Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
JCI Insight ; 8(23)2023 Dec 08.
Article En | MEDLINE | ID: mdl-37856222

Thyroid hormone (TH) levels are low during development, and the deiodinases control TH signaling through tissue-specific activation or inactivation of TH. Here, we studied human induced pluripotent stem cell-derived (iPSC-derived) hepatic organoids and identified a robust induction of DIO2 expression (the deiodinase that activates T4 to T3) that occurs in hepatoblasts. The surge in DIO2-T3 (the deiodinase that activates thyroxine [T4] to triiodothyronine [T3]) persists until the hepatoblasts differentiate into hepatocyte- or cholangiocyte-like cells, neither of which expresses DIO2. Preventing the induction of the DIO2-T3 signaling modified the expression of key transcription factors, decreased the number of hepatocyte-like cells by ~60%, and increased the number of cholangiocyte-like cells by ~55% without affecting the growth or the size of the mature liver organoid. Physiological levels of T3 could not fully restore the transition from hepatoblasts to mature cells. This indicates that the timed surge in DIO2-T3 signaling critically determines the fate of developing human hepatoblasts and the transcriptome of the maturing hepatocytes, with physiological and clinical implications for how the liver handles energy substrates.


Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Transcriptome , Liver/metabolism , Hepatocytes/metabolism , Thyroid Hormones/metabolism , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Organoids/metabolism
2.
Thyroid ; 33(1): 109-118, 2023 01.
Article En | MEDLINE | ID: mdl-36322711

Background: Non-Thyroidal Illness Syndrome (NTIS) caused by infection or fasting is hallmarked by reduced circulating thyroid hormone (TH) levels. To better understand the role of local TH-action in the development of NTIS, we assessed tissue-specific changes of TH signaling in Thyroid Hormone Action Indicator (THAI) mice. Methods: NTIS was induced in young adult THAI mice by bacterial lipopolysaccharide (LPS)-administration or by 24 or 48 hours' fasting. Tissue-specific TH-action was assessed by the detection of changes of the Luciferase reporter of THAI mice with quantitative polymerase chain reaction along with tissue-specific examination of regulators of TH metabolism and signaling. Age dependence of revealed alterations of hypothalamic TH-action was also studied in 1-year-old male THAI mice. Results: LPS-treatment increased TH-action in the hypothalamic arcuate nucleus-median eminence (ARC-ME) region preceded by an increase of type 2 deiodinase (D2) expression in the same region and followed by the suppression of proTrh expression in the hypothalamic paraventricular nucleus (PVN). In contrast, LPS decreased both TH-action and D2 activity in the pituitary at both ages. Tshß expression and serum free thyroxine (fT4) and free triiodothyronine (fT3) levels decreased in LPS-treated young adults. Tshß expression and serum fT4 levels were not significantly affected by LPS treatment in aged animals. In contrast to LPS treatment, TH-action remained unchanged in the ARC-ME of 24 and 48 hours fasted animals accompanied with a modest decrease of proTrh expression in the PVN in the 24-hour group. Tshß expression and fT3 level were decreased in both fasted groups, but the fT4 decreased only in the 48 hours fasted animals. Conclusions: Although the hypothalamo-pituitary-thyroid (HPT) axis is inhibited both in LPS and fasting-induced NTIS, LPS achieves this by centrally inducing local hyperthyroidism in the ARC-ME region, while fasting acts without affecting hypothalamic TH signaling. Lack of downregulation of Tshß and fT4 in LPS-treated aged THAI mice suggests age-dependent alterations in the responsiveness of the HPT axis. The LPS-induced tissue-specific hypo-, eu-, and hyperthyroidism in different tissues of the same animal indicate that under certain conditions TH levels alone could be a poor marker of tissue TH signaling. In conclusion, decreased circulating TH levels in these two forms of NTIS are associated with different patterns of hypothalamic TH signaling.


Euthyroid Sick Syndromes , Hypothalamus , Thyroid Hormones , Animals , Male , Mice , Euthyroid Sick Syndromes/chemically induced , Euthyroid Sick Syndromes/metabolism , Euthyroid Sick Syndromes/pathology , Fasting , Hyperthyroidism , Hypothalamo-Hypophyseal System/metabolism , Lipopolysaccharides/metabolism , Thyroid Hormones/metabolism , Hypothalamus/metabolism
3.
Behav Brain Res ; 436: 114082, 2023 01 05.
Article En | MEDLINE | ID: mdl-36041571

