Defending plasma T3 is a biological priority (2024)

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Defending plasma T3 is a biological priority (1)

About Author manuscriptsSubmit a manuscriptHHS Public Access; Author Manuscript; Accepted for publication in peer reviewed journal;

Clin Endocrinol (Oxf). Author manuscript; available in PMC 2016 Jan 4.

Published in final edited form as:

PMCID: PMC4699302

NIHMSID: NIHMS746217

PMID: 25040645

Author information Copyright and License information PMC Disclaimer

The publisher's final edited version of this article is available at Clin Endocrinol (Oxf)

Summary

Triiodothyronine (T3), the active form of thyroid hormone is produced predominantly outside the thyroid parenchyma secondary to peripheral tissue deiodination of thyroxine (T4), with <20% being secreted directly from the thyroid. In healthy individuals, plasma T3 is regulated by the negative feedback loop of the hypothalamus–pituitary–thyroid axis and by hom*oeostatic changes in deiodinase expression. Therefore, with the exception of a minimal circadian rhythmicity, serum T3 levels are stable over long periods of time. Studies in rodents indicate that different levels of genetic disruption of the feedback mechanism and deiodinase system are met with increase in serum T4 and thyroid-stimulating hormone (TSH) levels, while serum T3 levels remain stable. These findings have focused attention on serum T3 levels in patients with thyroid disease, with important clinical implications affecting therapeutic goals and choice of therapy for patients with hypothyroidism. Although monotherapy with levothyroxine is the standard of care for hypothyroidism, not all patients normalize serum T3 levels with many advocating for combination therapy with levothyroxine and liothyronine. The latter could be relevant for a significant number of patients that remain symptomatic on monotherapy with levothyroxine, despite normalization of serum TSH levels.

Introduction

The thyroid gland takes up iodide and produces iodinated molecules that have pleiotropic effects in vertebrates.1 The two main iodinated molecules secreted by the thyroid gland are thyroxine (T4) and triiodothyronine (T3). Even though both molecules can trigger biological effects, T3 is considered the biologically active thyroid hormone that binds to thyroid hormone receptors (TR), while T4 is a prohormone that must be converted to T3 in order to initiate signalling and gain biological activity. A corollary is that the level of T3 inside the cells defines how much T3 is bound to TR and hence the intensity of signalling and T3-dependent biological effects, aka ‘thyroid status’. In an organism, thyroid status can be considered the sum of all T3-dependent signalling events and depends on (i) circulating T3 levels and (ii) tissue/cell-specific factors influencing the intracellular concentration of T3. An organism is known to exhibit thyrotoxicosis when the intracellular levels of T3 are increased, whereas hypothyroidism results from thyroid hormone deficiency. In addition, individual tissues could be said to have specific thyroid status, that is, thyrotoxic or hypothyroid, relatively independent of serum thyroid hormone levels; this is because of tissue/cell-specific factors such as deiodinase activities and/or transport mechanisms.2 In this regard, the type 2 deiodinase (D2) catalyses T4 to T3 conversion and increases intracellular levels of T3 potentially leading to local thyrotoxicosis. The opposite is observed in cells expressing the type 3 deiodinase (D3), which depletes the cell of T3 by deiodination to T2 and can cause local hypothyroidism. Understanding these basic mechanisms that regulate thyroid hormone metabolism and action has clinical implications and might affect the choice of therapy for hypothyroid patients, a highly controversial area in the thyroid field.

What is the biologically active thyroid hormone?

Thyroid hormone signalling is initiated by binding of T3 to one of its TR isoforms, TRα or TRβ, thus affecting the expression of thyroid hormone-dependent genes.1 This is also known as the genomic effect of thyroid hormone and explains the biological actions of thyroid hormone in the various organs/systems, that is, development, growth, metabolic control and cognition. The fact that the plasma concentration (and presumably intracellular concentration) of free T4 and free T3 are similar (5–10 pM) and that the TR affinity for T3 is approximately 10-fold greater than T4, makes T3 the most potent TH. In fact, acknowledging that T4 is a prohormone indicates that conversion to the more active T3 molecule is required for biological activity. Notably, at higher concentrations, T4 can also bind to TR, modify gene transcription and trigger biological effects; however, this is considered minimal at physiological levels.1 In addition, T4 also exhibits significant nongenomic effects such as acceleration of the type 2 iodothyronine deiodinase (D2) inactivation via ubiquitination.3

Measuring plasma T3 levels

Similarly to T4, most T3 in human serum is bound to carrier proteins, namely thyroid-binding globulin (TBG), transthyretin and albumin, while <0·4% of T3 is free.4 Given its important biological activity, knowing the serum total T3 (TT3) levels is important to understand systemic thyroid hormone status. Ideally, one would prefer assessing circulating free T3 (FT3) levels as they provide information about the level of T3 that is biologically available to enter cells and initiate thyroid hormone action. Of course, this is not the only factor defining thyroid hormone action, also defined by cellular transporters that facilitate passage across the cell membrane, deiodinases that can activate or inactivate thyroid hormone as well as TRs and its co-regulators.5

Measuring serum FT3 levels is labour intensive and requires equipment not readily available in most clinical reference laboratories. Typically, the first step to measure serum FT3 is to physically separate the free T3 molecules from the T3 molecules that are bound to serum proteins, without disturbing the endogenous equilibrium between fractions. This is achievable by equilibrium dialysis or ultrafiltration; the second step is to measure T3 in the dialysate or ultrafiltrate, respectively. Ultrafiltration is far less labour intensive and therefore is the method used in many reference laboratories.68 Following equilibrium dialysis and ultrafiltration, a variety of detection techniques are available to measure free hormone concentration, including immunoassays and tandem mass spectrometry (MS/MS).

The majority of all commercially available assays utilize an alternative approach to analyse FT3 concentrations directly in the presence of proteins (unfractionated samples of serum). This is referred to as the direct FT3 method that unfortunately is subject to interferences and lack of specificity, exhibits inconsistent analytical performance and has poor correlation with equilibrium dialysis/ultrafiltration LC/MS/MS methods, currently regarded as gold standards.7 The main factors known to interfere with the direct FT3 methods originate in the sample and include abnormal binding proteins, heterophilic antibodies, autoantibodies and possibly free fatty acids.7

Thus, in the absence of equilibrium dialysis/ultrafiltration LC/MS/MS methods, it is not clear that using a direct FT3 assay offers any clinical advantage over measuring serum TT3 levels. In this case, indirect information about free T3 levels can be derived from the resin T3 uptake, which quantifies the empty T3 binding sites available in the serum carrier proteins.9

Where is T3 produced? How much T3 is produced? Via which pathway?

