerates oxidative phosphorylation. In turn, Dio3 expression is accelerated by hypoxia-inducible factor 1α (HIF-1a) (14), the key molecule that orchestrates a metabolic switch to anaerobic processes such as hepatic glucose production and glycolysis. Only recently loss-of-function mutations were identified in the gene that encodes the type 1 deiodinase (D1), DIO1, without any detectable metabolic phenotype (15). Nonetheless, a prevalent polymorphism in DIO2, Thr92Ala-DIO2, reduces catalytic activity (16, 17) and leaves a genetic fingerprint in the brain (18). Notably, Neanderthals and Denisovans carried only the Ala-DIO2 allele. Whether this could have played an adaptive dietary role—their diet was mainly based on the consumption of meat, with a low intake of carbohydrates—remains to be elucidated (19). In this regard, it is striking that carriers of this polymorphism exhibit clear metabolic consequences, such as an association with insulin resistance (20) and type 2 diabetes mellitus (21). In addition, individuals with compounded Thr92Ala-DIO2 and the Trp64Arg β3-adrenergic receptor polymorphism, a receptor variant that generates less cAMP (22), exhibited higher body mass index (23). Not surprisingly, these associations have not been reproduced in all populations studied (24-29), and more and larger trials are needed to identify relevant interfering linkage factors (28, 30, 31).

The purpose of this review article is to briefly summarize the metabolic roles played by the deiodinases with an emphasis on findings reported in the last 5 years (2016-2020). These were identified through PubMed searches using “deiodinase,” “deiodination,” “energy metabolism,” “adipose tissue,” “liver,” “skeletal muscle,” and “hypothalamus” as keywords.

Thyroidal Secretion and Deiodinases Are Coordinated

While circulating TH levels are largely stable (32), “local” deiodinase-mediated mechanisms within target cells modulate TH signaling (extensively reviewed elsewhere (33-38)). Deiodinases are dimeric integral membrane selenoproteins that contain a single N-terminal transmembrane segment connected to a larger globular area that contains a thioredoxin-fold catalytic domain (39-42). This group of enzymes can activate (ORD via D1 or D2) or inactivate (IRD via D1 and D3) TH molecules once they have entered target cells (33, 34, 43, 44). Mechanistic details of the deiodination reaction are available elsewhere (39, 40, 45, 46).

DIO2 is expressed in the brain, pituitary gland, brown adipose tissue (BAT), skin, and skeletal and smooth muscles. Alternatively, DIO3 expression is found in most fetal tissues, subsiding after birth (47, 48). In adults, DIO3 expression is mostly in the brain, placenta, pregnant uterus, skin, and pancreatic beta cells (49, 50). Nonetheless, DIO3 is also generally expressed during clinical illness (51, 52). D1 is unique in that it exhibits an affinity for T4 that is 3 orders of magnitude lower than that exhibited by D2. In fact, D1’s preferred substrate is reverse triiodothyronine (rT3), which is reflected in the elevated serum rT3 levels observed in the D1-deficient (D1KO) mouse (53) and in patients with loss-of-function mutations in DIO1 (15).

The integrated actions of the thyroid gland and the deiodinases stabilize plasma T3 levels, preserving TH signaling and clinical euthyroidism in most tissues. On one hand, the hypothalamus-pituitary-thyroid (HPT) axis is particularly driven to defend serum T3 levels (54, 55), and TSH-induced thyroidal T3 secretion is the gateway through which the HPT axis controls systemic TH signaling (56). On the other hand, deiodinases regulate T3 production and clearance outside the thyroid parenchyma (33, 57, 58). This is possible because T3 flows across cellular membranes via specific transmembrane transporters. Deiodinases have their catalytic active site located in the cytosol (39, 40, 59), and T3 molecules produced via D1 or D2 eventually exit cells and mix with the circulating pool of T3. In contrast, D3-expressing cells function as sinks for T4 and T3, dampening local TH signaling and consuming circulating TH (60).

In hypothyroidism, there is an increase in the D2-mediated fractional conversion of T4 to T3, and a decrease in the D3-mediated clearance of T3 (61-64). Regulation of D2 is mostly posttranslational via interaction with T4 (65, 66), which causes conjugation to ubiquitin via K48-linked ubiquitin chains (67) and inactivation of D2 (68, 69). Once D2 is ubiquitinated, it is retrotranslocated to the cytoplasm via interaction with the p97-ATPase complex and taken up by 26S proteasomes (67). This is the mechanism through which T3 production fluctuates according to the availability of T4 (69-78).

Deiodinases Customize TH Signaling

Distinct subcellular localization likely explains why D2-generated T3 remains in the cells where it was produced much longer than D1-generated T3. Thus, D2-expressing cells are subject to tissue-specific dynamic changes in TH signaling without antecedent changes in circulating levels of THs. For example, the sympathetic nervous system stimulates Dio2 expression and T3 production in BAT that adds to the intracellular T3 entering from the circulation. As a result, cellular T3 content and thyroid hormone receptor (TR) occupancy increases from its baseline level of about 75% (79, 80) to >95%, along with induction of T3-responsive genes (81).

The role played by Dio2 activation in TH signaling can be visualized through bioluminescence in the TH action indicator (THAI) mouse model (82), a transgenic mouse ubiquitously expressing a luciferase reporter gene regulated by a strong T3 response element that operates in the context of endogenously expressed levels of TH transporters, TRs, and transcriptional co-regulators. Exposing these mice to cold (4 °C) caused tissue-specific bioluminescence in the interscapular region (iBAT), along with a ~3.0- to 9.0-fold increase in luciferase activity and mRNA in iBAT, which was eliminated after surgical denervation of the iBAT (82). Such a role for D2 in defining local TH signaling is not unique to BAT (33), as it is also seen in the developing cochlea (83) and liver (84), where there is a surge in D2-generated T3. In the adult mouse, D2-generated T3 also plays a role in brain, lung, skeletal muscle, and skeleton (85-88).

D3 is located in the plasma membrane, but during hypoxic conditions it can relocate to the cell nucleus and more efficiently reduce TH signaling (60, 89, 90). D3 limits TH signaling because of T4 and T3 inactivation. For example, induction of DIO3 in skin cells by members of the Hedgehog family of proteins reduces local TH signaling. This is because the mRNA levels for cyclin-D1, a gene that is negatively regulated by T3, increase upon induction of DIO3, and this increase is followed by proliferation of keratinocytes (91). Most regions of the D3-deficient (D3KO) brain experience reduced TH signaling transiently during late neonatal life, while the opposite is true during early development and later life (92). However, the fact that D3 is relatively abundant in the brain by virtue of being expressed in neurons does not prevent TRs from being almost fully saturated with T3 (93). Thus, future studies should expand and clarify the role played by D3 in modulating local TH signaling.

Food Availability Adjusts Thyroidal and Deiodinase-Mediated T3 Production

Caloric intake stimulates the thyroid system, coupling availability of energy substrates and TH signaling (94, 95). During fasting, the default thyroid activity is low, along with a slow rate of energy expenditure. Once caloric intake is resumed, activity is accelerated and circulating TH levels increase; for example, in patients recovering from anorexia nervosa (96), weight gain and serum T3 are closely associated with a faster energy expenditure (96).

Fasting in rodents is associated with decreased thyroidal (97) and extrathyroidal D2-mediated T3 production (98). The activity of the D1 pathway is reduced as well (99, 100) but that may be a result of reduced T3 levels, not the cause (97, 101). Nonetheless, the inactivation of both Dio1 and Dio2 pathways in mice does not prevent the drop in circulating levels of T4 and T3 during fasting, pointing toward a major role played by other mechanisms (102). For example, upregulation of D3 in skeletal muscle, liver, and kidney play a role as well, along with enzymes involved in glucuronidation and sulfation of iodothyronines (103-105). Accordingly, in rats, semistarvation and refeeding in catch-up fat (a phase of weight regain after significant weight loss characterized by a disproportionately high rate of body fat recovery relative to lean tissue recovery) (106) slow down T3 formation in skeleton muscles due primarily to upregulation in the IRD enzymes, DIO1, and/or DIO3. Fasting has also been found to increase D3 activity in the liver and white adipose tissue (107). Fasting-induced constitutive androstane receptor activation increased Dio3 expression and activity in mice and rat hepatocytes. In addition, inhibition of the mammalian target of rapamycin (mTOR) by fasting increased Dio3 expression in mice liver and white adipose tissue, but only slightly increased D3 activity in rat hepatocytes.

Notwithstanding the different nuances associated with each individual animal model, all models of food restriction develop low levels of circulating T3 and diminished TH signaling in most tissues, explaining the reduction in the rate of metabolism.

