Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
Add more filters










Publication year range
1.
Endocr Res ; 44(4): 148-152, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30943372

ABSTRACT

Background: Reverse T3 (rT3; 3,3',5'-triiodo-L-thyronine) is widely regarded as an inactive naturally occurring analog of thyroid hormone. rT3 is known to bind to the thyroid hormone analog receptor on plasma membrane integrin αvß3. This integrin is generously expressed by tumor cells and is the initiation site for the stimulation by L-thyroxine (T4) at physiological free concentrations on cancer cell proliferation. Results: In the present studies, we show that rT3 caused increases of proliferation in vitro of 50% to 80% (P < 0.05-0.001) of human breast cancer and glioblastoma cells. Conclusion: rT3 may be a host factor supporting cancer growth.


Subject(s)
Cell Proliferation/drug effects , Neoplasms/pathology , Triiodothyronine, Reverse/pharmacology , Adenocarcinoma/pathology , Brain Neoplasms/pathology , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Female , Glioblastoma/pathology , Humans , MCF-7 Cells , Tumor Cells, Cultured
2.
Biochem Biophys Res Commun ; 506(3): 597-603, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30366665

ABSTRACT

Hypothyroidism has been associated with better recovery from cerebral ischemia-reperfusion (IR) injury in humans. However, any therapeutic advantage of inducing hypothyroidism for mitigating IR injury without invoking the adverse effect of whole body hypothyroidism remains a challenge. We hypothesize that a deiodinase II (D2) inhibitor reverse triiodothyronine (rT3) may render brain specific hypometabolic state to ensue reduced damage during an acute phase of cerebral ischemia without affecting circulating thyroid hormone levels. Preclinical efficacy of rT3 as a neuroprotective agent was determined in rat model of middle cerebral artery occlusion (MCAO) induced cerebral IR and in oxygen glucose deprivation/reoxygenation (OGD/R) model in vitro. rT3 administration in rats significantly reduced neuronal injury markers, infarct size and neurological deficit upon ischemic insult. Similarly, rT3 increased cellular survival in primary cerebral neurons under OGD/R stress. Based on our results from both in vivo as well as in vitro models of ischemia reperfusion injury we propose rT3 as a novel therapeutic agent in reducing neuronal damage and improving stroke outcome.


Subject(s)
Reperfusion Injury/drug therapy , Triiodothyronine, Reverse/therapeutic use , Animals , Blood Pressure/drug effects , Cells, Cultured , Glucose/deficiency , Heart Rate/drug effects , Iodide Peroxidase/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Protective Agents/pharmacology , Protective Agents/therapeutic use , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , Reperfusion Injury/prevention & control , Triiodothyronine, Reverse/pharmacology
3.
Mol Cell Endocrinol ; 470: 281-294, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29155306

ABSTRACT

In the present study we provide evidence that 3,3',5'-triiodothyronine (reverse T3, rT3) restores neurochemical parameters induced by congenital hypothyroidism in rat hippocampus. Congenital hypothyroidism was induced by adding 0.05% propylthiouracil in the drinking water from gestation day 8 and continually up to lactation day 15. In the in vivo rT3 exposure, hypothyroid 12-day old pups were daily injected with rT3 (50 ng/kg body weight) or saline until day 14. In the ex vivo rT3 treatment, hippocampal slices from 15-day-old hypothyroid pups were incubated for 30 min with or without rT3 (1 nM). We found that ex vivo and/or in vivo exposure to rT3 failed in restoring the decreased 14C-glutamate uptake; however, restored the phosphorylation of glial fibrillary acidic protein (GFAP), 45Ca2+ influx, aspartate transaminase (AST), glutamine synthetase (GS) and gamma-glutamate transferase (GGT) activities, as well as glutathione (GSH) levels in hypothyroid hippocampus. In addition, rT3 improved 14C-2-deoxy-D-glucose uptake and lactate dehydrogenase (LDH) activity. Receptor agonists/antagonists (RGD peptide and AP-5), kinase inhibitors of p38MAPK, ERK1/2, CaMKII, PKA (SB239063, PD98059, KN93 and H89, respectively), L-type voltage-dependent calcium channel blocker (nifedipine) and intracellular calcium chelator (BAPTA-AM) were used to determine the mechanisms of the nongenomic rT3 action on GGT activity. Using molecular docking analysis, we found rT3 interaction with αvß3 integrin receptors, nongenomically activating signaling pathways (PKA, CaMKII, p38MAPK) that restored GGT activity. We provide evidence that rT3 is an active TH metabolite and our results represent an important contribution to elucidate the nonclassical mechanism of action of this metabolite in hypothyroidism.