Most T3 is produced outside the thyroid gland via deiodination of T4, with <20% being secreted directly from the thyroid.10 Thyroidal T3 derives from intracellular thyroglobulin digestion that is supposedly enriched via intrathyroidal deiodination of T4 to T3, although it is uncertain how much thyrocyte deiodination contributes to the daily thyroidal T3 production. The molar ratio of T4 to T3 in the human thyroglobulin is 15:1, and some estimates put the thyroidal secretion as containing a molar ratio of 11:1, which is supportive of thyroidal deiodination of T4.1113 Furthermore, this ratio might decrease even further in situations such as iodine deficiency or thyroid disease through mechanisms that include changes in thyroglobulin iodination and possibly intrathyroidal deiodination.11,12

It is estimated that healthy adult subjects produce about 30 µg T3/day, of which about 5 µg are secreted directly from the thyroid and the rest is produced outside of the thyroid parenchyma via T4 deiodination.10 Deiodination is catalysed by a group of enzymes known as iodothyronine deiodinases that mediate (i) activation of T4 into T3 or (ii) inactivation of T4 into rT3 and of T3 into 3,3′-T2. All deiodinases are dimeric integral membrane thioredoxin-fold like containing selenoenzymes that are expressed in multiple tissues and cells, including the thyroid gland.2,14,15 There are two deiodinase pathways that lead to extrathyroidal T3 production, the type 1 deiodinase (D1) and D2 pathways. In turn, the third deiodinase pathway (type 3 deiodinase, aka D3) is involved in catabolism of both T4 and T3, terminating thyroid hormone action.

In a healthy adult individual, D2 is thought to mediate the bulk of T3 production, approximately 20 µg/day, with a minor contribution provided by D1 (5 µg/day).10 Relatively high D2-specific activity can be found in the brain, pituitary gland, brown adipose tissue (BAT) and heart.10,16 D2 is present in many other tissues at lower specific activity, including skin, skeletal muscle, skeleton, vascular smooth muscle and testis.1721 In contrast, D1 expression is restricted to the liver and kidney where it also processes conjugated T3, clearing these metabolites from the circulation.22 For example, T3 is a poor D1 substrate but once it is sulphated in its phenolic ring, T3S gains water solubility and is rapidly metabolized via D1, conceivably to conserve iodide before the molecule is eliminated in the bile.22,23 T3S has no biological activity but sulfatases present in tissues and the intestinal microflora can convert T3S back to T3. It is unclear at the moment how much these pathways play a role in the daily T3 economy in humans.

Given the widespread D2 expression throughout the body, it is likely that multiple organs/tissues collectively contribute to the daily T3 production via the D2 pathway. Brain also expresses relatively high levels of D3, the inactivating deiodinase, minimizing its potential contribution as an organ to plasma T3. Brown adipose tissue, on the other hand, has no D3 activity, and thus it is potentially an important source of circulating T3. This has previously been shown in rodents,24 and the more recent finding of D2-containing brown adipose tissue in humans25 also highlights its relevance as a source of T3. Less is known about the role played by tissues that express D2 at low specific activity such as skeletal muscle, vascular smooth muscle or skin, but, given the size of these organs/tissues, their contribution is probably relevant as well.

What are the cellular bases of deiodinase-mediated T3 production/catabolism?

T3 production via deiodination is an intracellular event. T4 enters cells via specific transmembrane transporters and can then be converted to T3.26 D1 is located in the plasma membrane and thus T3 produced via the D1 pathway exits the cells promptly and rapidly equilibrates with the plasma pool of T3.27,28 In fact, kinetics studies indicate that T3 molecules generated via the D1 pathway stay inside the cell for just about 30 min before exiting to plasma. In contrast, D2 is an endoplasmic reticulum (ER)-resident protein, and thus, T4 to T3 conversion through this pathway occurs in a cellular compartment that is physically associated with the cell nucleus, where the TRs are located.27,29 Thus, D2-originated T3 also diffuses to the nucleus and perhaps for this reason remains in the target cell for a longer period time, approximately 8 h.28,30 The significance of these observations is that D2-generated T3 also interacts with the local TRs and triggers biological effects in the same cell where it was produced. Eventually, D2-generated T3 exits the cell and equilibrates with the circulating pool of T3.

D3 is located in the plasma membrane and thus has easy access to incoming thyroid hormone molecules.31,32 Furthermore, under hypoxic or ischaemic conditions, D3 is preferentially inserted into the nuclear membrane where it inactivates thyroid hormone, thereby decreasing thyroid hormone signalling.33 T3 is inactivated by D3 via conversion to T2, a pathway that is present in the brain, skin and placenta.34,35 The latter explains in part the need for increased dose of thyroid hormone replacement to hypothyroid patients during pregnancy.36 Furthermore, D3 expression can be enhanced in multiple other tissues in disease states, including the liver, lungs, heart and brain, particularly during ischaemic or hypoxic states. In rare cases, D3 is greatly expressed in infantile hemangiomas to such a degree that inactivates T3 at a faster rate than it can be produced.37 Similarly, the use of the tyrosine kinase inhibitors imatinib and sunitinib in patients with cancer is also associated with hypothyroidism, which seems to be the result of marked D3 overexpression within the tumours.38 In fact, the use of these drugs commonly results in hypothyroidism when used for the treatment of a range of other tumours including differentiated thyroid cancer. The latter routinely requires an increase in their levothyroxine suppressive therapy when treated with tyrosine kinase inhibitors. The cause of hypothyroidism in these situations is not fully understood, and it could be multifactorial, including enhanced D3 expression in other tissues. All of these conditions characterize the syndrome of consumptive hypothyroidism, a state of systemic hypothyroidism caused by excessive degradation of T3 and T4 via abnormally high expression of D3.

An analogous situation is observed in patients treated with anti-epileptic drugs such as diphenylhydantonin or carbamazepine. There is accelerated biliary elimination and reduction in serum thyroid hormone levels due to enhanced conjugation to glucuronic acid.39 Notably, such patients do not exhibit an increase in serum thyroid-stimulating hormone (TSH) but warrant close monitoring as they may need treatment and/or adjustment of thyroid hormone replacement dose.

The corollary of these studies is that D1- and D2-expressing tissues produce a ‘positive’ flow of T3 that exits the cells, entering the circulation. In contrast, D3-expressing tissues function as a sink for TH, whereby there is a ‘negative’ flow of both T4 and T3 that enter the cells not to return to the circulation.2,40

Plasma T3 levels are relatively stable over time

Serum TT3 and FT3 exhibit minimal circadian rhythmicity that is due to a nocturnal increase in TSH secretion.41,42 Otherwise, serum T3 is remarkably stable over periods of days, weeks or months in healthy adult individuals, despite a relatively short half-life (approximately 12–18 h). This is likely the result of combined hom*oeostatic mechanisms involving (i) the hypothalamus–pituitary–thyroid axis as well as (ii) the group of deiodinases (Fig. 1).