Carbohydrates and carbohydrate-related hormones are potent modulators of circulating T3 levels through deiodination (94, 95). Insulin stimulates D2 activity in rat brown adipocytes (108), and insulin sensitizers stimulate Dio2 expression in cultures of skeletal myocytes (109). In addition, BAT D2 is upregulated by insulin-like growth factor 1 (IGF-1) and insulin (108, 110), which activate nutrient sensing pathways such as the phosphatidylinositol 3-kinase (PI3K)/mTOR (111, 112) pathways. Studies in cells and mice revealed that Dio2 is inhibited by forkhead box, subgroup O1 (Foxo1), a transcriptional regulator that binds the Dio2 promoter. In turn, insulin signals through the PI3K–mammalian target of rapamycin complex 2 (mTORC2)–serine/threonine kinase 1 (AKT) pathway and relieves Foxo1 repression. Dio2 expression in the brain is not modified by fasting, indicating that Dio2 regulation by nutrient availability is not universal, likely occurring in tissues such as BAT, skeletal muscle and neonatal liver where the metabolic pathways are responsive to T3 and insulin (43, 84). In addition, in neonatal rat ventricular myocytes, Foxo1 directly upregulates Dio2 and downregulates Dio3, indicating that these mechanisms are tissue-specific (113). Thus, the balance between PI3K-mTORC2-AKT and Foxo1 signaling in metabolically relevant tissues should provide nutritional input and fine-tuning to the regulation of circulating levels of T3 and T3-dependent processes (Fig. 1).

Figure 1.

Figure 1.

Metabolic roles played by Dio2 in iBAT and skeletal muscle. D2 catalyzes T4 to T3 conversion inside cells, next to the nuclear compartment. D2-generated T3 enters the nucleus and binds to TRs, enhancing local thyroid hormone signaling. D2-T3 accelerates expression of PGC1 in both iBAT and skeletal muscle, increasing thermogenic capacity. In the iBAT, D2-T3 also activates expression of UCP1, the key uncoupling protein that increases heat production. Dio2 is a cAMP-inducible gene. Dio2 is also responsive to insulin, which in the skeletal muscle may contribute to circulating levels of T3. Dio2 expression is typically repressed by FOXO1 in skeletal muscle, which can be minimized by insulin-induced FOXO1 phosphorylation.

Tissue-Specific Metabolic Control

The expressions of Dio2 and Dio3 in the medial basal hypothalamus strategically place both enzymes at the crossroads of neural regulation of metabolism. For example, studies in mice show that food deprivation increases hypothalamic Dio2 mRNA levels and D2 activity (114). Hence, this localized increase in TH signaling could explain the reduction in thyrotropin-releasing hormone mRNA observed in fasted rats (115, 116). This mechanism might also regulate the torpor state in which there is reduction in metabolism and body temperature (117). In hamsters, hypothalamic Dio2 expression is decreased during spontaneous daily torpor as well as fasting-induced torpor (117). Notably, the global-D3KO mouse exhibits increased TH signaling in the hypothalamus with abnormal expression of genes in the melanocortin system, suggesting leptin resistance (118). They also have decreased adiposity, reduced BAT size, and accelerated fat loss in response to treatment with LT3. Global-D3KO mice display increased locomotor activity and an increased rate of energy expenditure along with expanded nighttime activity periods, suggesting a disrupted circadian rhythm (118).

Angiogenesis allows vessel density to reach the metabolic demands of the tissues via active sprouting of new vessels from endothelial cells, a process that in humans is regulated by TH (119). D2 is required for rapid stimulation of PI3K by T4 in human umbilical vein endothelial cells (120), which promotes endothelial cell migration. In addition, the production of pro-angiogenic factors such as vascular endothelial growth factor alpha is reduced in D2KO myoblasts. TH are also involved in neovascularization that occurs in tumor masses. Notably, D2 is highly expressed in intestinal polyps of a mouse model of familial adenomatous polyposis, and treatment with the deiodinase inhibitor iopanoic acid or chemical thyroidectomy suppresses tumor formation accompanied by reduced tumor cell proliferation and angiogenesis (121). Altogether, these data suggest that conversion of T4 to T3 by D2 plays a role in TH-induced angiogenesis in physiological and pathological conditions.

Brown Adipose Tissue

The ability to accelerate metabolic rate, a process known as adaptive thermogenesis (122), is accomplished through the release of norepinephrine in tissues such as BAT. There, norepinephrine induces the expression of cAMP-responsive genes that are part of the thermogenic program, such as Dio2, PGC1α, and UCP1 (123, 124) (Fig. 1). All 3 β-adrenergic receptor subtypes are involved, with each playing slightly different roles in adaptive thermogenesis and metabolic control (125-127).

Mice defend body temperature and survive in the cold thanks to the activation of a thermoregulatory response that critically depends on TH. Acutely cold-exposed thyroidectomized rats rapidly become hypothermic; LT4 is effective in restoring thermogenesis, given its activation to T3 in BAT via D2 (128). Circulating T3 plays a role, but norepinephrine-induced D2 acceleration and BAT T3 production are critical because they amplify cAMP production, therefore directly inducing UCP1 expression (129-131). Lipogenic enzymes are also stimulated (132-134) through a mechanism that involves T3 induction of the carbohydrate response element binding protein (135). BAT expresses both TRα and TRβ (136, 137) and activation of BAT thermogenesis depends on the coordinated effort between both TR isoforms (138, 139). Activation of TRβ induces the BAT thermogenic program eg, expression of Dio2, UCP1, and PGC1α, but amplification of cAMP production depends on activation of both TR isoforms (138). The activation of the thermogenic program in white adipose cells, also known as browning, can also be prompted by stimulation of TRβ and involves induction of Dio2 (140).

Survival of hypothyroid mice in the cold is only possible if cold exposure is gradually increased over time. In this case, it involves massive increase in sympathetic activity (141) and shivering (142). Animals in which Dio2 is globally inactivated (global-D2KO) are more sensitive to cold; they stimulate cold-induced thermogenesis even at room temperature (21°C), paradoxically rendering them resistant to diet-induced obesity. This phenotype is reversed by acclimatization at thermoneutrality (30 °C), which “turns off” sympathetic activity to the BAT. Only then, as a result of the unopposed reduction in TH signaling, global-D2KO mice become sensitized to diet-induced obesity, gaining excessive weight and also developing severe hepatic steatosis (143).

Having a fast-responding, cAMP-inducible, T3 generating system inside brown adipocytes (Dio2) is a key element of the extraordinary metabolic capacity exhibited by this tissue.

Liver

The global-D2KO mouse exhibits liver steatosis when placed at thermoneutrality and fed on a high fat diet (HFD) (143-145). Dio2 is not expressed in the normal adult liver, but it can be ectopically present in the liver of mice with targeted deletion of both liver X receptors (LXR) α and β (145). That LXR and RXR signaling inhibit Dio2 expression was confirmed by the observation that 22(R)-OH-cholesterol negatively regulates the human DIO2 promoter confirms that LXR and RXR signaling inhibit Dio2 expression (144). Remarkably, adipocyte fatty acid–binding protein (AFABP), an adipokine, induces expression of Dio2 in BAT via inhibition of the nuclear receptor LXRα, thereby increasing local TH signaling. AFABP accelerates thermogenesis by activating D2-mediated T3 production in brown adipocytes. The thermogenic responses to T4 are abrogated in Afabp-KO mice, but enhanced by AFABP (146).

Although not expressed in the adult mouse liver, a transient surge of Dio2 was detected at around the first day of life, activating local T4 to T3 conversion and TH signaling. This T3 surge occurs at a time in which serum T3 levels are half of what is observed in adult mice (147, 148); hence, it doubles local T3 concentration and modifies the expression of genes involved in broad aspects of hepatocyte function, including lipid metabolism (84, 149). Liver-specific Dio2 inactivation (Alb-D2KO) delays the expression of lipid-related genes and modifies baseline and long-term hepatic transcriptional response to a HFD. Feeding on a HFD normally affects the expression of approximately 400 hepatic genes involved in synthesis of fatty acids and triglycerides. In contrast, the response to HFD in the Alb-D2KO animals is restricted to a different set of only approximately 200 genes, which are associated with reverse cholesterol transport and lipase activity (84).

Subsequent studies revealed that the Alb-D2KO liver exhibits signs of epigenetic modifications, ie, about 1500 sites of DNA hypermethylation that explain the dramatic phenotype of the adult Alb-D2KO mouse with greatly reduced susceptibility to diet-induced steatosis, hypertriglyceridemia, and obesity (84). The sites of DNA hypermethylation seem to be caused by a perinatal increase in H3K9me3 levels in discrete chromatin areas (150). The change in DNA methylation pattern reduces chromatin accessibility, with reduction in the expression of several hundred genes. Thus, the postnatal surge in hepatic D2 activity and TH signaling prevents discrete DNA methylation and modifies the transcriptome of the adult mouse liver (150).