Subject(s)
Hippocampus/enzymology , Hypothyroidism/enzymology , Integrin alphaVbeta3/metabolism , Signal Transduction , Triiodothyronine, Reverse/pharmacology , Animals , Biological Transport/drug effects , Calcium/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/metabolism , Glutathione/metabolism , Homeostasis/drug effects , Hypothyroidism/pathology , L-Lactate Dehydrogenase/metabolism , Models, Biological , Molecular Docking Simulation , Phosphorylation/drug effects , Rats, Wistar , Receptors, Glutamate/metabolism , Signal Transduction/drug effects , Transaminases/metabolism
4.
Neurosci Lett ; 588: 36-41, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25549541

ABSTRACT

The extent of myelination on the axon promotes transmission of impulses in the neural network, any disturbances in this process results in the neurodegenerative condition. Transplantation of oligodendrocyte precursors that supports in the regeneration of axons through myelination is an important step in the restoration of damaged neurons. Therefore, in the present study, the differentiation of human CD34+ stem cells into oligodendrocytes was carried out. The pure human CD34+ culture developed from the stem cells obtained from a peripheral blood of a donor were subjected to oligodendrocyte differentiation medium (ODM). The ODM at a concentration of 40ng/ml thyroxine, 40ng/ml 3,3',5-tri-iodo-thyronine showed distinct morphological changes from day 6 to 9 with cells exhibiting conspicuous stellate morphology and extensive foot processes. The real-time PCR analysis showed prominent expression of Olig2, CNPase, PDGFRα and PLP1/DM20 in the differentiated cells confirming the formed cells are oligodendrocyte precursors. The expression of these genes increased from days 6 to 9 corresponding to the morphological changes observed with almost no expression of GFAP+ cells. The distinct CNPase activity was observed in these differentiated cells compared to normal CD34+ stem cells correlating with results of real-time PCR conclusively explains the development of oligodendrocytes from human CD34+ stem cells.


Subject(s)
Antigens, CD34/metabolism , Cell Transdifferentiation , Neural Stem Cells/drug effects , Oligodendroglia/drug effects , Thyroxine/pharmacology , Triiodothyronine, Reverse/pharmacology , 2',3'-Cyclic-Nucleotide Phosphodiesterases/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Culture Media , Humans , Male , Myelin Proteolipid Protein/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/cytology , Oligodendroglia/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism
5.
PLoS One ; 8(10): e77176, 2013.
Article in English | MEDLINE | ID: mdl-24130850

ABSTRACT

There is increasing experimental evidence of the nongenomic action of thyroid hormones mediated by receptors located in the plasma membrane or inside cells. The aim of this work was to characterize the reverse T3 (rT3) action on calcium uptake and its involvement in immature rat Sertoli cell secretion. The results presented herein show that very low concentrations of rT3 are able to increase calcium uptake after 1 min of exposure. The implication of T-type voltage-dependent calcium channels and chloride channels in the effect of rT3 was evidenced using flunarizine and 9-anthracene, respectively. Also, the rT3-induced calcium uptake was blocked in the presence of the RGD peptide (an inhibitor of integrin-ligand interactions). Therefore, our findings suggest that calcium uptake stimulated by rT3 may be mediated by integrin αvß3. In addition, it was demonstrated that calcium uptake stimulated by rT3 is PKC and ERK-dependent. Furthermore, the outcomes indicate that rT3 also stimulates cellular secretion since the cells manifested a loss of fluorescence after 4 min incubation, indicating an exocytic quinacrine release that seems to be mediated by the integrin receptor. These findings indicate that rT3 modulates the calcium entry and cellular secretion, which might play a role in the regulation of a plethora of intracellular processes involved in male reproductive physiology.


Subject(s)
Calcium/metabolism , Exocytosis/drug effects , Integrins/metabolism , Sertoli Cells/cytology , Sertoli Cells/metabolism , Triiodothyronine, Reverse/pharmacology , Animals , Biological Transport/drug effects , Calcium Channels/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Chloride Channels/metabolism , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase C/metabolism , Rats , Rats, Wistar , Sertoli Cells/drug effects , Time Factors , Tumor Protein, Translationally-Controlled 1
6.
Clin Exp Dermatol ; 37(8): 850-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23083069