Postpartum depression is a mentally disabling disease with multifactorial etiology that affects women worldwide. It can also influence child development and lead to behavioral and cognitive alterations. Despite the high prevalence, the disease is underdiagnosed and poorly studied. To study the postpartum depression caused by maternal separation model in rats, dams were separated from their litter for 3 h daily starting from lactating day (LD) 2 through LD12. Maternal studies were conducted from LD5 to LD21 and the offspring studies from postnatal day (PND) 2 through PND90. The stress caused by the dam-offspring separation led to poor maternal care and a transient increase in anxiety in the offspring detected during infancy. The female offspring also exhibited a permanent impairment in sociability during adult life. These changes were associated with neurochemical alterations in the prefrontal cortex and hippocampus, and low TSH concentrations in the dams, and in the hypothalamus, hippocampus and striatum of the offspring. These results indicate that the postpartum depression resulted in a depressive phenotype, changes in the brain neurochemistry and in thyroid economy that remained until the end of lactation. Changes observed in the offspring were long-lasting and resemble what is observed in children of depressant mothers.


Depression, Postpartum , Animals , Corticosterone , Disease Models, Animal , Female , Lactation , Maternal Deprivation , Rats , Rats, Sprague-Dawley , Stress, Psychological/etiology , Thyrotropin
4.
Clin Case Rep ; 10(4): e05651, 2022 Apr.
Article En | MEDLINE | ID: mdl-35432999

We report a 54-year-old man with treatment-resistant depression (TRD) and hypothyroidism who responded to LT3/LT4 combination, rather than LT4 alone. He was able to discontinue all antidepressant medications eventually. Interestingly, the patient has a DIO2 polymorphism.

5.
Endocrinology ; 163(5)2022 05 01.
Article En | MEDLINE | ID: mdl-35238380

Adaptive thermogenesis in small mammals and infants takes place in brown adipose tissue (BAT). Heat is produced via uncoupling protein 1 (UCP1)-mediated uncoupling between oxidation of energy substrates and adenosine 5'-triphosphate synthesis. Thyroid hormone (TH) signaling plays a role in this process. The deiodinases activate thyroxine (T4) to 3,5,3'-triiodothyronine (T3) (D2) or inactivate T4 and T3 to 3,3,5'-triiodothyronine and T2 (D3), respectively. Using a mouse model with selective inactivation of Dio3 in BAT (flox-Dio3 × UCP1-cre = BAT-D3KO), we now show that knocking out D3 resulted in premature exposure of developing brown adipocytes (embryonic days 16.5-18.5) to T3 signaling, leading to an earlier expression of key BAT genes, including Cidea, Cox8b, Dio2, Ucp1, and Pgc1α. Adult BAT-D3KO mice exhibited increased expression of 1591 genes as assessed by RNA sequencing, including 19 gene sets related to mitochondria, 8 related to fat, and 8 related to glucose homeostasis. The expression of 243 genes was changed by more than 1.5-fold, 36 of which play a role in metabolic/thermogenic processes. BAT-D3KO mice weigh less and exhibit smaller white adipocyte area, but maintain normal energy expenditure at room temperature (22 °C) and in the cold (4 °C). They also defend their core temperature more effectively and do not lose as much body weight when exposed to cold. We conclude that the coordinated actions of Dio3 in the embryonic BAT define the timing and intensity of T3 signaling during brown adipogenesis. Enhanced T3 signaling during BAT embryogenesis (Dio3 inactivation) results in selective life-long modifications in the BAT transcriptome.


Adipose Tissue, Brown , Iodide Peroxidase , Adipose Tissue, Brown/metabolism , Animals , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Male , Mammals/genetics , Thermogenesis/genetics , Transcriptome , Triiodothyronine/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
6.
Nat Commun ; 12(1): 4446, 2021 07 21.
Article En | MEDLINE | ID: mdl-34290257

The type 2 deiodinase (D2) in the neonatal liver accelerates local thyroid hormone triiodothyronine (T3) production and expression of T3-responsive genes. Here we show that this surge in T3 permanently modifies hepatic gene expression. Liver-specific Dio2 inactivation (Alb-D2KO) transiently increases H3K9me3 levels during post-natal days 1-5 (P1-P5), and results in methylation of 1,508 DNA sites (H-sites) in the adult mouse liver. These sites are associated with 1,551 areas of reduced chromatin accessibility (RCA) within core promoters and 2,426 within intergenic regions, with reduction in the expression of 1,363 genes. There is strong spatial correlation between density of H-sites and RCA sites. Chromosome conformation capture (Hi-C) data reveals a set of 81 repressed genes with a promoter RCA in contact with an intergenic RCA ~300 Kbp apart, within the same topologically associating domain (χ2 = 777; p < 0.00001). These data explain how the systemic hormone T3 acts locally during development to define future expression of hepatic genes.