One of the repressed genes that underlies the Alb-D2KO phenotype is the gene encoding the zinc finger protein-125 (Zfp125) (84, 149). Zfp125 is a Foxo1-inducible transcriptional repressor that promotes lipid accumulation and liver steatosis in mice by reducing hepatic secretion of triglycerides and cholesterol (149). Zfp125 acts by repressing genes involved in lipoprotein structure and lipid binding and transport. While liver-specific knockdown of Zfp125 causes an “Alb-D2KO-like” metabolic phenotype, liver-specific normalization of Zfp125 expression in Alb-D2KO mice rescues the phenotype, restoring normal susceptibility to diet-induced obesity, liver steatosis, and hypercholesterolemia (149). In addition, mice with liver-specific Zfp125 knockdown exhibited a drop in respiratory quotient, reduced glycemia and pyruvate-stimulated liver glucose output, and higher levels of B-hydroxybutyrate (151). Liver-specific Zfp125 knockdown also amplified B-hydroxybutyrate production in response to 24 to 36 hours of fasting or feeding on a ketogenic diet. In isolated mouse hepatocytes, Zfp125 knockdown amplified the induction of ketogenesis by glucagon or insulin resistance, whereas Zfp125 overexpression amplified the expression of key gluconeogenic genes Pck1 and G6pc, placing Zfp125 at the center of fuel dysregulation of type 2 diabetes (151).

Overall, these studies indicate that the final differentiation of hepatoblasts to hepatocytes that occurs in the neonatal liver is influenced by TH signaling. The transient peak of D2-generated T3 is aligned with the overall positive effect of TH on liver lipogenesis and carries significant implications for future development of obesity and liver steatosis.

Zebrafish is another animal model in which this has been investigated, where Dio2 inactivation caused hyperglycemia due to reduced insulin sensitivity and expression of glycolytic enzymes in the skeletal muscles (152). As the fish aged, pancreatic islet size and β and α cell numbers increased along with insulin secretion. Glucagon receptors and downstream targets were also downregulated in the liver, indicating hepatic glucagon resistance and decreased gluconeogenesis (152).

Other Metabolically Relevant Tissues

The development of the flox-Dio2 and flox-Dio3 mice have allowed for a better understanding of the tissue-specific roles played by deiodinases (153, 154). Astro-D2KO mice (mice in which Dio2 has been inactivated in astrocytes) eat normally but exhibit a lower diurnal respiratory quotient and greater contributions of fatty acid oxidation to energy expenditure (155). In contrast, the Fat-D2KO mice (mice in which Dio2 has been inactivated in fat tissue) exhibit greater contribution of carbohydrate oxidation to energy expenditure. Furthermore, Fat-D2KO animals placed on HFDs gained more body weight and fat, suggesting impaired thermogenesis (possibly at the BAT level) and/or an inability to oxidize the fat excess (155).

DIO2 is expressed in human and murine skeletal muscle (33, 109, 156-158), but at levels at least 2 orders of magnitude lower than in the brain (159-161). While the role played by deiodinases in the context of skeletal muscle development seems clear (109, 158, 162-164), a metabolic role has not been well established. For example, Skm-D2KO mice (mice in which Dio2 has been inactivated in skeletal muscle) exhibited no apparent metabolic phenotype (155). The fact that there is BAT mixed with some muscle fibers (165), suggests that some of the local effects attributed to D2-generated T3 could be mediated by BAT tissue. With that in mind, Dio2 expression in skeletal muscle has been found to be induced by feeding on a methionine-restricted diet, which was associated with increased energy expenditure and protection against HFD-induced metabolic dysfunction (166).

Two models of induction of Dio2 in skeletal muscle have provided further understanding of the role played by this enzyme. The first is a cell model in which D2 expression is controlled by the “Tet-ON” system (167). D2 expression caused a shift from oxidative phosphorylation to glycolysis, with a consequent increase in the extracellular acidification rate; changes in metabolism coincided with a D2-induced shift from slow type-I to fast type-II myofibers, suggesting that D2 is essential in coordinating metabolic reprogramming of myocytes during myogenic differentiation (167). Indeed, in mice, Dio2 expression in the skeletal muscle can be induced by physical activity (86). A 20-minute treadmill exercise increased Dio2 expression/activity as well as PGC1α mRNA levels in rat and mouse skeletal muscle (Fig. 1). In contrast, induction of PGC1α was only partial (about 40% less) in the Skm-D2KO mice by acute and chronic treadmill exercise as well as in primary Skm-D2KO myocytes stimulated with cAMP (86). These findings that Dio2 expression mediates part of PGC1α induction by treadmill exercise are reminiscent of similar observation in BAT and lungs. Induction of PGC1α in BAT (168) and in the lung (9) is only partial in mice with Dio2 inactivation, suggesting a common pathway through which Dio2 affects mitochondrial biogenesis and metabolism.

Dio3 is expressed in embryonic and adult human and murine pancreatic β-cells (49, 50). It is conceivable that its presence minimizes the inhibitory effects of T3 on insulin secretion observed in isolated mature murine pancreatic islets (49, 50) and in MIN6 insulinoma cells (169). Dio3 expression in MIN6 cells is stimulated by glucagon-like peptide-1 and by Exendin-4, a glucagon-like peptide-1 receptor agonist (169). Mice with Dio3 inactivation in pancreatic islets are glucose intolerant and have impaired glucose-stimulated insulin secretion (49, 50).

Other Metabolic Signals Affect Dio2 Expression and Local TH Signaling

Endogenous and exogenous molecules that activate the Dio2 pathway include bile acids (170), flavonols (171), chemical chaperones (172), and the adipokine adipocyte-specific fatty acid–binding protein (AFABP) (146). These molecules have been linked to activation of D2-generated T3 and acceleration of energy expenditure (170), and protection against obesity (173). In vitro treatment of primary human brown adipocytes with chenodeoxycholic acid (CDCA) or specific TGR5 agonists increased mitochondrial uncoupling and DIO2 expression, an effect that was absent in human primary white adipocytes (174). Studies in humans support the idea that bile acids play a role in controlling energy expenditure (174, 175). Along the same lines but through a different pathway, kaempferol and other flavonols stimulate DIO2 expression via a cAMP-mediated mechanism in primary cultures of human skeletal myocytes, leading to D2-mediated T3 production, expression of thermogenic relevant genes, and acceleration of O2 consumption (171).

Endoplasmic reticulum (ER) stress may be a link between metabolic homeostasis and D2-mediated TH signaling (17, 176). From a metabolic perspective, ER stress can be triggered by HFD and obesity, contributing with downregulation of insulin sensitivity (177). It is notable that ER stress leads to a rapid loss of D2 activity without affecting Dio2 mRNA levels, resulting in a drop in D2-mediated T3 production (176). The drop in D2 activity requires eukaryotic initiation factor 2, which blocks Dio2 mRNA translation, hence inhibiting synthesis of D2.

Conclusions

Deiodinases can augment and reduce TH signaling in a tissue-specific fashion while plasma T3 levels remain tranquil. Although a hypothetical metabolic role was linked to the pathway that converts T4 to T3 in the 1950s, objective evidence that this was the case was only developed decades later. Several settings using cell, animal and human models illuminate how activation of T4 to T3 and inactivation of T4 and T3 play physiological and pathophysiological roles at various life moments. Further understanding of these mechanisms will allow a more refined therapeutic control of TH signaling with enormous implications for health and disease states.

Acknowledgments

Financial Support: This work was in part supported through grants from the NIDDK 58538, 65055.

Glossary

Abbreviations

AFABP

adipocyte fatty acid-binding protein

BAT

brown adipose tissue

cAMP

cyclic adenosine monophosphate

D1

type 1 deiodinase

D2

type 2 deiodinase (activating)

D3

type 3 deiodinase (inactivating)

ER

endoplasmic reticulum

FOXO1

forkhead box O1

HFD

high-fat diet

iBAT

interscapular brown adipose tissue

IRD

inner ring deiodination

LT3

L-triiodothyronine

LT4

levothyroxine

LXR

liver X receptor

mTOR

mammalian target of rapamycin

ORD

outer ring deiodination

PI3K

phosphatidylinositol 3-kinase

rT3

reverse triiodothyronine

T3

triiodothyronine

T4

thyroxine

TH

thyroid hormone

TR

thyroid hormone receptor

TSH

thyrotropin (thyroid-stimulating hormone)

Zfp125

zinc finger protein-125

Additional Information

Disclosures: Dr. Bianco is a consultant for Synthonics Inc, Allergan Inc, and BLA Technology LLC; the other authors have no disclosures.