ABSTRACT

BACKGROUND: Different elements of the hypothalamic-pituitary-thyroid axis have been found to be implicated in the normal physiology of the human skin. Their effects on wound healing and hair growth in rats have been described previously. There is close homology between the thyroid hormone receptors in humans and guinea pigs. AIM: To assess the effect of different doses of topical 3,3',5-triiodo-L-thyronine (T3) and recombinant human thyroid-stimulating hormone (TSH) on wound healing in guinea pigs. METHODS: Wounds were dressed every other day for 7 days, during which clinical measurements of the wounded areas were performed. Histological examination was performed at the end of the study. RESULTS: Application of high and low concentrations of topical T3 but not TSH demonstrated a significant dose-dependent reduction in the wound surface area through a process of contraction. The main significant histological result was an increase in the hair-follicle count. CONCLUSION: Topical T3 enhances wound healing in guinea pigs, primarily by wound contraction. As this is a critical stage in healing of chronic ulcers, topical T3 could be a useful treatment for wounds.


Subject(s)
Dermatologic Agents/pharmacology , Thyrotropin/pharmacology , Triiodothyronine, Reverse/pharmacology , Wound Healing/drug effects , Wounds and Injuries/drug therapy , Administration, Topical , Analysis of Variance , Animals , Dermatologic Agents/therapeutic use , Dose-Response Relationship, Drug , Guinea Pigs , Hair Follicle/drug effects , Male , Prospective Studies , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thyrotropin/therapeutic use , Triiodothyronine, Reverse/therapeutic use
7.
Steroids ; 77(6): 589-95, 2012 May.
Article in English | MEDLINE | ID: mdl-22366194

ABSTRACT

Thyroid hormones (THs) have a wide variety of essential roles in vertebrates, ranging from the regulation of key metabolic processes to cell proliferation and apoptosis. The classical mechanism of action of THs is genomic; 3,5,3'-triiodothyronine (T3) binds to specific nuclear receptors (TRs) and modifies the expression of specific genes. Recently, a new category of mechanisms, termed nongenomic, has been discovered for T3. These mechanisms include, among others, the rapid activation of signal transduction pathways, such as PI3K/Akt and MAPK, which eventually lead to cell proliferation. These effects are mediated in some cell types by a plasma membrane receptor, identified as integrin αvß3, and in other cell types by cytoplasmic TRß1. The aim of this work was to analyze the effect of T3 on the cell growth of chick embryo hepatocytes at two different stages of development, 14 and 19 days, and to determine the activation of the signal transduction pathways, focusing on the potential involvement of a plasma membrane receptor and the possible participation of PI3K/Akt and reactive oxygen species (ROS). Our results clearly show that T3 stimulates cell proliferation at both stages of development through the activation of the PI3K/Akt pathway and the production of small amounts of ROS, which operate as effective second messengers. Moreover, we prove that these effects are not initiated at the plasma membrane receptor for T3.


Subject(s)
Hepatocytes/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Triiodothyronine, Reverse/pharmacology , Animals , Cell Proliferation/drug effects , Chick Embryo , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Hepatocytes/cytology , Hepatocytes/enzymology , Hepatocytes/metabolism , NADPH Oxidases/metabolism
8.
Am J Physiol Endocrinol Metab ; 301(5): E818-24, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21771965

ABSTRACT

The FoxO3-dependent increase in type II deiodinase (D2), which converts the prohormone thyroxine (T(4)) to 3,5,3'-triiodothyronine (T(3)), is required for normal mouse skeletal muscle differentiation and regeneration. This implies a requirement for an increase in D2-generated intracellular T(3) under these conditions, which has not been directly demonstrated despite the presence of D2 activity in skeletal muscle. We directly show that D2-mediated T(4)-to-T(3) conversion increases during differentiation in C(2)C(12) myoblast and primary cultures of mouse neonatal skeletal muscle precursor cells, and that blockade of D2 eliminates this. In adult mice given (125)I-T(4) and (131)I-T(3), the intracellular (125)I-T(3)/(131)I-T(3) ratio is significantly higher than in serum in both the D2-expressing cerebral cortex and the skeletal muscle of wild-type, but not D2KO, mice. In D1-expressing liver and kidney, the (125)I-T(3)/(131)I-T(3) ratio does not differ from that in serum. Hypothyroidism increases D2 activity, and in agreement with this, the difference in (125)I-T(3)/(131)I-T(3) ratio is increased further in hypothyroid wild-type mice but not altered in the D2KO. Notably, in wild-type but not in D2KO mice, the muscle production of (125)I-T(3) is doubled after skeletal muscle injury. Thus, D2-mediated T(4)-to-T(3) conversion generates significant intracellular T(3) in normal mouse skeletal muscle, with the increased T(3) required for muscle regeneration being provided by increased D2 synthesis, not by T(3) from the circulation.