Epigenesis, Genetic , Liver/metabolism , Triiodothyronine/metabolism , Animals , Animals, Newborn , Chromatin/metabolism , DNA Methylation , Gene Expression , Gene Expression Regulation, Developmental , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Liver/growth & development , Mice , Mice, Knockout , Iodothyronine Deiodinase Type II
7.
J Clin Endocrinol Metab ; 106(11): e4400-e4413, 2021 10 21.
Article En | MEDLINE | ID: mdl-34185829

INTRODUCTION: Studies comparing levothyroxine (LT4) therapy with LT4 + liothyronine (LT3) or desiccated thyroid extract (DTE) did not detect consistent superiority of either treatment. Here, we investigated these therapies, focusing on the whole group of LT4-treated hypothyroid patients, while also exploring the most symptomatic patients. METHODOLOGY: Prospective, randomized, double-blind, crossover study of 75 hypothyroid patients randomly allocated to 1 of 3 treatment arms, LT4, LT4 + LT3, and DTE, for 22 weeks. The primary outcomes were posttreatment scores on the 36-point thyroid symptom questionnaire (TSQ-36), 12-point quality of life general health questionnaire (GHQ-12), the Wechsler memory scale-version IV (VMS-IV), and the Beck Depression Inventory (BDI). Secondary endpoints included treatment preference, biochemical and metabolic parameters, etiology of hypothyroidism, and Thr92Ala-DIO2 gene polymorphism. Analyses were performed with a linear mixed model using subject as a random factor and group as a fixed effect. RESULTS: Serum TSH remained within reference range across all treatment arms. There were no differences for primary and secondary outcomes, except for a minor increase in heart rate caused by DTE. Treatment preference was not different and there were no interferences of the etiology of hypothyroidism or Thr92Ala-DIO2 gene polymorphism in the outcomes. Subgroup analyses of the 1/3 most symptomatic patients on LT4 revealed strong preference for treatment containing T3, which improved performance on TSQ-36, GHQ-12, BDI, and visual memory index (VMS-IV component). CONCLUSIONS: As a group, outcomes were similar among hypothyroid patients taking DTE vs LT4 + T3 vs LT4. However, those patients that were most symptomatic on LT4 preferred and responded positively to therapy with LT4 + LT3 or DTE.


Hypothyroidism/drug therapy , Thyroid Gland/chemistry , Thyroxine/administration & dosage , Tissue Extracts/administration & dosage , Triiodothyronine/administration & dosage , Adult , Aged , Cross-Over Studies , Desiccation , Double-Blind Method , Female , Hormone Replacement Therapy/methods , Humans , Hypothyroidism/physiopathology , Male , Middle Aged , Placebos , Prospective Studies , Quality of Life , Surveys and Questionnaires , Treatment Outcome , Triiodothyronine/blood
8.
Endocrinology ; 161(12)2020 12 01.
Article En | MEDLINE | ID: mdl-33091112

More than a billion people worldwide are at risk of iodine deficiency (ID), with well-known consequences for development of the central nervous system. Furthermore, ID has also been associated with dyslipidemia and obesity in humans. To further understand the metabolic consequences of ID, here we kept 8-week-old C57/Bl6 mice at thermoneutrality (~28°C) while feeding them on a low iodine diet (LID). When compared with mice kept on control diet (LID + 0.71 µg/g iodine), the LID mice exhibited marked reduction in T4 and elevated plasma TSH, without changes in plasma T3 levels. LID mice grew normally, and had normal oxygen consumption, ambulatory activity, and heart expression of T3-responsive gene, confirming systemic euthyroidism. However, LID mice exhibited ~5% lower respiratory quotient (RQ), which reflected a ~2.3-fold higher contribution of fat to energy expenditure. LID mice also presented increased circulating levels of nonesterified fatty acids, ~60% smaller fat depots, and increased hepatic glycogen content, all indicative of accelerated lipolysis. LID mice responded much less to forced mobilization of energy substrates (50% food restriction for 3 days or starvation during 36 hours) because of limited size of the adipose depots. A 4-day treatment with T4 restored plasma T4 and TSH levels in LID mice and normalized RQ. We conclude that ID accelerates lipolysis and fatty acid oxidation, without affecting systemic thyroid hormone signaling. It is conceivable that the elevated plasma TSH levels trigger these changes by directly activating lipolysis in the adipose tissues.


Adipose Tissue/metabolism , Energy Metabolism/physiology , Iodine/deficiency , Lipid Metabolism/physiology , Thyrotropin/blood , Thyroxine/blood , Animals , Fatty Acids/metabolism , Lipolysis/physiology , Male , Mice
9.
J Clin Endocrinol Metab ; 105(2)2020 02 01.
Article En | MEDLINE | ID: mdl-31581295