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

  • 1. Thompson  WO, Thompson  PK. LOW BASAL METABOLISM FOLLOWING THYROTOXICOSIS: I. Temporary type without myxedema, with special reference to the rôle of iodine therapy. J Clin Invest.  1928;5(3):441-469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Thompson  WO, Thompson  PK. LOW BASAL METABOLISM FOLLOWING THYROTOXICOSIS: II. Permanent type without myxedema. J Clin Invest.  1928;5(3):471-501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Kurland  GS, Hamolsky  MW, Freedberg  AS. Studies in non-myxedematous hypometabolism. I. The clinical syndrome and the effects of thiiodothyronine, alone or combined with thyroxine. J Clin Endocrinol Metab.  1955;15(11):1354-1360. [DOI] [PubMed] [Google Scholar]
  • 4. Freedberg  AS, Kurland  GS, Hamolsky  MW. Effect of 1-tri-iodothyronine alone and combined with 1-thyroxine in nonmyxedematous hypometabolism; preliminary report. N Engl J Med.  1955;253(2):57-60. [DOI] [PubMed] [Google Scholar]
  • 5. Pitt-Rivers  R, Stanbury  JB, Rapp  B. Conversion of thyroxine to 3-5-3’-triiodothyronine in vivo. J Clin Endocrinol Metab.  1955;15(5):616-620. [DOI] [PubMed] [Google Scholar]
  • 6. lassiter  WR, Stanbury  JB. The in vivo conversion of thyroxine to 3:5:3’triiodothyronine. J Clin Endocrinol Metab.  1958;18(8):903-906. [DOI] [PubMed] [Google Scholar]
  • 7. Braverman  LE, Ingbar  SH, Sterling  K. Conversion of thyroxine (T4) to triiodothyronine (T3) in athyreotic subjects. J Clin Invest.  1970;49:855-864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Ma  SF, Xie  L, Pino-Yanes  M, et al.  Type 2 deiodinase and host responses of sepsis and acute lung injury. Am J Respir Cell Mol Biol.  2011;45(6):1203-1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Yu  G, Tzouvelekis  A, Wang  R, et al.  Thyroid hormone inhibits lung fibrosis in mice by improving epithelial mitochondrial function. Nat Med.  2018;24(1):39-49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Ingbar  D. Data from: Treatment of ARDS With Instilled T3 (ARDS+T3). 2020. ClinicalTrials.gov.
  • 11. Huang  SA, Tu  HM, Harney  JW, et al.  Severe hypothyroidism caused by type 3 iodothyronine deiodinase in infantile hemangiomas. N Engl J Med.  2000;343(3):185-189. [DOI] [PubMed] [Google Scholar]
  • 12. Bartha  T, Kim  SW, Salvatore  D, et al.  Characterization of the 5’-flanking and 5’-untranslated regions of the cyclic adenosine 3’,5’-monophosphate-responsive human type 2 iodothyronine deiodinase gene1. Endocrinology.  2000;141(1):229-237. [DOI] [PubMed] [Google Scholar]
  • 13. Canettieri  G, Celi  FS, Baccheschi  G, Salvatori  L, Andreoli  M, Centanni  M. Isolation of human type 2 deiodinase gene promoter and characterization of a functional cyclic adenosine monophosphate response element. Endocrinology.  2000;141(5):1804-1813. [DOI] [PubMed] [Google Scholar]
  • 14. Simonides  WS, Mulcahey  MA, Redout  EM, et al.  Hypoxia-inducible factor induces local thyroid hormone inactivation during hypoxic-ischemic disease in rats. J Clin Invest.  2008;118(3):975-983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Franca  MM, German  A, Fernandes  GW, et al.  Human type 1 Iodothyronine Deiodinase (DIO1) mutations cause abnormal thyroid hormone metabolism. Thyroid.  2021;31(2):202-207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Castagna  MG, Dentice  M, Cantara  S, et al.  DIO2 Thr92Ala reduces deiodinase-2 activity and serum-t3 levels in thyroid-deficient patients. J Clin Endocrinol Metab.  2017;102(5): 1623-1630. [DOI] [PubMed] [Google Scholar]
  • 17. Jo  S, Fonseca  TL, Da Costa Bocco  BM, et al.  Type 2 deiodinase polymorphism causes ER stress and hypothyroidism in the brain. J Clin Invest. 2019;129(1):230-245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. McAninch  EA, Jo  S, Preite  NZ, et al.  Prevalent polymorphism in thyroid hormone-activating enzyme leaves a genetic fingerprint that underlies associated clinical syndromes. J Clin Endocrinol Metab.  2015;100(3):920-933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Ricci  C, Kakularam  KR, Marzocchi  C, et al.  Thr92Ala polymorphism in the type 2 deiodinase gene: an evolutionary perspective. J Endocrinol Invest.  2020;43(12):1749-1757. [DOI] [PubMed] [Google Scholar]
  • 20. Peeters  RP, van der Deure  WM, Visser  TJ. Genetic variation in thyroid hormone pathway genes; polymorphisms in the TSH receptor and the iodothyronine deiodinases. Eur J Endocrinol.  2006;155(5):655-662. [DOI] [PubMed] [Google Scholar]
  • 21. Canani  LH, Capp  C, Dora  JM, et al.  The type 2 deiodinase A/G (Thr92Ala) polymorphism is associated with decreased enzyme velocity and increased insulin resistance in patients with type 2 diabetes mellitus. J Clin Endocrinol Metab.  2005;90(6):3472-3478. [DOI] [PubMed] [Google Scholar]
  • 22. Kimura  K, Sasaki  N, Asano  A, et al.  Mutated human beta3-adrenergic receptor (Trp64Arg) lowers the response to beta3-adrenergic agonists in transfected 3T3-L1 preadipocytes. Horm Metab Res.  2000;32(3):91-96. [DOI] [PubMed] [Google Scholar]
  • 23. Mentuccia  D, Proietti-Pannunzi  L, Tanner  K, et al.  Association between a novel variant of the human type 2 deiodinase gene Thr92Ala and insulin resistance: evidence of interaction with the Trp64Arg variant of the beta-3-adrenergic receptor. Diabetes.  2002;51(3):880-883. [DOI] [PubMed] [Google Scholar]
  • 24. Canani  LH, Leie  MA, Machado  WE, Capp  C, Maia  AL. Type 2 deiodinase Thr92Ala polymorphism is not associated with arterial hypertension in type 2 diabetes mellitus patients. Hypertension.  2007;49(6):e47; author reply e48. [DOI] [PubMed] [Google Scholar]
  • 25. Grarup  N, Andersen  MK, Andreasen  CH, et al.  Studies of the common DIO2 Thr92Ala polymorphism and metabolic phenotypes in 7342 Danish white subjects. J Clin Endocrinol Metab.  2007;92(1):363-366. [DOI] [PubMed] [Google Scholar]
  • 26. Gumieniak  O, Perlstein  TS, Williams  JS, et al.  Ala92 type 2 deiodinase allele increases risk for the development of hypertension. Hypertension. 2007;49(3):461-466. [DOI] [PubMed] [Google Scholar]
  • 27. Heemstra  KA, Hoftijzer  HC, van der Deure  WM, et al.  Thr92Ala polymorphism in the type 2 deiodinase is not associated with T4 dose in athyroid patients or patients with Hashimoto thyroiditis. Clin Endocrinol (Oxf).  2009;71(2):279-283. [DOI] [PubMed] [Google Scholar]
  • 28. Nair  S, Muller  YL, Ortega  E, Kobes  S, Bogardus  C, Baier  LJ. Association analyses of variants in the DIO2 gene with early-onset type 2 diabetes mellitus in Pima Indians. Thyroid.  2012;22(1):80-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. van der Deure  WM, Peeters  RP, Uitterlinden  AG, et al.  Impact of thyroid function and polymorphisms in the type 2 deiodinase on blood pressure: the Rotterdam Study and the Rotterdam Scan Study. Clin Endocrinol (Oxf).  2009;71(1):137-144. [DOI] [PubMed] [Google Scholar]
  • 30. Mentuccia  D, Thomas  MJ, Coppotelli  G, et al.  The Thr92Ala deiodinase type 2 (DIO2) variant is not associated with type 2 diabetes or indices of insulin resistance in the old order of Amish. Thyroid.  2005;15(11):1223-1227. [DOI] [PubMed] [Google Scholar]
  • 31. Bianco  AC, Kim  BS. Pathophysiological relevance of deiodinase polymorphism. Curr Opin Endocrinol Diabetes Obes.  2018;25(5):341-346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Martial  JA, Seeberg  PH, Guenzi  D, Goodman  HM, Baxter  JD. Regulation of growth hormone gene expression: synergistic effects of thyroid and glucocorticoid hormone. Proc Natl Acad Sci.  1977;74:4293-4295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Gereben  B, Zavacki  AM, Ribich  S, et al.  Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling. Endocr Rev.  2008;29(7):898-938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Bianco  AC, Salvatore  D, Gereben  B, Berry  MJ, Larsen  PR. Biochemistry, cellular and molecular biology, and physiological roles of the iodothyronine selenodeiodinases. Endocr Rev.  2002;23(1):38-89. [DOI] [PubMed] [Google Scholar]
  • 35. Peeters  RP, Visser  TJ.. Metabolism of thyroid hormone. In: De Groot  LJ, Chrousos  G, Dungan  K, et al. , eds. South Dartmouth (MA): Endotext, 2000. [Google Scholar]
  • 36. Galton  VA. The ups and downs of the thyroxine pro-hormone hypothesis. Mol Cell Endocrinol.  2017;458:105-111. [DOI] [PubMed] [Google Scholar]