Subject(s)
Iodide Peroxidase/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Regeneration , Triiodothyronine/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/chemistry , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Iodine Radioisotopes/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts/chemistry , Myoblasts/drug effects , Myoblasts/metabolism , Regeneration/physiology , Triiodothyronine, Reverse/pharmacology , Iodothyronine Deiodinase Type II
9.
PLoS One ; 6(6): e20861, 2011.
Article in English | MEDLINE | ID: mdl-21687737

ABSTRACT

BACKGROUND: Thyroid hormones regulate cell proliferation, differentiation as well as apoptosis. However molecular mechanism underlying apoptosis as a result of thyroid hormone signaling is poorly understood. The antiapoptotic role of Senescence Marker Protein-30 (SMP30) has been characterized in response to varieties of stimuli as well as in knock out model. Our earlier data suggest that thyroid hormone 3, 3'5 Triiodo L Thyronine (T(3)), represses SMP30 in rat liver. METHODOLOGY/PRINCIPAL FINDINGS: In highly metastatic MCF-7, human breast cancer cell line T3 treatment repressed SMP30 expression leading to enhanced apoptosis. Analysis by flow cytometry and other techniques revealed that overexpression and silencing of SMP30 in MCF-7 resulted in decelerated and accelerated apoptosis respectively. In order to identify the cis-acting elements involved in this regulation, we have analyzed hormone responsiveness of transiently transfected hSMP30 promoter deletion reporter vectors in MCF-7 cells. As opposed to the expected epigenetic outcome, thyroid hormone down regulated hSMP30 promoter activity despite enhanced recruitment of acetylated H3 on thyroid response elements (TREs). From the stand point of established epigenetic concept we have categorised these two TREs as negative response elements. Our attempt of siRNA mediated silencing of TRß, reduced the fold of repression of SMP30 gene expression. In presence of thyroid hormone, Trichostatin- A (TSA), which is a Histone deacetylase (HDAC) inhibitor further inhibited SMP30 promoter activity. The above findings are in support of categorisation of both the thyroid response element as negative response elements as usually TSA should have reversed the repressions. CONCLUSION: This is the first report of novel mechanistic insights into the remarkable downregulation of SMP30 gene expression by thyroid hormone which in turn induces apoptosis in MCF-7 human breast cancer cells. We believe that our study represents a good ground for future effort to develop new therapeutic approaches to challenge the progression of breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Calcium-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Response Elements/genetics , Thyroid Gland/metabolism , Triiodothyronine, Reverse/pharmacology , Apoptosis/genetics , Binding Sites , Breast Neoplasms/drug therapy , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Histone Acetyltransferases/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Neoplasm Metastasis , Promoter Regions, Genetic/genetics , Selective Estrogen Receptor Modulators/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Selective Estrogen Receptor Modulators/therapeutic use , Thyroid Gland/drug effects , Thyroid Hormone Receptors beta/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects
10.
Toxicol Sci ; 124(2): 339-47, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21565810

ABSTRACT

Halogenated contaminants, particularly brominated flame retardants, disrupt circulating levels of thyroid hormones (THs), potentially affecting growth and development. Disruption may be mediated by impacts on deiodinase (DI) activity, which regulate the levels of active hormones available to bind to nuclear receptors. The goal of this study was to develop a mass spectrometry-based method for measuring the activity of DIs in human liver microsomes and to examine the effect of halogenated phenolic contaminants on DI activity. Thyroxine (T4) and reverse triiodothyronine (rT3) deiodination kinetics were measured by incubating pooled human liver microsomes with T4 or rT3 and monitoring the production of T3, rT3, 3,3'-diiodothyronine, and 3-monoiodothyronine by liquid chromatography tandem mass spectrometry. Using this method, we examined the effects of several halogenated contaminants, including 2,2',4,4',5-pentabromodiphenyl ether (BDE 99), several hydroxylated polybrominated diphenyl ethers (OH-BDEs), tribromophenol, tetrabromobisphenol A, and triclosan, on DI activity. The Michaelis constants (K(M)) of rT3 and T4 deiodination were determined to be 3.2 ± 0.7 and 17.3 ± 2.3µM. The V(max) was 160 ± 5.8 and 2.8 ± 0.10 pmol/min.mg protein, respectively. All studied contaminants inhibited DI activity in a dose-response manner, with the exception of BDE 99 and two OH-BDEs. 5'-Hydroxy 2,2',4,4',5-pentabromodiphenyl ether was found to be the most potent inhibitor of DI activity, and phenolic structures containing iodine were generally more potent inhibitors of DI activity relative to brominated, chlorinated, and fluorinated analogues. This study suggests that some halogenated phenolics, including current use compounds such as plastic monomers, flame retardants, and their metabolites, may disrupt TH homeostasis through the inhibition of DI activity in vivo.