INTRODUCTION: The inflammatory response of critical illness is accompanied by nonthyroidal illness syndrome (NTIS). Feeding has been shown to attenuate this process, but this has not been explored prospectively over time in critically ill patients. OBJECTIVE: To explore the impact of calorie exposure on NTIS over time in critically ill patients. METHODS: Mechanically ventilated patients with systemic inflammatory response syndrome (SIRS) were randomized to receive either 100% or 40% of their estimated caloric needs (ECN). Thyroid hormones were measured daily for 7 days or until intensive care unit discharge or death. Mixed level regression modeling was used to explore the effect of randomization group on plasma triiodothyronine (T3), reverse triiodothyronine (rT3), thyroxine (T4), and thyroid stimulating hormone (TSH), as well as the T3/rT3 ratio. RESULTS: Thirty-five participants (n=19 in 100% ECN; n=16 in 40% ECN) were recruited. Adjusting for group differences in baseline T3/rT3 ratio, the parameters defining the fitted curves (intercept, linear effect of study day, and quadratic effect of study day) differed by randomization group (P = 0.001, P = 0.01, and P = 0.02 respectively). Plots of the fitted curves revealed that participants in the 100% ECN group had a 54% higher T3/rT3 ratio on postintervention day 1 compared with the 40% ECN group, a difference which attenuated over time. This was driven by a 23% higher plasma T3 and 10% lower plasma rT3 levels on postintervention 1. CONCLUSIONS: Higher caloric exposure in NTIS patients transiently attenuates the drop of the plasma T3/rT3 ratio, an effect that is minimized and finally lost over the following 3 days of continued higher caloric exposure.


Energy Intake/physiology , Enteral Nutrition/methods , Euthyroid Sick Syndromes/blood , Euthyroid Sick Syndromes/therapy , Critical Illness/therapy , Female , Humans , Male , Middle Aged , Regression Analysis , Respiration, Artificial , Thyrotropin/blood , Thyroxine/blood , Treatment Outcome , Triiodothyronine/blood , Triiodothyronine, Reverse/blood
10.
Article En | MEDLINE | ID: mdl-31156551

C3H/HeJ (C3H) mice are deficient of type I deiodinase (D1), an enzyme that activates thyroid hormone (TH), converting thyroxine (T4) to triiodothyronine (T3). Nevertheless, C3H mice present normal serum T3 and a gross euthyroid phenotype. To investigate if a global D1 deficiency interferes in the TH effects on bone, we compared bone growth, bone mass accrual and bone strength of C3H and C57BL/6J (B6) mice under abnormal TH status. Four-week-old female mice of both strains were grouped as Euthyroid, Hypothyroid (pharmacologically-induced), 1xT4 and 10xT4 (hypothyroid animals receiving 1- or 10-fold the physiological dose of T4 /day/16 weeks). Hypothyroidism and TH excess similarly impaired body weight (BW) gain and body growth in both mice strains. In contrast, whereas hypothyroidism only slightly impaired bone mineral density (BMD) accrual in B6 mice, it severely impaired BMD accrual in C3H mice. No differences were observed in serum and bone concentrations of T3 between hypothyroid animals of both strains. Interestingly, treatment with 10xT4 was less deleterious to BMD accrual in C3H than in B6 mice and resulted in less elevated T3 serum levels in B6 than in C3H mice, which is probably explained by the lower D1 activity in C3H mice. In addition, hypothyroidism decreased bone strength only in C3H but not in B6 mice, while TH excess decreased this parameter in both strains. These findings indicate that D1 deficiency contributes to the TH excess-induced differences in bone mass accrual in C3H vs. B6 mice and suggest that deiodinase-unrelated genetic factors might account for the different skeleton responses to hypothyroidism between strains.

11.
Endocr Rev ; 40(4): 1000-1047, 2019 08 01.
Article En | MEDLINE | ID: mdl-31033998

Thyroid hormone (TH) molecules enter cells via membrane transporters and, depending on the cell type, can be activated (i.e., T4 to T3 conversion) or inactivated (i.e., T3 to 3,3'-diiodo-l-thyronine or T4 to reverse T3 conversion). These reactions are catalyzed by the deiodinases. The biologically active hormone, T3, eventually binds to intracellular TH receptors (TRs), TRα and TRß, and initiate TH signaling, that is, regulation of target genes and other metabolic pathways. At least three families of transmembrane transporters, MCT, OATP, and LAT, facilitate the entry of TH into cells, which follow the gradient of free hormone between the extracellular fluid and the cytoplasm. Inactivation or marked downregulation of TH transporters can dampen TH signaling. At the same time, dynamic modifications in the expression or activity of TRs and transcriptional coregulators can affect positively or negatively the intensity of TH signaling. However, the deiodinases are the element that provides greatest amplitude in dynamic control of TH signaling. Cells that express the activating deiodinase DIO2 can rapidly enhance TH signaling due to intracellular buildup of T3. In contrast, TH signaling is dampened in cells that express the inactivating deiodinase DIO3. This explains how THs can regulate pathways in development, metabolism, and growth, despite rather stable levels in the circulation. As a consequence, TH signaling is unique for each cell (tissue or organ), depending on circulating TH levels and on the exclusive blend of transporters, deiodinases, and TRs present in each cell. In this review we explore the key mechanisms underlying customization of TH signaling during development, in health and in disease states.