  • 37. St Germain  DL, Galton  VA, Hernandez  A. Minireview: Defining the roles of the iodothyronine deiodinases: current concepts and challenges. Endocrinology.  2009;150(3):1097-1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Bianco  AC, Dumitrescu  A, Gereben  B, et al.  Paradigms of dynamic control of thyroid hormone signaling. Endocr Rev.  2019;40(4):1000-1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Callebaut  I, Curcio-Morelli  C, Mornon  JP, et al.  The iodothyronine selenodeiodinases are thioredoxin-fold family proteins containing a glycoside hydrolase clan GH-A-like structure. J Biol Chem.  2003;278(38):36887-36896. [DOI] [PubMed] [Google Scholar]
  • 40. Schweizer  U, Schlicker  C, Braun  D, Köhrle  J, Steegborn  C. Crystal structure of mammalian selenocysteine-dependent iodothyronine deiodinase suggests a peroxiredoxin-like catalytic mechanism. Proc Natl Acad Sci U S A.  2014;111(29): 10526-10531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Curcio-Morelli  C, Gereben  B, Zavacki  AM, et al.  In vivo dimerization of types 1, 2, and 3 iodothyronine selenodeiodinases. Endocrinology.  2003;144(3):937-946. [DOI] [PubMed] [Google Scholar]
  • 42. Sagar  GD, Gereben  B, Callebaut  I, et al.  The thyroid hormone-inactivating deiodinase functions as a homodimer. Mol Endocrinol.  2008;22(6):1382-1393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Mullur  R, Liu  YY, Brent  GA. Thyroid hormone regulation of metabolism. Physiol Rev.  2014;94(2):355-382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Groeneweg  S, Visser  WE, Visser  TJ. Disorder of thyroid hormone transport into the tissues. Best Pract Res Clin Endocrinol Metab.  2017;31(2):241-253. [DOI] [PubMed] [Google Scholar]
  • 45. Bayse  CA, Marsan  ES, Garcia  JR, Tran-Thompson  AT. Thyroxine binding to type III iodothyronine deiodinase. Sci Rep.  2020;10(1):15401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Schweizer  U, Steegborn  C. New insights into the structure and mechanism of iodothyronine deiodinases. J Mol Endocrinol.  2015;55(3):R37-R52. [DOI] [PubMed] [Google Scholar]
  • 47. Becker  KB, Stephens  KC, Davey  JC, Schneider  MJ, Galton  VA. The type 2 and type 3 iodothyronine deiodinases play important roles in coordinating development in Rana catesbeiana tadpoles. Endocrinology.  1997;138(7):2989-2997. [DOI] [PubMed] [Google Scholar]
  • 48. Darras  VM, Hume  R, Visser  TJ. Regulation of thyroid hormone metabolism during fetal development. Mol Cell Endocrinol.  1999;151(1-2):37-47. [DOI] [PubMed] [Google Scholar]
  • 49. Medina  MC, Fonesca  TL, Molina  J, et al.  Maternal inheritance of an inactive type III deiodinase gene allele affects mouse pancreatic β-cells and disrupts glucose homeostasis. Endocrinology.  2014;155(8):3160-3171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Medina  MC, Molina  J, Gadea  Y, et al.  The thyroid hormone-inactivating type III deiodinase is expressed in mouse and human beta-cells and its targeted inactivation impairs insulin secretion. Endocrinology.  2011;152(10):3717-3727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Peeters  RP, Wouters  PJ, van Toor  H, Kaptein  E, Visser  TJ, Van den Berghe  G. Serum 3,3’,5’-triiodothyronine (rT3) and 3,5,3’-triiodothyronine/rT3 are prognostic markers in critically ill patients and are associated with postmortem tissue deiodinase activities. J Clin Endocrinol Metab.  2005;90(8):4559-4565. [DOI] [PubMed] [Google Scholar]
  • 52. Peeters  RP, Wouters  PJ, Kaptein  E, van Toor  H, Visser  TJ, Van den Berghe  G. Reduced activation and increased inactivation of thyroid hormone in tissues of critically ill patients. J Clin Endocrinol Metab.  2003;88(7):3202-3211. [DOI] [PubMed] [Google Scholar]
  • 53. Schneider  MJ, Fiering  SN, Thai  B, et al.  Targeted disruption of the type 1 selenodeiodinase gene (Dio1) results in marked changes in thyroid hormone economy in mice. Endocrinology.  2006;147(1):580-589. [DOI] [PubMed] [Google Scholar]
  • 54. Christoffolete  MA, Arrojo e Drigo  R, Gazoni  F, et al.  Mice with impaired extrathyroidal thyroxine to 3,5,3’-triiodothyronine conversion maintain normal serum 3,5,3’-triiodothyronine concentrations. Endocrinology.  2007;148(3):954-960. [DOI] [PubMed] [Google Scholar]
  • 55. Abdalla  SM, Bianco  AC. Defending plasma T3 is a biological priority. Clin Endocrinol (Oxf).  2014;81(5):633-641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Citterio  CE, Morishita  Y, Dakka  N, Veluswamy  B, Arvan  P. Relationship between the dimerization of thyroglobulin and its ability to form triiodothyronine. J Biol Chem.  2018;293(13):4860-4869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Gereben  B, Salvatore  D. Pretranslational regulation of type 2 deiodinase. Thyroid.  2005;15(8):855-864. [DOI] [PubMed] [Google Scholar]
  • 58. Huang  SA. Physiology and pathophysiology of type 3 deiodinase in humans. Thyroid.  2005;15(8):875-881. [DOI] [PubMed] [Google Scholar]
  • 59. Baqui  MM, Gereben  B, Harney  JW, Larsen  PR, Bianco  AC. Distinct subcellular localization of transiently expressed types 1 and 2 iodothyronine deiodinases as determined by immunofluorescence confocal microscopy. Endocrinology.  2000;141(11):4309-4312. [DOI] [PubMed] [Google Scholar]
  • 60. Jo  S, Kalló  I, Bardóczi  Z, et al.  Neuronal hypoxia induces Hsp40-mediated nuclear import of type 3 deiodinase as an adaptive mechanism to reduce cellular metabolism. J Neurosci.  2012;32(25):8491-8500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Inada  M, Kasagi  K, Kurata  S, et al.  Estimation of thyroxine and triiodothyronine distribution and of the conversion rate of thyroxine to triiodothyronine in man. J Clin Invest.  1975;55(6):1337-1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Nicoloff  JT, Lum  SM, Spencer  CA, Morris  R. Peripheral autoregulation of thyroxine to triiodothyronine conversion in man. Horm Metab Res Suppl.  1984;14:74-79. [PubMed] [Google Scholar]
  • 63. Lum  SM, Nicoloff  JT, Spencer  CA, Kaptein  EM. Peripheral tissue mechanism for maintenance of serum triiodothyronine values in a thyroxine-deficient state in man. J Clin Invest.  1984;73(2):570-575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Peeters  R, Fekete  C, Goncalves  C, et al.  Regional physiological adaptation of the central nervous system deiodinases to iodine deficiency. Am J Physiol Endocrinol Metab.  2001;281(1):E54-E61. [DOI] [PubMed] [Google Scholar]
  • 65. Steinsapir  J, Bianco  AC, Buettner  C, Harney  J, Larsen  PR. Substrate-induced down-regulation of human type 2 deiodinase (hD2) is mediated through proteasomal degradation and requires interaction with the enzyme’s active center. Endocrinology.  2000;141(3):1127-1135. [DOI] [PubMed] [Google Scholar]
  • 66. Steinsapir  J, Harney  J, Larsen  PR. Type 2 iodothyronine deiodinase in rat pituitary tumor cells is inactivated in proteasomes. J Clin Invest.  1998;102(11):1895-1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Arrojo E Drigo  R, Egri  P, Jo  S, Gereben  B, Bianco  AC. The type II deiodinase is retrotranslocated to the cytoplasm and proteasomes via p97/Atx3 complex. Mol Endocrinol.  2013;27(12):2105-2115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Sagar  GD, Gereben  B, Callebaut  I, et al.  Ubiquitination-induced conformational change within the deiodinase dimer is a switch regulating enzyme activity. Mol Cell Biol.  2007;27(13):4774-4783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Gereben  B, Goncalves  C, Harney  JW, Larsen  PR, Bianco  AC. Selective proteolysis of human type 2 deiodinase: a novel ubiquitin-proteasomal mediated mechanism for regulation of hormone activation. Mol Endocrinol.  2000;14(11):1697-1708. [DOI] [PubMed] [Google Scholar]
  • 70. McAninch  EA, Bianco  AC. New insights into the variable effectiveness of levothyroxine monotherapy for hypothyroidism. Lancet Diabetes Endocrinol.  2015;3(10):756-758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Gereben  B, McAninch  EA, Ribeiro  MO, Bianco  AC. Scope and limitations of iodothyronine deiodinases in hypothyroidism. Nat Rev Endocrinol.  2015;11(11):642-652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Zeöld  A, Pormüller  L, Dentice  M, et al.  Metabolic instability of type 2 deiodinase is transferable to stable proteins independently of subcellular localization. J Biol Chem.  2006;281(42):31538-31543. [DOI] [PubMed] [Google Scholar]