Defending plasma T3 is a biological priority (2)

Schematic representation of the feedback mechanisms in the hypothalamus–pituitary–thyroid axis. The thyroid gland secretes T4 and T3, the biologically active thyroid hormone; T3 is also produced outside the thyroid gland via deiodination of T4; T3 initiates the negative feedback at the hypothalamus and anterior pituitary gland by suppressing TRH and TSH expression and secretion, respectively; only about 20% of the T3 produced daily is directly secreted by the thyroid gland; most T3 (about 80%) is produced outside of the thyroid gland (periphery) via two deiodination pathways, D1 and D2; T3 from both sources mixes in the plasma, defining the pool of circulating T3; D2-generated T3 also acts locally, before exiting the cell where deiodination of T4 took place; this occurs in the hypothalamus and pituitary gland and explains how circulating T4 can signal the feedback mechanism; in the hypothalamus, D2 is expressed in specialized ependymal cells (tanycytes), whereas in the anterior pituitary gland, D2 is expressed in the TSH-producing cells, thyrotrophs; the current model for the feedback mechanism is depicted in (a) and predicts that plasma T3 mixes with D2-generated T3 both in the hypothalamus (to suppress TRH) and in the pituitary gland (to suppress TSH); the data obtained in the genetically modified mice support a new model (depicted in (b)) in which plasma and D2-generated T3 do not mix in the hypothalamus; consequently, plasma T3 can be monitored independently of circulating T4.

It is well accepted that TSH secretion is negatively regulated by T4 and T3, although the hypothalamus–pituitary axis seems to be particularly sensitive to circulating T4 levels. In fact, few examples exist in which serum T3 is low in the face of normal serum T4 and TSH levels. In most such examples, there is nonthyroidal illness that results in central hypothyroidism and inappropriately low/normal serum TSH levels. A unique experimental setup illustrating that serum T3 per se is an important determinant of TSH secretion is the acute administration of large doses of PTU to thyroidectomized individuals kept on levothyroxine replacement therapy.43 The approximately 20% drop in serum T3 as a result of D1 inhibition was sufficient to double serum TSH even as serum T4 levels remained stable.43 It is unknown whether this TSH response is originated at the hypothalamus, pituitary or both levels.

The deiodinase group has the potential to defend serum T3 levels given the inverse relationship between D2 and D3 observed during hypo- or hyperthyroidism.2,35,44 Whereas Dio2 is negatively regulated by T3, the opposite is observed for Dio3. As a result, during hypothyroidism, there is increase in the fractional conversion of T4 to T3 that reflects the higher D2 activity and decreased clearance of T3 that reflects lower D3 activity, both presumably helping maintain serum T3 levels within the normal range.4547

Perhaps the most dramatic deiodinase-mediated hom*oeostatic mechanism is the post-translational loss of D2 activity induced by interaction with T4 via conjugation to ubiquitin.3,48 D2 is a type I endoplasmic reticulum (ER)-resident D2 with a variable short half-life that depends on whether its natural substrate T4 is available. In the presence of T4, D2 is inactivated with an approximately 20 min half-life, whereas in the absence of T4, its half-life is prolonged to hours.49 This provides a mechanism through which the production of T3 can be regulated according to the availability of T4.

D2 ubiquitination is the molecular mechanism underlying these changes in D2 half-life, that is, the covalent attachment of multiple ubiquitin molecules to D2, which both inactivates the enzyme and targets it to degradation in the proteasomes.48 Ubiquitination is thought to inactivate D2 by disrupting the conformation of the D2:D2 dimer, critical for enzyme activity.3,50 A unique 18-amino acid loop confers intrinsic metabolic instability to D2, facilitating binding to proteins involved in the ubiquitination process.51,52

The ubiquitin-activating enzymes UBC6 and UBC7 are critical in the process of D2 ubiquitination,53,54 as well as two ubiquitin ligases, the hedgehog-inducible WSB-1, and TEB4, a ligase involved in the degradation of proteins in the endoplasmic reticulum.52,55 D2 is structured as an hom*odimer, D2:D2, and monomers are inactive.15 Ubiquitinated D2 (UbD2) is not immediately taken up by the proteasomes. Instead, UbD2 can be reactivated by deubiquitination, a process catalysed by two USP-class D2-interacting de-ubiquitinases (DUBs), USP-20 and USP-33.56 The other two deiodinases, D1 and D3, are not known to be ubiquitinated or undergo post-translational modifications.

D2 ubiquitination occurs via K48-linked ubiquitin chains, and exposure to its natural substrate, T4, accelerates UbD2 formation.57 UbD2 is taken up by proteasomes located in the cytoplasm. D2 retrotranslocation to the cytoplasm occurs via interaction with the p97–ATPase complex. D2 retrotranslocation also includes deubiquitination by the p97-associated DUB Ataxin-3. Once in the cytosol, D2 is delivered to the proteasomes as evidenced by coprecipitation with 19S proteasome subunit S5a and increased co-localization with the 20S proteasome.57

Therefore, a number of transcriptional and post-transcriptional mechanisms underlie the hom*oeostatic role played by the deiodinase system in defending serum T3 levels. In combination with the hypothalamus–pituitary–thyroid axis, both central and peripheral pathways explain the stability of serum T3 levels.

The hypothalamus–pituitary–thyroid axis is wired to preserve serum T3

Based on the above discussion and on the fact that the deiodinases contribute with approximately 60% of circulating T3 in rodents (and about 80% in humans), one would expect major interference in serum T3 hom*oeostasis upon disruption of the deiodinase system. However, serum T3 levels are normal in mice with single or combined targeted inactivation of both Dio1 and Dio2 genes.5861 In these animal models, there is resetting in the hypothalamus–pituitary–thyroid axis that includes increase in serum T4 and/or serum TSH, leading to increased thyroidal T3 secretion that makes up for the lack of extrathyroidal T3 production. Even if the disruption in Dio2 is restricted to the TSH-producing cells, serum TSH and T4 are increased while preserving serum T3 levels.62

A response mediated by the hypothalamus–pituitary–thyroid axis also seems to be involved in the maintenance of serum T3 levels during iodine deficiency or mild hypothyroidism.63,64 In both cases, there is an increase in serum TSH levels due to the drop in serum T4 while serum T3 remains within normal range.