Signal Transduction , Thyroid Hormones/metabolism , Animals , Female , Humans , Iodide Peroxidase/metabolism , Male , Receptors, Thyroid Hormone/metabolism
12.
Alcohol Clin Exp Res ; 43(7): 1376-1383, 2019 07.
Article En | MEDLINE | ID: mdl-30908637

BACKGROUND: A mouse with hepatocyte-specific deiodinase type II inactivation (Alb-D2KO) is resistant to diet-induced obesity, hepatic steatosis, and hypertriglyceridemia due to perinatal epigenetic modifications in the liver. This phenotype is linked to low levels of Zfp125, a hepatic transcriptional repressor that promotes liver steatosis by inhibiting genes involved in packaging and secretion of very-low-density lipoprotein. METHODS: Here, we used chronic and binge ethanol (EtOH) in mice to cause liver steatosis. RESULTS: The EtOH treatment causes a 2.3-fold increase in hepatic triglyceride content; Zfp125 levels were approximately 50% higher in these animals. In contrast, Alb-D2KO mice did not develop EtOH-induced liver steatosis. They also failed to elevate Zfp125 to the same levels, despite being on the EtOH-containing diet for the same period of time. Their phenotype was associated with 1.3- to 2.9-fold up-regulation of hepatic genes involved in lipid transport and export that are normally repressed by Zfp125, that is, Mttp, Abca1, Ldlr, Apoc1, Apoc3, Apoe, Apoh, and Azgp1. Furthermore, genes involved in the EtOH metabolic pathway, that is, Aldh2 and Acss2, were also 1.6- to 3.1-fold up-regulated in Alb-D2KO EtOH mice compared with control animals kept on EtOH. CONCLUSIONS: EtOH consumption elevates expression of Zfp125. Alb-D2KO animals, which have lower levels of Zfp125, are much less susceptible to EtOH-induced liver steatosis.


Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/prevention & control , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Liver/metabolism , Alcoholism/complications , Alcoholism/genetics , Animals , Binge Drinking , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diet , Ethanol/metabolism , Fatty Liver , Fatty Liver, Alcoholic/metabolism , Gene Expression Regulation , Lipid Metabolism/genetics , Metabolic Networks and Pathways/genetics , Mice , Mice, Knockout , Triglycerides/metabolism , Iodothyronine Deiodinase Type II
13.
J Clin Invest ; 129(1): 230-245, 2019 01 02.
Article En | MEDLINE | ID: mdl-30352046

Levothyroxine (LT4) is a form of thyroid hormone used to treat hypothyroidism. In the brain, T4 is converted to the active form T3 by type 2 deiodinase (D2). Thus, it is intriguing that carriers of the Thr92Ala polymorphism in the D2 gene (DIO2) exhibit clinical improvement when liothyronine (LT3) is added to LT4 therapy. Here, we report that D2 is a cargo protein in ER Golgi intermediary compartment (ERGIC) vesicles, recycling between ER and Golgi. The Thr92-to-Ala substitution (Ala92-D2) caused ER stress and activated the unfolded protein response (UPR). Ala92-D2 accumulated in the trans-Golgi and generated less T3, which was restored by eliminating ER stress with the chemical chaperone 4-phenyl butyric acid (4-PBA). An Ala92-Dio2 polymorphism-carrying mouse exhibited UPR and hypothyroidism in distinct brain areas. The mouse refrained from physical activity, slept more, and required additional time to memorize objects. Enhancing T3 signaling in the brain with LT3 improved cognition, whereas restoring proteostasis with 4-PBA eliminated the Ala92-Dio2 phenotype. In contrast, primary hypothyroidism intensified the Ala92-Dio2 phenotype, with only partial response to LT4 therapy. Disruption of cellular proteostasis and reduced Ala92-D2 activity may explain the failure of LT4 therapy in carriers of Thr92Ala-DIO2.


Brain , Endoplasmic Reticulum Stress , Hypothyroidism , Iodide Peroxidase , Polymorphism, Genetic , Unfolded Protein Response , Amino Acid Substitution , Animals , Brain/enzymology , Brain/pathology , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Golgi Apparatus/enzymology , Golgi Apparatus/genetics , HEK293 Cells , Humans , Hypothyroidism/drug therapy , Hypothyroidism/enzymology , Hypothyroidism/genetics , Hypothyroidism/pathology , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Mice , Mice, Transgenic , Mutation, Missense , Thyroxine/therapeutic use , Triiodothyronine/therapeutic use , Iodothyronine Deiodinase Type II
14.
Thyroid ; 28(11): 1425-1433, 2018 11.
Article En | MEDLINE | ID: mdl-30301431

BACKGROUND: Liothyronine (LT3) has limited short-term clinical applications, all of which aim at suppressing thyrotropin (TSH) secretion. A more controversial application is chronic administration along with levothyroxine in the treatment of hypothyroidism. Long-term treatment with LT3 is complicated by its unique pharmacokinetics that result in a substantial triiodothyronine (T3) peak in the blood three to four hours after oral dosing. This is a significant problem, given that T3 levels in the blood are normally stable, varying by <10% throughout the day. METHODS: A metal coordinated form of LT3 (Zn[T3][H2O])n, known as poly-zinc-liothyronine (PZL), was synthesized and loaded into coated gelatin capsules for delivery to the duodenum where sustained release of T3 from PZL occurs. Male Wistar rats were made hypothyroid by feeding on a low iodine diet and water containing 0.05% methimazole for five to six weeks. Rats were given a capsule containing 24 µg/kg PZL or equimolar amounts of LT3. Blood samples were obtained multiple times from the tail vein during the first 16 hours, and processed for T3 and TSH serum levels. Some animals were treated daily for eight days, and blood samples were collected daily. RESULTS: Rats given LT3 exhibited the expected serum T3 peak (about fivefold baseline) at 3.5 hours, followed by a rapid decline, with serum levels almost returning to baseline values by 16 hours. In contrast, serum T3 in PZL-treated rats exhibited about a 30% lower T3 peak at nine hours. Furthermore, the plateau time, that is, the time-span during which the serum T3 concentration is at least half of T3 peak, increased from 4.9 to 7.7 hours in LT3- versus PZL-treated rats, respectively. Serum TSH dropped in both groups, but PZL-treated rats exhibited a more gradual decrease, which was delayed by about four hours compared to LT3-treated rats. Chronic treatment with either LT3 or PZL restored growth, lowered serum cholesterol, and stimulated hepatic expression of the Dio1 mRNA and other T3-dependent markers in the central nervous system. CONCLUSION: Capsules of PZL given orally restore T3-dependent biological effects while exhibiting a reduced and delayed serum T3 peak after dosing, thus providing a longer period of relatively stable serum T3 levels compared to capsules of LT3.


Hypothyroidism/drug therapy , Triiodothyronine/blood , Animals , Hypothyroidism/blood , Male , Rats , Rats, Wistar , Thyrotropin/blood , Treatment Outcome , Triiodothyronine/therapeutic use
15.
Endocrinology ; 159(8): 3090-3104, 2018 08 01.
Article En | MEDLINE | ID: mdl-29905787

Status epilepticus (SE) is an abnormally prolonged seizure that results from either a failure of mechanisms that terminate seizures or from initiating mechanisms that inherently lead to prolonged seizures. Here we report that mice experiencing a 3 hours of SE caused by pilocarpine exhibit a rapid increase in expression of type 2 iodothyronine deiodinase gene (Dio2) and a decrease in the expression of type 3 iodothyronine deiodinase gene in hippocampus, amygdala and prefrontal cortex. Type 3 iodothyronine deiodinase in hippocampal sections was seen concentrated in the neuronal nuclei, typical of ischemic injury of the brain. An unbiased analysis of the hippocampal transcriptome of mice undergoing 3 hours of SE revealed a number of genes, including those involved with response to oxidative stress, cellular homeostasis, cell signaling, and mitochondrial structure. In contrast, in mice with targeted disruption of Dio2 in astrocytes (Astro D2KO mouse), the highly induced genes in the hippocampus were related to inflammation, apoptosis, and cell death. We propose that Dio2 induction caused by SE accelerates production of T3 in different areas of the central nervous system and modifies the hippocampal gene expression profile, affecting the balance between adaptive and maladaptive mechanisms.


Gene Expression , Hippocampus/metabolism , Iodide Peroxidase/genetics , Status Epilepticus/genetics , Triiodothyronine/metabolism , Amygdala/metabolism , Animals , Apoptosis/genetics , Astrocytes/metabolism , Cell Death/genetics , Cell Nucleus/metabolism , Inflammation/genetics , Iodide Peroxidase/metabolism , Male , Mice , Mice, Knockout , Muscarinic Agonists/toxicity , Neurons/metabolism , Oxidative Stress/genetics , Pilocarpine/toxicity , Prefrontal Cortex/metabolism , Signal Transduction , Status Epilepticus/chemically induced , Iodothyronine Deiodinase Type II
16.
Cell Rep ; 22(2): 523-534, 2018 01 09.
Article En | MEDLINE | ID: mdl-29320745

Liver-specific disruption of the type 2 deiodinase gene (Alb-D2KO) results in resistance to both diet-induced obesity and liver steatosis in mice. Here, we report that this is explained by an ∼60% reduction in liver zinc-finger protein-125 (Zfp125) expression. Zfp125 is a Foxo1-inducible transcriptional repressor that causes lipid accumulation in the AML12 mouse hepatic cell line and liver steatosis in mice by reducing liver secretion of triglycerides and hepatocyte efflux of cholesterol. Zfp125 acts by repressing 18 genes involved in lipoprotein structure, lipid binding, and transport. The ApoE promoter contains a functional Zfp125-binding element that is also present in 17 other lipid-related genes repressed by Zfp125. 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.


DNA-Binding Proteins/genetics , Fatty Liver/genetics , Forkhead Box Protein O1/genetics , Hypercholesterolemia/genetics , Animals , DNA-Binding Proteins/metabolism , Fatty Liver/pathology , Forkhead Box Protein O1/metabolism , Mice
17.
Thyroid ; 27(4): 577-586, 2017 04.
Article En | MEDLINE | ID: mdl-27967605

BACKGROUND: Myogenesis is positively regulated by thyroid hormone (triiodothyronine [T3]), which is amplified by the type 2 deiodinase (D2) activation of thyroxine to T3. Global inactivation of the Dio2 gene impairs skeletal muscle (SKM) differentiation and regeneration in response to muscle injury. Given that newborn and adult mice with late developmental SKM Dio2 disruption do not develop a significant phenotype, it was hypothesized that D2 plays an early role in this process. METHODS: This was tested in mice with SKM disruption of Dio2 driven by two early developmental promoters: MYF5 and MYOD. RESULTS: MYF5 myoblasts in culture differentiate normally into myotubes, despite loss of almost all D2 activity. Dio2 mRNA levels in developing SKM obtained from MYF5-D2KO embryos (E18.5) were about 54% of control littermates, but the expression of the T3-responsive genes Myh1 and 7 and Atp2a1 and 2 were not affected. In MYF5-D2KO and MYOD-D2KO neonatal hind-limb muscle, the expression of Myh1 and 7 and Atp2a2 remained unaffected, despite 60-70% loss in D2 activity and/or mRNA. Only in MYOD-D2KO neonatal muscle was there a 40% reduction in Atp2a1 mRNA. Postnatal growth of both mouse models and SKM function as assessed by exercise capacity and measurement of muscle strength were normal. Furthermore, an analysis of the adult soleus revealed no changes in the expression of T3-responsive genes, except for an about 18% increase in MYOD-D2KO SOL Myh7 mRNA. CONCLUSION: Two mouse models of early developmental disruption of Dio2 in myocyte precursor exhibit no significant SKM phenotype.


Iodide Peroxidase/genetics , Muscle Development/genetics , Muscle, Skeletal/growth & development , Myoblasts/metabolism , RNA, Messenger/metabolism , Triiodothyronine/metabolism , Animals , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , MyoD Protein/genetics , Myogenic Regulatory Factor 5/genetics , Myosin Heavy Chains/genetics , Phenotype , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Signal Transduction , Iodothyronine Deiodinase Type II
18.
Endocrinology ; 157(9): 3682-95, 2016 09.
Article En | MEDLINE | ID: mdl-27501182

Millions of levothyroxine-treated hypothyroid patients complain of impaired cognition despite normal TSH serum levels. This could reflect abnormalities in the type 2 deiodinase (D2)-mediated T4-to-T3 conversion, given their much greater dependence on the D2 pathway for T3 production. T3 normally reaches the brain directly from the circulation or is produced locally by D2 in astrocytes. Here we report that mice with astrocyte-specific Dio2 inactivation (Astro-D2KO) have normal serum T3 but exhibit anxiety-depression-like behavior as found in open field and elevated plus maze studies and when tested for depression using the tail-suspension and the forced-swimming tests. Remarkably, 4 weeks of daily treadmill exercise sessions eliminated this phenotype. Microarray gene expression profiling of the Astro-D2KO hippocampi identified an enrichment of three gene sets related to inflammation and impoverishment of three gene sets related to mitochondrial function and response to oxidative stress. Despite normal neurogenesis, the Astro-D2KO hippocampi exhibited decreased expression of four of six known to be positively regulated genes by T3, ie, Mbp (∼43%), Mag (∼34%), Hr (∼49%), and Aldh1a1 (∼61%) and increased expression of 3 of 12 genes negatively regulated by T3, ie, Dgkg (∼17%), Syce2 (∼26%), and Col6a1 (∼3-fold) by quantitative real-time PCR. Notably, in Astro-D2KO animals, there was also a reduction in mRNA levels of genes known to be affected in classical animal models of depression, ie, Bdnf (∼18%), Ntf3 (∼43%), Nmdar (∼26%), and GR (∼20%), which were also normalized by daily exercise sessions. These findings suggest that defects in Dio2 expression in the brain could result in mood and behavioral disorders.


Anxiety/enzymology , Astrocytes/enzymology , Depression/enzymology , Hippocampus/metabolism , Iodide Peroxidase/deficiency , Animals , Gene Expression , Hindlimb Suspension , Iodide Peroxidase/genetics , Male , Mice, Knockout , Physical Conditioning, Animal/physiology , Transcriptome , Triiodothyronine/blood , Iodothyronine Deiodinase Type II
19.
Proc Natl Acad Sci U S A ; 112(45): 14018-23, 2015 Nov 10.
Article En | MEDLINE | ID: mdl-26508642

Thyroid hormone binds to nuclear receptors and regulates gene transcription. Here we report that in mice, at around the first day of life, there is a transient surge in hepatocyte type 2 deiodinase (D2) that activates the prohormone thyroxine to the active hormone triiodothyronine, modifying the expression of ∼165 genes involved in broad aspects of hepatocyte function, including lipid metabolism. Hepatocyte-specific D2 inactivation (ALB-D2KO) is followed by a delay in neonatal expression of key lipid-related genes and a persistent reduction in peroxisome proliferator-activated receptor-γ expression. Notably, the absence of a neonatal D2 peak significantly modifies the baseline and long-term hepatic transcriptional response to a high-fat diet (HFD). Overall, changes in the expression of approximately 400 genes represent the HFD response in control animals toward the synthesis of fatty acids and triglycerides, whereas in ALB-D2KO animals, the response is limited to a very different set of only approximately 200 genes associated with reverse cholesterol transport and lipase activity. A whole genome methylation profile coupled to multiple analytical platforms indicate that 10-20% of these differences can be related to the presence of differentially methylated local regions mapped to sites of active/suppressed chromatin, thus qualifying as epigenetic modifications occurring as a result of neonatal D2 inactivation. The resulting phenotype of the adult ALB-D2KO mouse is dramatic, with greatly reduced susceptibility to diet-induced steatosis, hypertriglyceridemia, and obesity.


Disease Susceptibility/enzymology , Fatty Liver/enzymology , Gene Expression Regulation, Developmental/genetics , Hepatocytes/metabolism , Iodide Peroxidase/metabolism , Obesity/enzymology , Analysis of Variance , Animals , Animals, Newborn , Calorimetry, Indirect , DNA Methylation , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Gene Expression Profiling , In Situ Hybridization , Mice , Mice, Knockout , Microarray Analysis , Obesity/etiology , Triiodothyronine/blood
20.
Endocrinology ; 156(10): 3842-52, 2015 Oct.
Article En | MEDLINE | ID: mdl-26214036

The type 2 deiodinase (D2) activates the prohormone T4 to T3. D2 is expressed in skeletal muscle (SKM), and its global inactivation (GLOB-D2KO mice) reportedly leads to skeletal muscle hypothyroidism and impaired differentiation. Here floxed Dio2 mice were crossed with mice expressing Cre-recombinase under the myosin light chain 1f (cre-MLC) to disrupt D2 expression in the late developmental stages of skeletal myocytes (SKM-D2KO). This led to a loss of approximately 50% in D2 activity in neonatal and adult SKM-D2KO skeletal muscle and about 75% in isolated SKM-D2KO myocytes. To test the impact of Dio2 disruption, we measured soleus T3 content and found it to be normal. We also looked at the expression of T3-responsive genes in skeletal muscle, ie, myosin heavy chain I, α-actin, myosin light chain, tropomyosin, and serca 1 and 2, which was preserved in neonatal SKM-D2KO hindlimb muscles, at a time that coincides with a peak of D2 activity in control animals. In adult soleus the baseline level of D2 activity was about 6-fold lower, and in the SKM-D2KO soleus, the expression of only one of five T3-responsive genes was reduced. Despite this, adult SKM-D2KO animals performed indistinguishably from controls on a treadmill test, running for approximately 16 minutes and reached a speed of about 23 m/min; muscle strength was about 0.3 mN/m·g body weight in SKM-D2KO and control ankle muscles. In conclusion, there are multiple sources of D2 in the mouse SKM, and its role is limited in postnatal skeletal muscle fibers.


Iodide Peroxidase/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Signal Transduction , Thyroid Hormones/metabolism , Adipose Tissue, Brown/metabolism , Animals , Animals, Newborn , Cells, Cultured , Gene Expression , Iodide Peroxidase/genetics , Male , Mice, Knockout , Mice, Transgenic , Muscle Strength/genetics , Muscle Strength/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Myosin Heavy Chains/genetics , Physical Conditioning, Animal/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Thyroxine/metabolism , Time Factors , Triiodothyronine/metabolism , Tropomyosin/genetics , Iodothyronine Deiodinase Type II
...