  • 73. Dentice  M, Bandyopadhyay  A, Gereben  B, et al.  The Hedgehog-inducible ubiquitin ligase subunit WSB-1 modulates thyroid hormone activation and PTHrP secretion in the developing growth plate. Nat Cell Biol.  2005;7(7):698-705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Botero  D, Gereben  B, Goncalves  C, De Jesus  LA, Harney  JW, Bianco  AC. Ubc6p and ubc7p are required for normal and substrate-induced endoplasmic reticulum-associated degradation of the human selenoprotein type 2 iodothyronine monodeiodinase. Mol Endocrinol.  2002;16(9):1999-2007. [DOI] [PubMed] [Google Scholar]
  • 75. Kim  BW, Zavacki  AM, Curcio-Morelli  C, et al.  Endoplasmic reticulum-associated degradation of the human type 2 iodothyronine deiodinase (D2) is mediated via an association between mammalian UBC7 and the carboxyl region of D2. Mol Endocrinol.  2003;17(12):2603-2612. [DOI] [PubMed] [Google Scholar]
  • 76. Zavacki  AM, Arrojo E Drigo  R, Freitas  BC, et al.  The E3 ubiquitin ligase TEB4 mediates degradation of type 2 iodothyronine deiodinase. Mol Cell Biol.  2009;29(19):5339-5347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Egri  P, Gereben  B. Minimal requirements for ubiquitination-mediated regulation of thyroid hormone activation. J Mol Endocrinol.  2014;53(2):217-226. [DOI] [PubMed] [Google Scholar]
  • 78. Curcio-Morelli  C, Zavacki  AM, Christofollete  M, et al.  Deubiquitination of type 2 iodothyronine deiodinase by von Hippel-Lindau protein-interacting deubiquitinating enzymes regulates thyroid hormone activation. J Clin Invest.  2003;112(2):189-196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Larsen  PR, Silva  JE, Kaplan  MM. Relationships between circulating and intracellular thyroid hormones: physiological and clinical implications. Endocr Rev.  1981;2(1):87-102. [DOI] [PubMed] [Google Scholar]
  • 80. Bianco  AC, Silva  JE. Nuclear 3,5,3’-triiodothyronine (T3) in brown adipose tissue: receptor occupancy and sources of T3 as determined by in vivo techniques. Endocrinology.  1987;120(1):55-62. [DOI] [PubMed] [Google Scholar]
  • 81. Bianco  AC, Silva  JE. Cold exposure rapidly induces virtual saturation of brown adipose tissue nuclear T3 receptors. Am J Physiol.  1988;255(4 Pt 1):E496-E503. [DOI] [PubMed] [Google Scholar]
  • 82. Mohácsik  P, Erdélyi  F, Baranyi  M, et al.  A transgenic mouse model for detection of tissue-specific thyroid hormone action. Endocrinology.  2018;159(2):1159-1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Campos-Barros  A, Amma  LL, Faris  JS, Shailam  R, Kelley  MW, Forrest  D. Type 2 iodothyronine deiodinase expression in the cochlea before the onset of hearing. Proc Natl Acad Sci U S A.  2000;97(3):1287-1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Fonseca  TL, Fernandes  GW, McAninch  EA, et al.  Perinatal deiodinase 2 expression in hepatocytes defines epigenetic susceptibility to liver steatosis and obesity. Proc Natl Acad Sci U S A.  2015;112(45):14018-14023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Bocco  BM, Werneck-de-Castro  JP, Oliveira  KC, et al.  Type 2 deiodinase disruption in astrocytes results in anxiety-depressive-like behavior in male mice. Endocrinology.  2016;157(9):3682-3695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Bocco  BM, Louzada  RA, Silvestre  DH, et al.  Thyroid hormone activation by type 2 deiodinase mediates exercise-induced peroxisome proliferator-activated receptor-γ coactivator-1α expression in skeletal muscle. J Physiol.  2016;594(18):5255-5269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Bassett  JH, Boyde  A, Howell  PG, et al.  Optimal bone strength and mineralization requires the type 2 iodothyronine deiodinase in osteoblasts. Proc Natl Acad Sci U S A.  2010;107(16):7604-7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Galton  VA. The roles of the iodothyronine deiodinases in mammalian development. Thyroid.  2005;15(8):823-834. [DOI] [PubMed] [Google Scholar]
  • 89. Freitas  BC, Gereben  B, Castillo  M, et al.  Paracrine signaling by glial cell-derived triiodothyronine activates neuronal gene expression in the rodent brain and human cells. J Clin Invest.  2010;120(6):2206-2217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Kalló  I, Mohácsik  P, Vida  B, et al.  A novel pathway regulates thyroid hormone availability in rat and human hypothalamic neurosecretory neurons. PLoS One.  2012;7(6):e37860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Dentice  M, Luongo  C, Huang  S, et al.  Sonic hedgehog-induced type 3 deiodinase blocks thyroid hormone action enhancing proliferation of normal and malignant keratinocytes. Proc Natl Acad Sci U S A.  2007;104(36):14466-14471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Hernandez  A, Quignodon  L, Martinez  ME, Flamant  F, St Germain  DL. Type 3 deiodinase deficiency causes spatial and temporal alterations in brain T3 signaling that are dissociated from serum thyroid hormone levels. Endocrinology.  2010;151(11):5550-5558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Crantz  FR, Silva  JE, Larsen  PR. An analysis of the sources and quantity of 3,5,3’-triiodothyronine specifically bound to nuclear receptors in rat cerebral cortex and cerebellum. Endocrinology.  1982;110(2):367-375. [DOI] [PubMed] [Google Scholar]
  • 94. Spaulding  SW, Chopra  IJ, Sherwin  RS, Lyall  SS. Effect of caloric restriction and dietary composition of serum T3 and reverse T3 in man. J Clin Endocrinol Metab.  1976;42(1):197-200. [DOI] [PubMed] [Google Scholar]
  • 95. Danforth  E  Jr, Horton  ES, O’Connell  M, et al.  Dietary-induced alterations in thyroid hormone metabolism during overnutrition. J Clin Invest.  1979;64(5):1336-1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Schebendach  JE, Golden  NH, Jacobson  MS, Hertz  S, Shenker  IR. The metabolic responses to starvation and refeeding in adolescents with anorexia nervosa. Ann N Y Acad Sci.  1997;817:110-119. [DOI] [PubMed] [Google Scholar]
  • 97. Kinlaw  WB, Schwartz  HL, Oppenheimer  JH. Decreased serum triiodothyronine in starving rats is due primarily to diminished thyroidal secretion of thyroxine. J Clin Invest.  1985;75(4):1238-1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Lartey  LJ, Werneck-de-Castro  JP, O-Sullivan  I, Unterman  TG, Bianco  AC. Coupling between nutrient availability and thyroid hormone activation. J Biol Chem.  2015;290(51): 30551-30561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Balsam  A, Ingbar  SH. The influence of fasting, diabetes, and several pharmacological agents on the pathways of thyroxine metabolism in rat liver. J Clin Invest.  1978;62(2):415-424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. de Vries  EM, van Beeren  HC, Ackermans  MT, Kalsbeek  A, Fliers  E, Boelen  A. Differential effects of fasting vs food restriction on liver thyroid hormone metabolism in male rats. J Endocrinol.  2015;224(1):25-35. [DOI] [PubMed] [Google Scholar]
  • 101. Zavacki  AM, Ying  H, Christoffolete  MA, et al.  Type 1 iodothyronine deiodinase is a sensitive marker of peripheral thyroid status in the mouse. Endocrinology.  2005;146(3): 1568-1575. [DOI] [PubMed] [Google Scholar]
  • 102. Galton  VA, Hernandez  A, St Germain  DL. The 5’-deiodinases are not essential for the fasting-induced decrease in circulating thyroid hormone levels in male mice: possible roles for the type 3 deiodinase and tissue sequestration of hormone. Endocrinology.  2014;155(8):3172-3181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Boelen  A, van Beeren  M, Vos  X, et al.  Leptin administration restores the fasting-induced increase of hepatic type 3 deiodinase expression in mice. Thyroid.  2012;22(2):192-199. [DOI] [PubMed] [Google Scholar]
  • 104. Vella  KR, Ramadoss  P, Lam  FS, et al.  NPY and MC4R signaling regulate thyroid hormone levels during fasting through both central and peripheral pathways. Cell Metab.  2011;14(6):780-790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. De Andrade  PB, Neff  LA, Strosova  MK, et al.  Caloric restriction induces energy-sparing alterations in skeletal muscle contraction, fiber composition and local thyroid hormone metabolism that persist during catch-up fat upon refeeding. Front Physiol.  2015;6:254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Calonne  J, Arsenijevic  D, Scerri  I, Miles-Chan  JL, Montani  JP, Dulloo  AG. Low 24-hour core body temperature as a thrifty metabolic trait driving catch-up fat during weight regain after caloric restriction. Am J Physiol Endocrinol Metab.  2019;317(4):E699-E709. [DOI] [PubMed] [Google Scholar]