That the level of serum T3 is a main target around which serum T4 and TSH are adjusted constitutes a shift in the paradigm traditionally accepted for the function of the hypothalamus–pituitary–thyroid axis. For example, it is unexpected that the hypothalamus–pituitary–thyroid axis would tolerate an increased serum T4 in order to preserve serum T3.6062 This observation challenges the dogma that locally produced T3 via D2 mixes at a prereceptor level with incoming plasma T3 in the hypothalamus and pituitary so that both, indistinctively, reduce expression of TRH and TSH. The new evidence obtained in the genetically modified animals suggests the existence of a distinct unknown mechanism within the hypothalamus–pituitary axis that selectively detects plasma T3.

Of course there are a number of physiological conditions in which the hypothalamus–pituitary–thyroid axis does adjust in response to environment cues such as food availability, temperature, breeding season or pregnancy, all of which reflect in adaptive changes in serum T3. In addition, there are a number of pathophysiological conditions in which the hypothalamus–pituitary–thyroid axis does not defend serum T3, which are let drop below the reference range. These are largely centrally mediated and include fasting/caloric restriction, nonthyroidal illness, hypothalamic hyperthyroidism due to Dio3 inactivation, or targeted inactivation of TRH gene.65,66 In fact, the finding of low serum T3 in the presence of normal serum T4 and TSH levels is a prevalent event as nonthyroidal illness is common. In addition, certain species exhibit significant seasonal variation in thyroid hormone levels that is mediated through adjustments in the hypothalamus–pituitary–thyroid axis.

Thus, experimental data support the concept that in healthy adult mice the hypothalamus–pituitary–thyroid axis is wired to preserve serum T3,6062 except when it deliberately ‘allows’ serum T3 to drop in response to physiological or pathophysiological mechanisms, or its function is disrupted to the point that is no longer capable of reacting adequately to a fall in serum T3.65,66

How well defended is serum T3 in the absence of a functional thyroid gland?

Experimental evidence indicates that in the absence of T3-producing deiodinases, the hypothalamus–pituitary–thyroid axis resets and is capable of defending serum T3. Would the deiodinases be able to do the same in the absence of a functional thyroid gland? Studies in thyroidectomized rats indicate that monotherapy with levothyroxine alone does not normalize serum and tissue T3 and T4 simultaneously at normal TSH levels.67 Only the combination of levothyroxine and liothyronine therapy was able to normalize serum TSH as well as serum and tissue T4 and T3 concentrations simultaneously in all tissues.68

In humans, multiple studies have looked at serum T3 levels in clinically euthyroid athyreotic or hypothyroid patients maintained on levothyroxine replacement therapy. The consensus has been that such patients maintain levels of T3 that are slightly but significantly lower and of T4 that are slightly higher than control individuals.69 In addition, normal serum T3 levels can be achieved but at the expense of having relatively lower/suppressed serum TSH.70,71 A large cross-sectional study involving approximately 1800 athyreotic patients clinically euthyroid on levothyroxine monotherapy revealed that the distribution of serum FT3 levels is shifted to the left and that of FT4 levels is shifted to the right when compared to the distribution patterns obtained in approximately 3900 controls.72 In this study, for every quintile of serum TSH distributed within the normal range, athyreotic patients on levothyroxine monotherapy exhibited significantly lower serum FT3 and higher serum FT4 when compared to the controls on the same serum TSH quintile.

Notably, a study of 50 patients undergoing elective total thyroidectomy indicated that monotherapy with levothyroxine alone was able to bring serum T3 levels back to the same presurgical levels without suppressing serum TSH, although serum FT4 levels were significantly higher after surgery.9

Is having a relatively high serum T4 and low serum T3 clinically relevant?

Whereas it has become widely accepted that serum T3 is relatively lower in hypothyroid patients maintained on levothyroxine alone, it is unclear whether this is clinically relevant. In addition, it is equally unclear whether those patients that remain symptomatic despite having normal serum TSH levels are the same patients that cannot maintain serum T3 levels within the normal range. Clinical trials are needed to answer these questions, particularly trials with outcomes including objective biological responses to T3. For example, an acute drop in serum T3 of approximately 20% in thyroidectomized individuals maintained on levothyroxine replacement therapy resulted in doubling of serum TSH,43 indicating that at least from the thyrotroph’s perspective, a small reduction in serum T3 is clinically relevant. The long-term systemic consequences of a relatively low serum T3 are unknown.

Is there evidence-based support for treatment of hypothyroid patients with combination therapy?

The renewed attention to serum T3 and the possibility that its levels might not be fully restored in hypothyroid patients after conventional treatment has led to clinical trials analysing patient satisfaction and quality of life questionnaires in hypothyroid patients kept on monotherapy vs combination therapy with levothyroxine and liothyronine. Readers are also referred to recent comprehensive reviews on this highly controversial area.7375

Some studies suggest that patients tend to prefer combination therapy based on patient preference/satisfaction,76,77 body weight,76 quality of life, depression, neuropsychological function, mood and anxiety rating scales;77,78 the majority of the studies indicate that both forms of treatment provide similar clinical outcomes.7981 However, a meta-analysis of eleven randomized controlled trials and a total of 1216 participants comparing combination therapy vs monotherapy revealed no difference in the effectiveness of combination therapy vs monotherapy with regard to bodily pain, depression, fatigue, body weight, total serum cholesterol, triglyceride levels, low-density lipoprotein, high density lipoprotein or adverse events.82 In addition, a systematic review of ten randomized controlled trials comparing combination therapy vs monotherapy found no statistically significant differences in biochemical markers, mood states, adverse effects or drop-out rates, suggesting that combination therapy does not improve well-being, cognitive function, or quality of life as compared to monotherapy.83

There is new evidence that patients bearing the Thr92Ala D2 polymorphism might benefit from combination therapy.84 This genetic polymorphism causes an amino acid change in a critical loop of the D2 molecule that controls its half-life.52 It is not known how this polymorphism affects D2 function but it could hypothetically disrupt thyroid hormone signalling in D2-expressing tissues, making these cells more susceptible to a reduction in serum T3.

In view of the experimental evidence that (i) serum T3 levels are stable in healthy adult individuals, and (ii) preserving a stable plasma T3 is the default condition after disruption of the hypothalamus–pituitary–thyroid axis, the failure of combination therapy to provide superior clear cut clinical outcome might reside on the fact that current forms of liothyronine replacement, that is, tablets, do not ensure stable serum T3 levels throughout the day. Given the relatively short T3 half-life and the relatively fast absorption of liothyronine, patients receiving a tablet of liothyronine experience a transient increase in serum T3 that subsides during the next few hours.85 In fact, it has been reported that at least three tablets daily of liothyronine are necessary to avoid peaks of serum T3 that are above the normal range, which makes this approach impractical.86 Most importantly, these observations put in check the conclusions obtained in clinical trials that used a single dose of liothyronine when comparing monotherapy vs combination therapy.

New randomized double-blind placebo controlled trials should be performed once new technology allows for stable deliver of liothyronine, for example slow release formulations. For example, oral administration of 3,5,3′-triiodothyronine sulphate (T3S) that banks on the lack of biological activity of T3S and the endogenous desulphating pathway that slowly produces T3 from T3S. In hypothyroid humans, T3S is absorbed following oral administration and results in steady state serum T3 levels for 48 h.87

Conclusion

Disruption of key elements in the hypothalamus–pituitary–thyroid axis as well as the deiodinase system in animals suggests that maintaining a stable serum T3 within normal range is a biological priority. At the same time, an analysis of a large number of hypothyroid patients maintained on levothyroxine replacement therapy indicates that monotherapy restores serum TSH levels without normalizing serum T3 in a portion of patients. The clinical relevance of a relatively lower serum T3 is unknown. Many clinical trials comparing monotherapy vs combination therapy might not have been useful, given limitations to normalize serum T3 with a single tablet of liothyronine daily.

Acknowledgments

The authors are grateful to Dr. Elizabeth McAninch for critically reviewing the manuscript. Part of the studies discussed here was sponsored by the NIDDK.

References

1. Brent GA. Mechanisms of thyroid hormone action. The Journal of Clinical Investigation. 2012;122:3035–3043. [PMC free article] [PubMed] [Google Scholar]

2. Gereben B, Zavacki AM, Ribich S, et al. Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling. Endocrine Reviews. 2008;29:898–938. [PMC free article] [PubMed] [Google Scholar]

3. Sagar GD, Gereben B, Callebaut I, et al. Ubiquitination-induced conformational change within the deiodinase dimer is a switch regulating enzyme activity. Molecular and Cellular Biology. 2007;27:4774–4783. [PMC free article] [PubMed] [Google Scholar]

4. Schussler GC. The thyroxine-binding proteins. Thyroid. 2000;10:141–149. [PubMed] [Google Scholar]

5. Jansen J, Friesema EC, Milici C, et al. Thyroid hormone transporters in health and disease. Thyroid. 2005;15:757–768. [PubMed] [Google Scholar]

6. Jonklaas J, Kahric-Janicic N, Soldin OP, et al. Correlations of free thyroid hormones measured by tandem mass spectrometry and immunoassay with thyroid-stimulating hormone across 4 patient populations. Clinical Chemistry. 2009;55:1380–1388. [PMC free article] [PubMed] [Google Scholar]

7. Soldin OP, Soldin SJ. Thyroid hormone testing by tandem mass spectrometry. Clinical Biochemistry. 2011;44:89–94. [PMC free article] [PubMed] [Google Scholar]

8. Konno N, Hagiwara K, Taguchi H, et al. Measurements of serum-free thyroid hormone concentrations by ultrafiltration–a comparison with equilibrium dialysis & mathematical calculation. Annals of Nuclear Medicine. 1987;1:15–22. [PubMed] [Google Scholar]

9. Jonklaas J, Davidson B, Bhagat S, et al. Triiodothyronine levels in athyreotic individuals during levothyroxine therapy. JAMA. 2008;299:769–777. [PubMed] [Google Scholar]

10. Bianco AC, Salvatore D, Gereben B, et al. Biochemistry, cellular and molecular biology, and physiological roles of the iodothyronine selenodeiodinases. Endocrine Reviews. 2002;23:38–89. [PubMed] [Google Scholar]

11. Laurberg P. Mechanisms governing the relative proportions of thyroxine and 3,5,3′-triiodothyronine in thyroid secretion. Metabolism. 1984;33:379–392. [PubMed] [Google Scholar]

12. Laurberg P. Thyroxine and 3,5,3′-triiodothyronine content of thyroglobulin in thyroid needle aspirates in hyperthyroidism and hypothyroidism. The Journal of Clinical Endocrinology and Metabolism. 1987;64:969–974. [PubMed] [Google Scholar]

13. Pilo A, Iervasi G, Vitek F, et al. Thyroidal and peripheral production of 3,5,3’-triiodothyronine in humans by multi-compartmental analysis. The American Journal of Physiology. 1990;258:E715–E726. [PubMed] [Google Scholar]

14. 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. The Journal of Biological Chemistry. 2003;278:36887–36896. [PubMed] [Google Scholar]

15. Curcio-Morelli C, Gereben B, Zavacki AM, et al. In vivo dimerization of types 1, 2, and 3 iodothyronine selenodeiodinases. Endocrinology. 2003;144:937–946. [PubMed] [Google Scholar]

16. St Germain DL, Hernandez A, Schneider MJ, et al. Insights into the role of deiodinases from studies of genetically modified animals. Thyroid. 2005;15:905–916. [PubMed] [Google Scholar]

17. Hosoi Y, Murakami M, Mizuma H, et al. Expression and regulation of type II iodothyronine deiodinase in cultured human skeletal muscle cells. The Journal of Clinical Endocrinology and Metabolism. 1999;84:3293–3300. [PubMed] [Google Scholar]

18. Mizuma H, Murakami M, Mori M. Thyroid hormone activation in human vascular smooth muscle cells: expression of type II iodothyronine deiodinase. Circulation Research. 2001;88:313–318. [PubMed] [Google Scholar]

19. Wajner SM, dos Santos Wagner M, Melo RC, et al. Type 2 iodothyronine deiodinase is highly expressed in germ cells of adult rat testis. The Journal of Endocrinology. 2007;194:47–54. [PubMed] [Google Scholar]

20. Williams AJ, Robson H, Kester MH, et al. Iodothyronine deiodinase enzyme activities in bone. Bone. 2008;43:126–134. [PMC free article] [PubMed] [Google Scholar]

21. Gouveia CH, Christoffolete MA, Zaitune CR, et al. Type 2 iodothyronine selenodeiodinase is expressed throughout the mouse skeleton and in the MC3T3-E1 mouse osteoblastic cell line during differentiation. Endocrinology. 2005;146:195–200. [PubMed] [Google Scholar]

22. 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;147:580–589. [PubMed] [Google Scholar]

23. Visser TJ. Role of sulfation in thyroid hormone metabolism. Chemico-Biological Interactions. 1994;92:293–303. [PubMed] [Google Scholar]

24. Silva JE, Larsen PR. The potential of brown adipose tissue type II T4 5’-deiodinase as a local and systemic source of T3 in rats. The Journal of Clinical Investigation. 1985;76:2296–2305. [PMC free article] [PubMed] [Google Scholar]

25. Skarulis MC, Celi FS, Mueller E, et al. Thyroid hormone induced brown adipose tissue and amelioration of diabetes in a patient with extreme insulin resistance. The Journal of Clinical Endocrinology and Metabolism. 2010;95:256–262. [PMC free article] [PubMed] [Google Scholar]

26. Visser TJ. Thyroid hormone transporters. Hormone Research. 2007;68(Suppl 5):28–30. [PubMed] [Google Scholar]

27. Baqui MM, Gereben B, Harney JW, et al. Distinct subcellular localization of transiently expressed types 1 and 2 iodothyronine deiodinases as determined by immunofluorescence confocal microscopy. Endocrinology. 2000;141:4309–4312. [PubMed] [Google Scholar]

28. Silva JE, Dick TE, Larsen PR. The contribution of local tissue thyroxine monodeiodination to the nuclear 3,5,3’-triiodothyronine in pituitary, liver, and kidney of euthyroid rats. Endocrinology. 1978;103:1196–1207. [PubMed] [Google Scholar]

29. Curcio C, Baqui MM, Salvatore D, et al. The human type 2 iodothyronine deiodinase is a selenoprotein highly expressed in a mesothelioma cell line. The Journal of Biological Chemistry. 2001;276:30183–30187. [PubMed] [Google Scholar]

30. Bianco AC, Silva JE. Nuclear 3,5,3’-triiiodothyronine (T3) in brown adipose tissue: receptor occupancy and sources of T3 as determined by in vivo techniques. Endocrinology. 1987;120:55–62. [PubMed] [Google Scholar]

31. Baqui M, Botero D, Gereben B, et al. Human type 3 iodothyronine selenodeiodinase is located in the plasma membrane and undergoes rapid internalization to endosomes. The Journal of Biological Chemistry. 2003;278:1206–1211. [PubMed] [Google Scholar]

32. Kallo I, Mohacsik P, Vida B, et al. A novel pathway regulates thyroid hormone availability in rat and human hypothalamic neurosecretory neurons. PLoS One. 2012;7:e37860. [PMC free article] [PubMed] [Google Scholar]

33. Jo S, Kallo I, Bardoczi Z, et al. Neuronal hypoxia induces hsp40-mediated nuclear import of type 3 deiodinase as an adaptive mechanism to reduce cellular metabolism. The Journal of Neuroscience. 2012;32:8491–8500. [PMC free article] [PubMed] [Google Scholar]

34. Hernandez A. Structure and function of the type 3 deiodinase gene. Thyroid. 2005;15:865–874. [PubMed] [Google Scholar]

35. Huang SA. Physiology and pathophysiology of type 3 deiodinase in humans. Thyroid. 2005;15:875–881. [PubMed] [Google Scholar]

36. Alexander EK, Marqusee E, Lawrence J, et al. Timing and magnitude of increases in levothyroxine requirements during pregnancy in women with hypothyroidism. The New England Journal of Medicine. 2004;351:241–249. [PubMed] [Google Scholar]

37. Huang SA, Tu HM, Harney JW, et al. Severe hypothyroidism caused by type 3 iodothyronine deiodinase in infantile hemangiomas. The New England Journal of Medicine. 2000;343:185–189. [PubMed] [Google Scholar]

38. Maynard MA, Marino-Enriquez A, Fletcher JA, et al. Thyroid hormone inactivation in gastrointestinal stromal tumors. The New England Journal of Medicine. 2014;370:1327–1334. [PMC free article] [PubMed] [Google Scholar]

39. Curran PG, DeGroot LJ. The effect of hepatic enzyme-inducing drugs on thyroid hormones and the thyroid gland. Endocrine Reviews. 1991;12:135–150. [PubMed] [Google Scholar]

40. Bianco AC, Kim BW. Deiodinases: implications of the local control of thyroid hormone action. The Journal of Clinical Investigation. 2006;116:2571–2579. [PMC free article] [PubMed] [Google Scholar]

41. Andersen S, Bruun NH, Pedersen KM, et al. Biologic variation is important for interpretation of thyroid function tests. Thyroid. 2003;13:1069–1078. [PubMed] [Google Scholar]

42. Russell W, Harrison RF, Smith N, et al. Free triiodothyronine has a distinct circadian rhythm that is delayed but parallels thyrotropin levels. The Journal of Clinical Endocrinology and Metabolism. 2008;93:2300–2306. [PubMed] [Google Scholar]

43. Geffner DL, Azukizawa M, Hershman JM. Propylthiouracil blocks extrathyroidal conversion of thyroxine to triiodothyronine and augments thyrotropin secretion in man. The Journal of Clinical Investigation. 1975;55:224–229. [PMC free article] [PubMed] [Google Scholar]

44. Gereben B, Salvatore D. Pretranslational regulation of type 2 deiodinase. Thyroid. 2005;15:855–864. [PubMed] [Google Scholar]

45. 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. The Journal of Clinical Investigation. 1975;55:1337–1348. [PMC free article] [PubMed] [Google Scholar]

46. Nicoloff JT, Lum SM, Spencer CA, et al. Peripheral autoregulation of thyroxine to triiodothyronine conversion in man. Hormone and Metabolic Research Supplement. 1984;14:74–79. [PubMed] [Google Scholar]

47. Lum SM, Nicoloff JT, Spencer CA, et al. Peripheral tissue mechanism for maintenance of serum triiodothyronine values in a thyroxine-deficient state in man. The Journal of Clinical Investigation. 1984;73:570–575. [PMC free article] [PubMed] [Google Scholar]

48. Gereben B, Goncalves C, Harney JW, et al. Selective proteolysis of human type 2 deiodinase: a novel ubiquitin-proteasomal mediated mechanism for regulation of hormone activation. Molecular Endocrinology. 2000;14:1697–1708. [PubMed] [Google Scholar]

49. Arrojo e Drigo R, Bianco AC. Type 2 deiodinase at the crossroads of thyroid hormone action. International Journal of Biochemistry & Cell Biology. 2011;43:1432–1441. [PMC free article] [PubMed] [Google Scholar]

50. Sagar GD, Gereben B, Callebaut I, et al. The thyroid hormone-inactivating deiodinase functions as a hom*odimer. Molecular Endocrinology. 2008;22:1382–1393. [PMC free article] [PubMed] [Google Scholar]

51. Zeold A, Pormuller L, Dentice M, et al. Metabolic instability of type 2 deiodinase is transferable to stable proteins independently of subcellular localization. The Journal of Biological Chemistry. 2006;281:31538–31543. [PubMed] [Google Scholar]

52. 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. Nature Cell Biology. 2005;7:698–705. [PMC free article] [PubMed] [Google Scholar]

53. Botero D, Gereben B, Goncalves C, et al. Ubc6p and ubc7p are required for normal and substrate-induced endoplasmic reticulum-associated degradation of the human selenoprotein type 2 iodothyronine monodeiodinase. Molecular Endocrinology. 2002;16:1999–2007. [PubMed] [Google Scholar]

54. 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. Molecular Endocrinology. 2003;17:2603–2612. [PubMed] [Google Scholar]

55. Zavacki AM, Arrojo e Drigo R, Freitas BC, et al. The E3 ubiquitin ligase TEB4 mediates degradation of type 2 iodothyronine deiodinase. Molecular and Cellular Biology. 2009;29:5339–5347. [PMC free article] [PubMed] [Google Scholar]

56. 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. The Journal of Clinical Investigation. 2003;112:189–196. [PMC free article] [PubMed] [Google Scholar]

57. Arrojo EDR, Egri P, Jo S, et al. The type II deiodinase is retrotranslocated to the cytoplasm and proteasomes via p97/Atx3 complex. Molecular Endocrinology. 2013;27:2105–2115. [PMC free article] [PubMed] [Google Scholar]

58. Schneider MJ, Fiering SN, Pallud SE, et al. Targeted disruption of the type 2 selenodeiodinase gene (DIO2) results in a phenotype of pituitary resistance to T4. Molecular Endocrinology. 2001;15:2137–2148. [PubMed] [Google Scholar]

59. Berry MJ, Grieco D, Taylor BA, et al. Physiological and genetic analyses of inbred mouse strains with a type I iodothyronine 5’ deiodinase deficiency. The Journal of Clinical Investigation. 1993;92:1517–1528. [PMC free article] [PubMed] [Google Scholar]

60. 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;148:954–960. [PubMed] [Google Scholar]

61. Galton VA, Schneider M, Clark AS, et al. Life without T4 to T3 conversion: studies in mice devoid of the 5’-deiodinases. Endocrinology. 2009;150:2957–2963. [PMC free article] [PubMed] [Google Scholar]

62. Fonseca TL, Correa-Medina M, Campos MP, et al. Coordination of hypothalamic and pituitary T3 production regulates TSH expression. The Journal of Clinical Investigation. 2013;123:1492–1500. [PMC free article] [PubMed] [Google Scholar]

63. Riesco G, Taurog A, Larsen PR, et al. Acute and chronic responses to iodine deficiency in rats. Endocrinology. 1977;100:303–313. [PubMed] [Google Scholar]

64. Santisteban P, Obregon MJ, Rodriguez-Pena A, et al. Are iodine-deficient rats euthyroid? Endocrinology. 1982;110:1780–1789. [PubMed] [Google Scholar]

65. Bianco AC, Anderson G, Forrest D, et al. American thyroid association guide to investigating thyroid hormone economy and action in rodent and cell models. Thyroid. 2014;24:88–168. [PMC free article] [PubMed] [Google Scholar]

66. Huang SA, Bianco AC. Reawakened interest in type III iodothyronine deiodinase in critical illness and injury. Nature Clinical Practice Endocrinology & Metabolism. 2008;4:148–155. [PMC free article] [PubMed] [Google Scholar]

67. Escobar- Morreale HF, Obregon MJ, Escobar del Rey F, et al. Replacement therapy for hypothyroidism with thyroxine alone does not ensure euthyroidism in all tissues, as studied in thyroidectomized rats. The Journal of Clinical Investigation. 1995;96:2828–2838. [PMC free article] [PubMed] [Google Scholar]

68. Escobar- Morreale HF, Rey F, Obregon MJ, et al. Only the combined treatment with thyroxine and triiodothyronine ensures euthyroidism in all tissues of the thyroidectomized rat. Endocrinology. 1996;137:2490–2502. [PubMed] [Google Scholar]

69. Stock JM, Surks MI, Oppenheimer JH. Replacement dosage of L-thyroxine in hypothyroidism. A re-evaluation. The New England Journal of Medicine. 1974;290:529–533. [PubMed] [Google Scholar]

70. Pearce CJ, Himsworth RL. Total and free thyroid hormone concentrations in patients receiving maintenance replacement treatment with thyroxine. British Medical Journal. 1984;288:693–695. [PMC free article] [PubMed] [Google Scholar]

71. 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. [PubMed] [Google Scholar]

72. Gullo D, Latina A, Frasca F, et al. Levothyroxine monotherapy cannot guarantee euthyroidism in all athyreotic patients. PLoS One. 2011;6:e22552. [PMC free article] [PubMed] [Google Scholar]

73. Wiersinga WM, Duntas L, Fadeyev V, et al. 2012 ETA guidelines: the use of L-T4+ L-T3 in the treatment of hypothyroidism. European Thyroid Journal. 2012;1:55–71. [PMC free article] [PubMed] [Google Scholar]

74. Wiersinga WM. Paradigm shifts in thyroid hormone replacement therapies for hypothyroidism. Nature Reviews Endocrinology. 2014;10:164–174. [PubMed] [Google Scholar]

75. Biondi B, Wartofsky L. Combination treatment with T4 and T3: toward personalized replacement therapy in hypothyroidism? The Journal of Clinical Endocrinology and Metabolism. 2012;97:2256–2271. [PubMed] [Google Scholar]

76. 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 and Metabolism. 2005;90:2666–2674. [PubMed] [Google Scholar]

77. Nygaard B, Jensen EW, Kvetny J, et al. 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;161:895–902. [PubMed] [Google Scholar]

78. Bunevicius R, Kazanavicius G, Zalinkevicius R, et al. Effects of thyroxine as compared with thyroxine plus triiodothyronine in patients with hypothyroidism. The New England Journal of Medicine. 1999;340:424–429. [PubMed] [Google Scholar]

79. Clyde PW, Harari AE, Getka EJ, et al. Combined levothyroxine plus liothyronine compared with levothyroxine alone in primary hypothyroidism: a randomized controlled trial. JAMA. 2003;290:2952–2958. [PubMed] [Google Scholar]

80. Siegmund W, Spieker K, Weike AI, et al. Replacement therapy with levothyroxine plus triiodothyronine (bioavailable molar ratio 14:1) is not superior to thyroxine alone to improve well-being and cognitive performance in hypothyroidism. Clinical Endocrinology. 2004;60:750–757. [PubMed] [Google Scholar]

81. 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 and Metabolism. 2003;88:4543–4550. [PubMed] [Google Scholar]

82. Grozinsky-Glasberg S, Fraser A, Nahshoni E, et al. Thyroxine-triiodothyronine combination therapy versus thyroxine monotherapy for clinical hypothyroidism: meta-analysis of randomized controlled trials. The Journal of Clinical Endocrinology and Metabolism. 2006;91:2592–2599. [PubMed] [Google Scholar]

83. Ma C, Xie J, Huang X, et al. Thyroxine alone or thyroxine plus triiodothyronine replacement therapy for hypothyroidism. Nuclear Medicine Communications. 2009;30:586–593. [PubMed] [Google Scholar]

84. 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 and Metabolism. 2009;94:1623–1629. [PubMed] [Google Scholar]

85. Saravanan P, Siddique H, Simmons DJ, et al. Twenty-four hour hormone profiles of TSH, Free T3 and free T4 in hypothyroid patients on combined T3/T4 therapy. Experimental and Clinical Endocrinology & Diabetes. 2007;115:261–267. [PubMed] [Google Scholar]

86. Celi FS, Zemskova M, Linderman JD, et al. Metabolic effects of liothyronine therapy in hypothyroidism: a randomized, double-blind, crossover trial of liothyronine versus levothyroxine. The Journal of Clinical Endocrinology and Metabolism. 2011;96:3466–3474. [PMC free article] [PubMed] [Google Scholar]

87. Santini F, Giannetti M, Ricco I, et al. Steady state serum T3 concentrations for 48 hours following the oral administration of a single dose of 3,5,3’-Triiodothyronine Sulfate (T3S) Endocrine Practice. 2014;???:1–25. [PubMed] [Google Scholar]

Defending plasma T3 is a biological priority (2024)

FAQs

What is the biological importance of T3? ›

Moreover, T3-regulated secreted proteins participate in physiological processes or cellular transformation. T3 has been employed as a marker in several disorders, such as cardiovascular disorder in chronic kidney disease, as well as diseases of the liver, immune system, endocrine hormone metabolism and coronary artery.

Is T3 biologically active? ›

Even though both molecules can trigger biological effects, T3 is considered the biologically active thyroid hormone that binds to thyroid hormone receptors (TR), while T4 is a prohormone that must be converted to T3 in order to initiate signalling and gain biological activity.

What is the biological reference interval for T3? ›

A normal Total T3 level in adults ranges from 80-220 ng/dL. Free T3 assays are often unreliable and not routinely used to assess thyroid function.

Why is free T3 so important? ›

Free T3 testing may be used to determine whether thyroid replacement therapy is effective. It can also help diagnose abnormalities of the proteins that bind thyroid hormones. Total T3 is considered by many experts to be a more reliable measurement than free T3.

What is the biological importance of T3 and T4? ›

The thyroid gland releases triiodothyronine (T3) and thyroxine (T4). These hormones play an important role in regulation of your weight, energy levels, internal temperature, skin, hair, nail growth, metabolism and is an important part of the endocrine system.

What happens if your T3 is low? ›

Low T3 levels (free or total) may mean hypothyroidism (too little thyroid hormone). Low or normal T3 levels with: High TSH levels and low free T4 levels may mean that a problem with your thyroid gland is causing too little thyroid hormone.

What organ does T3 target? ›

Triiodothyronine is a thyroid hormone that plays vital roles in the body's metabolic rate, heart and digestive functions, muscle control, brain development and function, and the maintenance of bones.

How does T3 make you feel? ›

T3 testing generally does not cause side effects, but there is a slight risk of throbbing, soreness, or mild bruising at the injection site. Some people might also feel lightheaded during or just after the procedure.

What produces T3 in the body? ›

T3 is an important hormone that is produced by the thyroid gland. Together with the other hormones produced by the thyroid (T4, rT3, and calcitonin), T3 is crucial for processes including metabolism, heart rate, growth, and development.

How to reduce T3 levels naturally? ›

Additional studies have indicated that bugleweed and lemon balm may block or decrease thyroid-stimulating hormone (TSH) and reduce T3 and T4 hormone levels, which would reduce the symptoms of hyperthyroidism [53].

What is the optimal T3 level? ›

The standard lab reference range for free T3 is 2.0−4.4 pg/mL. Low free T3 (or high reverse T3) can be a byproduct of poor nutrition, gut inflammation, and chronic illness [6].

What if T3 is high but T4 is normal? ›

T3 tests are often useful to diagnosis hyperthyroidism or to determine the severity of the hyperthyroidism. Patients who are hyperthyroid will have an elevated T3 level. In some individuals with a low TSH, only the T3 is elevated and the FT4 or FTI is normal.

Why don't doctors test for free T3? ›

Providers don't usually order free T3 tests because they're not as reliable, but it is possible to test these levels. In general, normal ranges of free T3 for healthy people include: Infants up to 3 days old: 1.4 – 5.4 picograms per milliliter (pg/mL).

Can low T3 cause anxiety? ›

Can thyroid problems cause anxiety and panic attacks? Hypothyroidism, which means your thyroid is not making enough hormones, can be responsible for anxiety and even panic attacks. You may get relief when you are treated for your thyroid disorder with a medication to replace the thyroid hormone you are not making.

How do you know if you need T3? ›

The T3 test is particularly useful in diagnosing hyperthyroidism. In this condition, the thyroid overproduces hormones, causing symptoms such as a fast heart rate, weight loss, trembling, and sweating. Tell the doctor about any medicines your child takes because some can affect the test results.

Is T3 active or inactive? ›

The thyroid produces approximately 90% inactive thyroid hormone, or thyroxine (T4), and 10% active thyroid hormone, or triiodothyronine (T3).

Is T3 or T4 biologically active? ›

T3 is regarded as the biologically active hormone because it binds to TH receptors (TRs) with greater affinity than T4. However, it is currently unclear what structural mechanisms underlie this difference in affinity.

How is T3 activated? ›

Even though the thyroid contributes with a small fraction of the circulating T3, thyroidal T3 secretion is upregulated in response to TSH stimulation. This occurs through an increase in the T3/T4 ratio in the thyroglobulin and through increased thyroidal conversion of T4 to T3.

Is T3 or T3 active? ›

T3 is the active form of thyroid hormone, meaning it impacts cells in your body, whereas T4 is the inactive form of thyroid hormone. Your liver and kidneys convert most of the T4 your thyroid releases into T3.

Top Articles
Latest Posts
Article information

Author: Horacio Brakus JD

Last Updated:

Views: 6185

Rating: 4 / 5 (51 voted)

Reviews: 90% of readers found this page helpful

Author information

Name: Horacio Brakus JD

Birthday: 1999-08-21

Address: Apt. 524 43384 Minnie Prairie, South Edda, MA 62804

Phone: +5931039998219

Job: Sales Strategist

Hobby: Sculling, Kitesurfing, Orienteering, Painting, Computer programming, Creative writing, Scuba diving

Introduction: My name is Horacio Brakus JD, I am a lively, splendid, jolly, vivacious, vast, cheerful, agreeable person who loves writing and wants to share my knowledge and understanding with you.