Subject(s)
Endocrine Disruptors/toxicity , Halogenated Diphenyl Ethers/toxicity , Iodide Peroxidase/metabolism , Microsomes, Liver/drug effects , Polybrominated Biphenyls/toxicity , Chromatography, Liquid , Diiodothyronines/metabolism , Endocrine Disruptors/chemistry , Halogenated Diphenyl Ethers/chemistry , Humans , In Vitro Techniques , Microsomes, Liver/enzymology , Polybrominated Biphenyls/chemistry , Tandem Mass Spectrometry , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroxine/metabolism , Thyroxine/pharmacology , Triiodothyronine, Reverse/metabolism , Triiodothyronine, Reverse/pharmacology
11.
J Endocrinol ; 210(1): 59-69, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21508094

ABSTRACT

Iodothyronines influence lipid metabolism and energy homeostasis. Previous studies demonstrated that 3,5-l-diiodothyronine (T(2)), as well as 3,3',5-L-triiodothyronine (T(3)), was able to both prevent and reverse hepatic steatosis in rats fed a high-fat diet, and this effect depends on a direct action of iodothyronines on the hepatocyte. However, the involvement of thyroid hormone receptors (TRs) in mediating the lipid-lowering effect of iodothyronines was not elucidated. In this study, we investigated the ability of T(2) and T(3) to reduce the lipid overloading using the rat hepatoma FaO cells defective for functional TRs. The absence of constitutive mRNA expression of both TRα1 and TRß1 in FaO cells was verified by RT-qPCR. To mimic the fatty liver condition, FaO cells were treated with a fatty acid mixture and then exposed to pharmacological doses of T(2) or T(3) for 24 h. Lipid accumulation, mRNA expression of the peroxisome proliferator-activated receptors (PPAR-α, -γ, -δ) the acyl-CoA oxidase (AOX), and the stearoyl CoA desaturase (SCD1), as well as fuel-stimulated O(2) consumption in intact cells, were evaluated. Lipid accumulation was associated with an increase in triacylglycerol content, PPARγ mRNA expression, and a decrease in PPARδ and SCD1 mRNA expression. The addition of T(2) or T(3) to lipid-overloaded cells resulted in i) reduction in lipid content; ii) downregulation of PPARα, PPARγ, and AOX expression; iii) increase in PPARδ expression; and iv) stimulation of mitochondrial uncoupling. These data demonstrate, for the first time, that in the hepatocyte, the lipid-lowering actions of both T(2) and T(3) are not mediated by TRs.


Subject(s)
Diiodothyronines/pharmacology , Fatty Liver/drug therapy , Hepatocytes/drug effects , Hypolipidemic Agents/pharmacology , Triiodothyronine, Reverse/pharmacology , Uncoupling Agents/pharmacology , Aldehyde Oxidase/genetics , Aldehyde Oxidase/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver Neoplasms/metabolism , PPAR delta/genetics , PPAR delta/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism
12.
Arch Biochem Biophys ; 503(1): 129-36, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20599658

ABSTRACT

The hypothalamic-pituitary-thyroid axis plays a key role in skeletal development, acquisition of peak bone mass and regulation of adult bone turnover. Euthyroid status is essential for maintenance of optimal bone mineralization and strength. In population studies, hypothyroidism and hyperthyroidism have both been associated with an increased risk of fracture. Furthermore, recent studies in healthy euthyroid post-menopausal women indicate that thyroid status in the upper normal range is also associated with low bone mineral density and an increased risk of non-vertebral fracture. Studies in mutant mice have demonstrated that thyroid hormone receptor α is the major mediator of T3 action in bone and that thyroid hormones exert anabolic actions during growth but have catabolic effects on the adult skeleton. Nevertheless, TSH has also been proposed to be a direct negative regulator of bone turnover, although the relative importance of T3 and TSH actions in the skeleton has yet to be clarified.


Subject(s)
Bone and Bones , Thyroid Gland , Animals , Bone Remodeling/drug effects , Bone and Bones/cytology , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/physiology , Humans , Thyroid Diseases/drug therapy , Thyroid Diseases/metabolism , Thyroid Diseases/physiopathology , Thyroid Gland/drug effects , Thyroid Gland/physiopathology , Thyrotropin/metabolism , Thyrotropin/pharmacology , Triiodothyronine, Reverse/metabolism , Triiodothyronine, Reverse/pharmacology
13.
Article in English | MEDLINE | ID: mdl-16820313

ABSTRACT

We previously isolated cDNAs encoding conger eel (Conger myriaster) thyroid hormone (TH) receptors (TRs). In the present study, we investigated the transactivation activities of conger eel TRs treated with THs (3,3',5-triiodo-l-thyronine [T3], l-thyroxine [T4], and 3,3',5'-triiodo-l-thyronine [rT3]), or ligands and activators of other nuclear receptors. Following transient transfection into the Japanese eel (Anguilla japonica) hepatocyte cell line Hepa-E1, the conger eel TRs (cTRs) showed TH-dependent activation of transcription from a TH-responsive promoter. However, no transactivation activity of any of the four cTRs was observed with ligands or activators of other nuclear receptors. Although T3 activation for cTRs is stronger than other THs (T3>T4>rT3), the transactivation sensitivity was different from the activity of cTRs with THs, respectively. Therefore, we conclude that cTRs can act in concert with THs in fish metamorphosis.


Subject(s)
Eels/metabolism , Receptors, Thyroid Hormone/metabolism , Thyroid Hormones/pharmacology , Transcriptional Activation , Animals , Cell Line , Hepatocytes/cytology , Hepatocytes/metabolism , Liver/metabolism , Receptors, Thyroid Hormone/agonists , Receptors, Thyroid Hormone/genetics , Thyroxine/pharmacology , Triiodothyronine, Reverse/pharmacology
14.
Thyroid ; 16(6): 521-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16839253

ABSTRACT

3,5,3' Triiodo-L-thyronine (T3) stimulated the uptake of 2-deoxy-D-glucose (2-DOG) into L6 cells, nongenomically, starting at subpicomolar concentrations and reaching a peak at concentrations of 1-10 nM. Stimulation at the peak was usually approximately 250%. The uptake of 2-DOG declined with higher concentrations of T(3). The dose-response curve of insulin is similar in shape to that of T(3), and its peak stimulation can even reach 600% over the control. Wortmannin, an inhibitor of the PI-3k, completely inhibited the stimulation of 2-DOG uptake by T(3), with no effect on the control cells. L6 cells exposed for 10 minutes to T3 resulted in a 200%-300% stimulation of PI-3k, as measured by the production of labeled (32)P-PI-3P. Similar results were obtained with insulin. After incubation for 5 minutes with L6 cells, T(3) increased phosphorylation of the insulin receptor beta subunit; this correlated significantly with the degree of stimulation of 2-DOG uptake at 90 minutes (r = 0.89, p

Subject(s)
Deoxyglucose/metabolism , Muscle, Skeletal/cytology , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Insulin/metabolism , Triiodothyronine, Reverse/pharmacology , Animals , Biological Transport , Cell Proliferation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Insulin/metabolism , Muscle, Skeletal/metabolism , Phosphorylation , Rats , Time Factors
15.
J Med Chem ; 49(1): 23-6, 2006 Jan 12.
Article in English | MEDLINE | ID: mdl-16392786

ABSTRACT

Steered molecular dynamics simulations of ligand dissociation from Thyroid hormone receptors indicate that dissociation is favored via rearrangements in a mobile part of the LBD comprising H3, the loop between H1 and H2, and nearby beta-sheets, contrary to current models in which the H12 is mostly involved. Dissociation is facilitated in this path by the interaction of the hydrophilic part of the ligand with external water molecules, suggesting strategies to enhance ligand binding affinity.


Subject(s)
Models, Chemical , Receptors, Thyroid Hormone/chemistry , Triiodothyronine, Reverse/chemistry , Triiodothyronine/analogs & derivatives , Computer Simulation , Ligands , Molecular Structure , Receptors, Thyroid Hormone/drug effects , Structure-Activity Relationship , Time Factors , Triiodothyronine/chemistry , Triiodothyronine/pharmacology , Triiodothyronine, Reverse/pharmacology
16.
J Vet Med A Physiol Pathol Clin Med ; 52(9): 429-35, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16268952

ABSTRACT

Previously, it has been observed that dexamethasone or adrenaline-induced hyperlipaemia in blood of chicken was significantly reduced after administration of reverse triiodothyronine (rT3). The present experiment was performed on chicken to determine the altered circulating non-esterified fatty acids (NEFA) induced by physiologically enhanced endogenous corticosterone and catecholamines may also be influenced by rT3. Rise of both hormones were induced by insulin administration. Changes in circulating glucose, corticosterone and catecholamines were additionally measured. Following insulin injection blood glucose fell on the average by 32.7% below control at 2 h of the experiment. Additional treatment with rT3 (rT3 + insulin group) gradually attenuated this decrease and at 4 and 6 h of the experiment it was 17.1% and 12.9% below control, respectively, suggesting on slight inhibition by rT3 of insulin-stimulated glucose utilization. Exposure to insulin significantly increased NEFA levels to about 670% above control group. Additional treatment with rT3 reduced this increase to 309% of control, suggesting inhibition of lipolysis by rT3. Similar alterations were observed in plasma corticosterone levels. Insulin treatment peaked the corticosterone levels maximally by 507.6% above control. Additional treatment with rT3 abolished this rise in the averages to 194.2% above control, possibly by interaction of rT3 with hypothalamo-adrenal axis. Insulin injection increased plasma catecholamines on the average by 21.5% and 53.4% for adrenaline and noradrenaline respectively. Supplementary treatment with rT3 intensified this rise by 55.6% and 71.6% respectively. The obtained results suggest on inhibitory effect of rT3 on hypoglycaemia, hyperlipaemia and plasma corticosterone concentrations in chickens treated with insulin. Contrary to this, rT3 enhanced the rise of plasma catecholamines due to insulin treatment. The obtained data favour the assumption that hypometabolic properties of rT3 depends mainly upon reduced supply of NEFA as a result of restricted lipolysis and to a lesser extent upon the supply of glucose.


Subject(s)
Chickens/physiology , Corticosterone/blood , Fatty Acids, Nonesterified/blood , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Triiodothyronine, Reverse/pharmacology , Animals , Blood Glucose/analysis , Blood Glucose/drug effects , Chickens/blood , Hypoglycemia/blood , Hypoglycemia/chemically induced , Hypoglycemia/veterinary
17.
Eur J Endocrinol ; 153(3): 429-34, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16131606

ABSTRACT

OBJECTIVES: Intrapituitary triiodothyronine (T3) production plays a pivotal role in the control of TSH secretion. Its production is increased in the presence of decreased serum thyroxine (T4) concentrations and the enzyme responsible, deiodinase type 2 (D2), is highest in hypothyroidism. In order to document the role of this enzyme in adult rats we developed an experimental model that inhibited this enzyme using the specific inhibitor, reverse T3 (rT3). METHODS: Hypothyroidism was induced with propylthiouracil (PTU; 0.025 g/l in drinking water) which in addition blocked deiodinase type 1 (D1) activity, responsible for the rapid clearance of rT3 in vivo. During the last 7 days of the experiment, the hypothyroid rats were injected (s.c.) for 4 days with 0.4 or 0.8 nmol T4 per 100 g body weight (bw) per day. For the last 3 days, the same amount of T4 was infused via s.c. minipumps. In additional groups, 25 nmol rT3/100 g bw per day were added to the 3-day infusion of T4. RESULTS: Infusion of 0.4 nmol T4/100 g bw per day did not affect the high serum TSH levels, 0.8 nmol T4/100 g bw per day decreased them to 57% of the hypothyroid values. The infusions of rT3 inhibited D2 activity in all organs where it was measured: the pituitary, brain cortex and brown adipose tissue (BAT). In the pituitary, the activity was 27%, to less than 15% of the activity in hypothyroidism. Despite that, serum TSH levels did not increase, serum T4 concentrations did not change and the changes in serum T3 were minimal. CONCLUSIONS: We conclude that in partly hypothyroid rats, a 3-day inhibition of D2 activity, without concomitant change in serum T4 and minimal changes in serum T3 levels, is not able to upregulate TSH secretion and we postulate that this may be a reflection of absent or only minimal changes in circulating T3 concentrations.


Subject(s)
Enzyme Inhibitors/pharmacology , Hypothyroidism/enzymology , Iodide Peroxidase/antagonists & inhibitors , Thyrotropin/metabolism , Thyroxine/metabolism , Triiodothyronine, Reverse/pharmacology , Animals , Hypothyroidism/blood , Iodide Peroxidase/metabolism , Male , Propylthiouracil , Rats , Rats, Wistar , Thyrotropin/antagonists & inhibitors , Thyrotropin/blood , Thyroxine/blood
19.
Diabetologia ; 48(4): 624-33, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15756538

ABSTRACT

AIMS/HYPOTHESIS: The aims of this work were to determine the effect of hypothyroidism on insulin-stimulated glucose turnover and to unravel the potential mechanisms involved in such an effect. METHODS: Hypothyroidism was induced by administration of propylthiouracil, with partial T4 substitution. Euglycaemic-hyperinsulinaemic clamps, associated with the labelled 2-deoxy-D-glucose technique for measuring tissue-specific glucose utilisation, were used. To assess a possible involvement of leptin in the modulation of glucose metabolism by hypothyroidism, leptin was infused intracerebroventricularly for 6 days. A group of leptin-infused rats was treated with rT3 to determine a potential role of T3 in mediating the leptin effects. RESULTS: Compared with euthyroid rats, hypothyroid animals exhibited decreased overall glucose turnover and decreased glucose utilisation indices in skeletal muscle and adipose tissue. Leptinaemia in hypothyroid rats was lower while resistin mRNA expression in adipose tissue was higher than in euthyroid animals. Intracerebroventricular leptin infusion in hypothyroid rats partially restored overall, muscle and adipose tissue insulin-stimulated glucose utilisation and improved the reduced glycaemic response observed during insulin tolerance tests. The leptin effects were due neither to the observed increase in plasma T3 levels nor to changes in the high adipose tissue resistin expression of hypothyroid rats. The administration of leptin to hypothyroid animals was accompanied by increased expression of muscle and adipose tissue carnitine palmitoyl transferases, decreased plasma NEFA levels and reduced muscle triglyceride content. CONCLUSIONS/INTERPRETATION: Hypothyroidism is characterised by decreased insulin responsiveness, partly mediated by an exaggerated glucose-fatty acid cycle that is partly alleviated by intracerebroventricular leptin administration.


Subject(s)
Energy Metabolism/drug effects , Glucose/metabolism , Hyperthyroidism/metabolism , Leptin/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Carnitine O-Palmitoyltransferase/genetics , Fatty Acids, Nonesterified/blood , Gene Expression/genetics , Glucose/pharmacology , Glucose Clamp Technique , Hormones, Ectopic/genetics , Hyperthyroidism/chemically induced , Hyperthyroidism/genetics , Insulin/blood , Insulin/pharmacology , Insulin Resistance/physiology , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Leptin/administration & dosage , Leptin/blood , Male , Muscle, Skeletal/chemistry , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Propylthiouracil , Rats , Rats, Wistar , Resistin , Thyrotropin/blood , Thyroxine/blood , Thyroxine/pharmacology , Triglycerides/analysis , Triglycerides/blood , Triiodothyronine/blood , Triiodothyronine, Reverse/pharmacology , Iodothyronine Deiodinase Type II
20.
Blood ; 105(1): 85-94, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15358620

ABSTRACT

Outgrowth, long-term self-renewal, and terminal maturation of human erythroid progenitors derived from umbilical cord blood in serum-free medium can be modulated by steroid hormones. Homogeneous erythroid cultures, as characterized by flow cytometry and dependence on a specific mixture of physiologic proliferation factors, were obtained within 8 days from a starting population of mature and immature mononuclear cells. Due to previous results in mouse and chicken erythroblasts, the proliferation-promoting effect of glucocorticoids was not unexpected. Surprisingly, however, androgen had a positive effect on the sustained expansion of human female but not male erythroid progenitors. Under optimal conditions, sustained proliferation of erythroid progenitors resulted in a more than 10(9)-fold expansion within 60 days. Terminal erythroid maturation was significantly improved by adding human serum and thyroid hormone (3,5,3'-triiodothyronine [T3]) to the differentiation medium. This resulted in highly synchronous differentiation of the cells toward enucleated erythrocytes within 6 days, accompanied by massive size decrease and hemoglobin accumulation to levels comparable to those in peripheral blood erythrocytes. Thus, obviously, different ligand-activated nuclear hormone receptors massively influence the decision between self-renewal and terminal maturation in the human erythroid compartment.


Subject(s)
Cell Differentiation/drug effects , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Steroids/pharmacology , Androgens/pharmacology , Cell Lineage , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Serum-Free , Dexamethasone/pharmacology , Erythroblasts/cytology , Erythroblasts/drug effects , Erythropoietin/pharmacology , Female , Fetal Blood/cytology , Fetal Blood/drug effects , Humans , Male , Sex Characteristics , Stem Cell Factor/pharmacology , Time Factors , Triiodothyronine, Reverse/pharmacology , Umbilical Cord/cytology , Umbilical Cord/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...