  • 107. de Vries  EM, van Beeren  HC, van Wijk  ACWA, et al.  Regulation of type 3 deiodinase in rodent liver and adipose tissue during fasting. Endocr Connect.  2020;9(6):552-562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Mills  I, Barge  RM, Silva  JE, Larsen  PR. Insulin stimulation of iodothyronine 5’-deiodinase in rat brown adipocytes. Biochem Biophys Res Commun.  1987;143(1):81-86. [DOI] [PubMed] [Google Scholar]
  • 109. Grozovsky  R, Ribich  S, Rosene  ML, et al.  Type 2 deiodinase expression is induced by peroxisomal proliferator-activated receptor-gamma agonists in skeletal myocytes. Endocrinology.  2009;150(4):1976-1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. 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. J Mol Endocrinol.  2005;34(1):139-151. [DOI] [PubMed] [Google Scholar]
  • 111. Laplante  M, Sabatini  DM. mTOR signaling at a glance. J Cell Sci.  2009;122(Pt 20):3589-3594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Hoeffer  CA, Klann  E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends Neurosci.  2010;33(2):67-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Ferdous  A, Wang  ZV, Luo  Y, et al.  FoxO1-Dio2 signaling axis governs cardiomyocyte thyroid hormone metabolism and hypertrophic growth. Nat Commun.  2020;11(1):2551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Fontes  KN, Cabanelas  A, Bloise  FF, et al.  Differential regulation of thyroid hormone metabolism target genes during non-thyroidal [corrected] illness syndrome triggered by fasting or sepsis in adult mice. Front Physiol.  2017;8:828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Diano  S, Naftolin  F, Goglia  F, Horvath  TL. Fasting-induced increase in type II iodothyronine deiodinase activity and messenger ribonucleic acid levels is not reversed by thyroxine in the rat hypothalamus. Endocrinology.  1998;139(6):2879-2884. [DOI] [PubMed] [Google Scholar]
  • 116. Kong  WM, Martin  NM, Smith  KL, et al.  Triiodothyronine stimulates food intake via the hypothalamic ventromedial nucleus independent of changes in energy expenditure. Endocrinology.  2004;145(11):5252-5258. [DOI] [PubMed] [Google Scholar]
  • 117. Cubuk  C, Markowsky  H, Herwig  A. Hypothalamic control systems show differential gene expression during spontaneous daily torpor and fasting-induced torpor in the Djungarian hamster (Phodopus sungorus). PLoS One.  2017;12(10):e0186299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Wu  Z, Martinez  ME, St Germain  DL, Hernandez  A. Type 3 deiodinase role on central thyroid hormone action affects the leptin-melanocortin system and circadian activity. Endocrinology.  2017;158(2):419-430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. 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. Physiol Behav.  2010;99(2):237-239. [DOI] [PubMed] [Google Scholar]
  • 120. Aoki  T, Tsunekawa  K, Araki  O, et al.  Type 2 iodothyronine deiodinase activity is required for rapid stimulation of PI3K by thyroxine in human umbilical vein endothelial cells. Endocrinology.  2015;156(11):4312-4324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Kojima  Y, Kondo  Y, Fujishita  T, et al.  Stromal iodothyronine deiodinase 2 (DIO2) promotes the growth of intestinal tumors in ApcΔ716 mutant mice. Cancer Sci.  2019;110(8): 2520-2528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. McAninch  EA, Bianco  AC. Thyroid hormone signaling in energy homeostasis and energy metabolism. Ann N Y Acad Sci.  2014;1311:77-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Bianco  AC. Minireview: cracking the metabolic code for thyroid hormone signaling. Endocrinology.  2011;152(9): 3306-3311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Silva  JE. Thyroid hormone and the energetic cost of keeping body temperature. Biosci Rep.  2005;25(3-4):129-148. [DOI] [PubMed] [Google Scholar]
  • 125. Preite  NZ, Nascimento  BP, Muller  CR, et al.  Disruption of beta3 adrenergic receptor increases susceptibility to DIO in mouse. J Endocrinol.  2016;231(3):259-269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Fernandes  GW, Ueta  CB, Fonseca  TL, et al.  Inactivation of the adrenergic receptor β2 disrupts glucose homeostasis in mice. J Endocrinol.  2014;221(3):381-390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Ueta  CB, Fernandes  GW, Capelo  L, et al.  beta1 Adrenergic receptor is key to cold- and diet-induced thermogenesis in mice. J Endocrinol.  2012;214(3):359-365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Bianco  AC, Silva  JE. Intracellular conversion of thyroxine to triiodothyronine is required for the optimal thermogenic function of brown adipose tissue. J Clin Invest.  1987;79(1):295-300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Carvalho  SD, Kimura  ET, Bianco  AC, Silva  JE. Central role of brown adipose tissue thyroxine 5’-deiodinase on thyroid hormone-dependent thermogenic response to cold. Endocrinology.  1991;128(4):2149-2159. [DOI] [PubMed] [Google Scholar]
  • 130. Branco  M, Ribeiro  M, Negrão  N, Bianco  AC. 3,5,3’-Triiodothyronine actively stimulates UCP in brown fat under minimal sympathetic activity. Am J Physiol.  1999;276(1):E179-E187. [DOI] [PubMed] [Google Scholar]
  • 131. Bianco  AC, Kieffer  JD, Silva  JE. Adenosine 3’,5’-monophosphate and thyroid hormone control of uncoupling protein messenger ribonucleic acid in freshly dispersed brown adipocytes. Endocrinology.  1992;130(5):2625-2633. [DOI] [PubMed] [Google Scholar]
  • 132. Bianco  AC, Carvalho  SD, Carvalho  CR, Rabelo  R, Moriscot  AS. Thyroxine 5’-deiodination mediates norepinephrine-induced lipogenesis in dispersed brown adipocytes. Endocrinology.  1998;139(2):571-578. [DOI] [PubMed] [Google Scholar]
  • 133. Carvalho  SD, Negrão  N, Bianco  AC. Hormonal regulation of malic enzyme and glucose-6-phosphate dehydrogenase in brown adipose tissue. Am J Physiol.  1993;264(6 Pt 1):E874-E881. [DOI] [PubMed] [Google Scholar]
  • 134. Bianco  AC, Silva  JE. Optimal response of key enzymes and uncoupling protein to cold in BAT depends on local T3 generation. Am J Physiol.  1987;253(3 Pt 1):E255-E263. [DOI] [PubMed] [Google Scholar]
  • 135. Katz  LS, Xu  S, Ge  K, Scott  DK, Gershengorn  MC. T3 and glucose coordinately stimulate ChREBP-mediated Ucp1 expression in brown adipocytes from male mice. Endocrinology.  2018;159(1):557-569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Hernández  A, Obregón  MJ. Presence and mRNA expression of T3 receptors in differentiating rat brown adipocytes. Mol Cell Endocrinol.  1996;121(1):37-46. [DOI] [PubMed] [Google Scholar]
  • 137. Minakhina  S, Bansal  S, Zhang  A, et al.  A direct comparison of thyroid hormone receptor protein levels in mice provides unexpected insights into thyroid hormone action. Thyroid.  2020;30(8):1193-1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Ribeiro  MO, Carvalho  SD, Schultz  JJ, et al.  Thyroid hormone–sympathetic interaction and adaptive thermogenesis are thyroid hormone receptor isoform–specific. J Clin Invest.  2001;108(1):97-105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Amorim  BS, Ueta  CB, Freitas  BC, et al.  A TRbeta-selective agonist confers resistance to diet-induced obesity. J Endocrinol.  2009;203(2):291-299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Lin  JZ, Martagón  AJ, Cimini  SL, et al.  Pharmacological activation of thyroid hormone receptors elicits a functional conversion of white to brown fat. Cell Rep.  2015;13(8):1528-1537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Christoffolete  MA, Linardi  CC, de Jesus  L, et al.  Mice with targeted disruption of the Dio2 gene have cold-induced overexpression of the uncoupling protein 1 gene but fail to increase brown adipose tissue lipogenesis and adaptive thermogenesis. Diabetes.  2004;53(3):577-584. [DOI] [PubMed] [Google Scholar]
  • 142. de Jesus  LA, Carvalho  SD, Ribeiro  MO, et al.  The type 2 iodothyronine deiodinase is essential for adaptive thermogenesis in brown adipose tissue. J Clin Invest.  2001;108(9):1379-1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Castillo  M, Hall  JA, Correa-Medina  M, et al.  Disruption of thyroid hormone activation in type 2 deiodinase knockout mice causes obesity with glucose intolerance and liver steatosis only at thermoneutrality. Diabetes.  2011;60(4):1082-1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Christoffolete  MA, Doleschall  M, Egri  P, et al.  Regulation of thyroid hormone activation via the liver X-receptor/retinoid X-receptor pathway. J Endocrinol.  2010;205(2):179-186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Kalaany  NY, Gauthier  KC, Zavacki  AM, et al.  LXRs regulate the balance between fat storage and oxidation. Cell Metab.  2005;1(4):231-244. [DOI] [PubMed] [Google Scholar]
  • 146. Shu  L, Hoo  RL, Wu  X, et al.  A-FABP mediates adaptive thermogenesis by promoting intracellular activation of thyroid hormones in brown adipocytes. Nat Commun.  2017;8:14147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Fukuda  H, Greer  MA. Postnatal development of pituitary-thyroid function in male and female rats: comparison of plasma and thyroid T3 and T4 concentration. J Endocrinol Invest.  1978;1(4):311-314. [DOI] [PubMed] [Google Scholar]
  • 148. Dussault  JH, Labrie  F. Development of the hypothalamic-pituitary-thyroid axis in the neonatal rat. Endocrinology.  1975;97(5):1321-1324. [DOI] [PubMed] [Google Scholar]
  • 149. Fernandes  GW, Bocco  BM, Fonseca  TL, et al.  The FoxO1-inducible transcriptional repressor Zfp125 causes hepatic steatosis and hypercholesterolemia. Cell Rep.  2018;22(2):523-534 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Fonseca  T, Garcia  T, Fernandes  GW, Nair  TM, Bianco  AC. Neonatal thyroxine activation modifies epigenetic programming of the liver. bioRxiv.  2020. doi: 10.1101/2020.12.07.414938 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Fernandes  GW, Bocco  BMLC, Fonseca  T, et al.  The transcriptional repressor Zfp125 modifies hepatic energy metabolism in response to fasting and insulin resistance. bioRxiv.  2020. doi: 10.1101/2020.07.02.185165 [DOI] [Google Scholar]
  • 152. Houbrechts  AM, Van Houcke  J, Darras  VM. Disruption of deiodinase type 2 in zebrafish disturbs male and female reproduction. J Endocrinol.  2019:JOE-18-0549.R3. doi: 10.1530/JOE-18-0549. [DOI] [PubMed] [Google Scholar]
  • 153. Fonseca  TL, Correa-Medina  M, Campos  MP, et al.  Coordination of hypothalamic and pituitary T3 production regulates TSH expression. J Clin Invest.  2013;123(4):1492-1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Dentice  M, Ambrosio  R, Damiano  V, et al.  Intracellular inactivation of thyroid hormone is a survival mechanism for muscle stem cell proliferation and lineage progression. Cell Metab.  2014;20(6):1038-1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Fonseca  TL, Werneck-De-Castro  JP, Castillo  M, et al.  Tissue-specific inactivation of type 2 deiodinase reveals multilevel control of fatty acid oxidation by thyroid hormone in the mouse. Diabetes.  2014;63(5):1594-1604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Salvatore  D, Simonides  WS, Dentice  M, Zavacki  AM, Larsen  PR. Thyroid hormones and skeletal muscle–new insights and potential implications. Nat Rev Endocrinol.  2014;10(4): 206-214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Werneck-de-Castro  JP, Fonseca  TL, Ignacio  DL, et al.  Thyroid hormone signaling in male mouse skeletal muscle is largely independent of D2 in myocytes. Endocrinology.  2015;156(10):3842-3852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Ambrosio  R, Damiano  V, Sibilio  A, et al.  Epigenetic control of type 2 and 3 deiodinases in myogenesis: role of Lysine-specific Demethylase enzyme and FoxO3. Nucleic Acids Res.  2013;41(6):3551-3562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Marsili  A, Ramadan  W, Harney  JW, et al.  Type 2 iodothyronine deiodinase levels are higher in slow-twitch than fast-twitch mouse skeletal muscle and are increased in hypothyroidism. Endocrinology.  2010;151(12):5952-5960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Ramadan  W, Marsili  A, Huang  S, Larsen  PR, Silva  JE. Type-2 iodothyronine 5’deiodinase in skeletal muscle of C57BL/6 mice. I. Identity, subcellular localization, and characterization. Endocrinology.  2011;152(8):3082-3092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Ramadan  W, Marsili  A, Larsen  PR, Zavacki  AM, Silva  JE. Type-2 iodothyronine 5’deiodinase (D2) in skeletal muscle of C57Bl/6 mice. II. Evidence for a role of D2 in the hypermetabolism of thyroid hormone receptor alpha-deficient mice. Endocrinology.  2011;152(8):3093-3102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Otto  A, Collins-Hooper  H, Patel  K. The origin, molecular regulation and therapeutic potential of myogenic stem cell populations. J Anat.  2009;215(5):477-497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Mauro  A. Satellite cell of skeletal muscle fibers. J Biophys Biochem Cytol.  1961;9:493-495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Dentice  M, Marsili  A, Ambrosio  R, et al.  The FoxO3/type 2 deiodinase pathway is required for normal mouse myogenesis and muscle regeneration. J Clin Invest.  2010;120(11):4021-4030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Almind  K, Manieri  M, Sivitz  WI, Cinti  S, Kahn  CR. Ectopic brown adipose tissue in muscle provides a mechanism for differences in risk of metabolic syndrome in mice. Proc Natl Acad Sci U S A.  2007;104(7):2366-2371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Yang  Y, Zhang  J, Wu  G, et al.  Dietary methionine restriction regulated energy and protein homeostasis by improving thyroid function in high fat diet mice. Food Funct.  2018;9(7):3718-3731. [DOI] [PubMed] [Google Scholar]
  • 167. Sagliocchi  S, Cicatiello  AG, Di Cicco  E, et al.  The thyroid hormone activating enzyme, type 2 deiodinase, induces myogenic differentiation by regulating mitochondrial metabolism and reducing oxidative stress. Redox Biol.  2019;24:101228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Hall  JA, Ribich  S, Christoffolete  MA, et al.  Absence of thyroid hormone activation during development underlies a permanent defect in adaptive thermogenesis. Endocrinology.  2010;151(9):4573-4582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Akiyama  S, Ogiwara  T, Aoki  T, Tsunekawa  K, Araki  O, Murakami  M. Glucagon-like peptide-1 stimulates type 3 iodothyronine deiodinase expression in a mouse insulinoma cell line. Life Sci.  2014;115(1-2):22-28. [DOI] [PubMed] [Google Scholar]
  • 170. 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. [DOI] [PubMed] [Google Scholar]
  • 171. da-Silva  WS, Harney  JW, Kim  BW, et al.  The small polyphenolic molecule kaempferol increases cellular energy expenditure and thyroid hormone activation. Diabetes.  2007;56(3):767-776. [DOI] [PubMed] [Google Scholar]
  • 172. da-Silva  WS, Ribich  S, Arrojo e Drigo  R, Castillo  M, Patti  ME, Bianco  AC. The chemical chaperones tauroursodeoxycholic and 4-phenylbutyric acid accelerate thyroid hormone activation and energy expenditure. FEBS Lett.  2011;585(3):539-544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Thomas  C, Gioiello  A, Noriega  L, et al.  TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab.  2009;10(3):167-177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Broeders  EP, Nascimento  EB, Havekes  B, et al.  The bile acid chenodeoxycholic acid increases human brown adipose tissue activity. Cell Metab.  2015;22(3):418-426. [DOI] [PubMed] [Google Scholar]
  • 175. Ockenga  J, Valentini  L, Schuetz  T, et al.  Plasma bile acids are associated with energy expenditure and thyroid function in humans. J Clin Endocrinol Metab.  2012;97(2):535-542. [DOI] [PubMed] [Google Scholar]
  • 176. Arrojo e Drigo  R, Fonseca  TL, Castillo  M, et al.  Endoplasmic reticulum stress decreases intracellular thyroid hormone activation via an eIF2a-mediated decrease in type 2 deiodinase synthesis. Mol Endocrinol.  2011;25(12):2065-2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Ozcan  U, Cao  Q, Yilmaz  E, et al.  Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science.  2004;306(5695):457-461. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.


Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES