Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
FASEB J ; 34(11): 15480-15491, 2020 11.
Article in English | MEDLINE | ID: mdl-32969079

ABSTRACT

Thyroid hormones are important for homeostatic control of energy metabolism and body temperature. Although skeletal muscle is considered a key site for thyroid action, the contribution of thyroid hormone receptor signaling in muscle to whole-body energy metabolism and body temperature has not been resolved. Here, we show that T3-induced increase in energy expenditure requires thyroid hormone receptor alpha 1 (TRα1 ) in skeletal muscle, but that T3-mediated elevation in body temperature is achieved in the absence of muscle-TRα1 . In slow-twitch soleus muscle, loss-of-function of TRα1 (TRαHSACre ) alters the fiber-type composition toward a more oxidative phenotype. The change in fiber-type composition, however, does not influence the running capacity or motivation to run. RNA-sequencing of soleus muscle from WT mice and TRαHSACre mice revealed differentiated transcriptional regulation of genes associated with muscle thermogenesis, such as sarcolipin and UCP3, providing molecular clues pertaining to the mechanistic underpinnings of TRα1 -linked control of whole-body metabolic rate. Together, this work establishes a fundamental role for skeletal muscle in T3-stimulated increase in whole-body energy expenditure.


Subject(s)
Energy Metabolism/drug effects , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/physiology , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormones/pharmacology , Animals , Male , Mice , Mice, Knockout , Muscle Fibers, Fast-Twitch/cytology , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/cytology , Muscle Fibers, Slow-Twitch/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Physical Conditioning, Animal , Transcriptome
2.
Int J Mol Sci ; 22(5)2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33801253

ABSTRACT

P43 is a truncated form of thyroid hormone receptor α localized in mitochondria, which stimulates mitochondrial respiratory chain activity. Previously, we showed that deletion of p43 led to reduction of pancreatic islet density and a loss of glucose-stimulated insulin secretion in adult mice. The present study was designed to determine whether p43 was involved in the processes of ß cell development and maturation. We used neonatal, juvenile, and adult p43-/- mice, and we analyzed the development of ß cells in the pancreas. Here, we show that p43 deletion affected only slightly ß cell proliferation during the postnatal period. However, we found a dramatic fall in p43-/- mice of MafA expression (V-Maf Avian Musculoaponeurotic Fibrosarcoma Oncogene Homolog A), a key transcription factor of beta-cell maturation. Analysis of the expression of antioxidant enzymes in pancreatic islet and 4-hydroxynonenal (4-HNE) (a specific marker of lipid peroxidation) staining revealed that oxidative stress occurred in mice lacking p43. Lastly, administration of antioxidants cocktail to p43-/- pregnant mice restored a normal islet density but failed to ensure an insulin secretion in response to glucose. Our findings demonstrated that p43 drives the maturation of ß cells via its induction of transcription factor MafA during the critical postnatal window.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Insulin Secretion , Insulin-Secreting Cells/cytology , Maf Transcription Factors, Large/metabolism , Thyroid Hormone Receptors alpha/physiology , Animals , Female , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/genetics , Male , Mice , Mice, Knockout , Oxidative Stress
3.
Int J Mol Sci ; 22(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478016

ABSTRACT

The aim of this study was to assess the prognostic value of the steroid hormone receptor expression, counting the retinoid X receptor (RXR) and thyroid hormone receptors (THRs), on the two different breast cancer (BC) entities: multifocal/multicentric versus unifocal. The overall and disease-free survival were considered as the prognosis determining aspects and analyzed by uni- and multi-variate analysis. Furthermore, histopathological grading and TNM staging (T = tumor size, N = lymph node involvement, M = distant metastasis) were examined in relation to RXR and THRs expression. A retrospective statistical analysis was carried out on survival-related events in a series of 319 sporadic BC patients treated at the Department of Gynecology and Obstetrics at the Ludwig-Maximillian's University in Munich between 2000 and 2002. The expression of RXR and THRs, including its two major isoforms THRα1 and THRα2, was analyzed by immunohistochemistry and showed to have a significant correlation for both BC entities in regard to survival analysis. Patients with multifocal/multicentric BC were exposed to a significantly worse disease-free survival (DFS) when expressing RXR. Patients with unifocal BC showed a significantly worse DFS when expressing THRα1. In contrast, a statistically significant positive association between THRα2 expression and enhanced DFS in multifocal/multicentric BC was shown. Especially the RXR expression in multifocal/multicentric BC was found to play a remarkably contradictory role for BC prognosis. The findings imply the need for a critical review of possible molecular therapies targeting steroid hormone receptors in BC treatment. Our results strengthen the need to further investigate the behavior of the nuclear receptor family, especially in relation to BC focality.


Subject(s)
Breast Neoplasms/diagnosis , Retinoid X Receptors/physiology , Thyroid Hormone Receptors alpha/physiology , Adult , Aged , Biomarkers, Tumor/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cohort Studies , Female , Germany/epidemiology , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Prognosis , Retrospective Studies , Survival Analysis , Tumor Burden
4.
J Vasc Res ; 55(4): 224-234, 2018.
Article in English | MEDLINE | ID: mdl-30092589

ABSTRACT

Thyroid hormone (TH) regulates gene transcription by binding to TH receptors (TRs). TRs regulate the genes of lipid metabolism and the renin-angiotensin system (RAS). We examined the effect of TRα deletion in ApoE-/- mice (DKO mice) on the following: (i) the expression of genes controlling cholesterol metabolism and tissue (t)RAS in the liver and aorta and (ii) the expression of these genes and the regulation of cholesterol content in cultured vascular smooth muscle cells (VSMCs). TRα deletion in ApoE-/- mice led to the repression of genes involved in the synthesis and influx of cholesterol in the liver. However, TRα deletion in the arterial wall suppressed the expression of genes involved in the esterification and excretion of cholesterol and enhanced the expression of angiotensinogen (AGT). The VSMCs of the ApoE-/- and DKO mice increased their cholesterol content during cholesterol loading, but failed to increase the expression of ATP-binding cassette transporter A1 (ABCA1). T3 addition partially corrected these abnormalities in the cells of the ApoE-/- mice but not those of the DKO mice. In conclusion, TRα deletion in ApoE-/- mice slightly increases the expression of tRAS in the aorta and aggravates the dysregulation of cholesterol content in the VSMCs.


Subject(s)
Apolipoproteins E/deficiency , Cholesterol/metabolism , Muscle, Smooth, Vascular/metabolism , Renin-Angiotensin System/physiology , Thyroid Hormone Receptors alpha/deficiency , ATP Binding Cassette Transporter 1/genetics , Animals , Aorta/chemistry , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Atherosclerosis/diagnostic imaging , Cells, Cultured , Cholesterol/administration & dosage , Cholesterol/genetics , Gene Expression , Hybridization, Genetic , Liver/chemistry , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , RNA, Messenger , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/physiology , Triiodothyronine/pharmacology , Ultrasonography
5.
Heart Fail Rev ; 20(3): 273-82, 2015 May.
Article in English | MEDLINE | ID: mdl-25501869

ABSTRACT

Thyroid hormone (TH) appears to have a critical role in cardiac repair after injury beyond its role in development and metabolism homeostasis. This unique action is due to the fact that TH effect on the heart is shown to be differentiated depending on its administration on injured or healthy myocardium. Thus, TH can limit ischemia-reperfusion injury via a fine balance between pro-apoptotic and pro-survival signaling pathways. This response is thyroid hormone receptor (TRα1) dependent. Furthermore, an interaction between stress-induced growth kinase signaling and TRα1 is shown to occur and determine postischemic remodeling and cardiac recovery depending on the availability of TH. This new evidence is consistent with clinical observations showing the cardioprotective effect of TH treatment in cardiac surgery, transplantation and heart failure. TH and/or thyroid analogs may be novel agents in treating heart diseases.


Subject(s)
Heart Failure/therapy , Myocardial Ischemia/drug therapy , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormones/therapeutic use , Cardiac Surgical Procedures , Heart/drug effects , Heart Transplantation , Humans , Myocardium/metabolism , Ventricular Remodeling
6.
Biochim Biophys Acta ; 1830(7): 4004-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23528896

ABSTRACT

BACKGROUND: Thyroid hormone acts via receptor subtypes (TRα1, TRß1, TRß2) with differing tissue distributions, encoded by distinct genes (THRA, THRB). THRB mutations cause a disorder with central (hypothalamic-pituitary) resistance to thyroid hormone action with markedly elevated thyroid hormone and normal TSH levels. SCOPE OF REVIEW: This review describes the clinical features, genetic and molecular pathogenesis of a homologous human disorder mediated by defective THRA. Clinical features include growth retardation, skeletal dysplasia and constipation associated with low-normal T4 and high-normal T3 levels and a low T4/T3 ratio, together with subnormal reverse T3 levels. Heterozygous TRa1 mutations in affected individuals generate defective mutant receptors which inhibit wild-type receptor action in a dominant negative manner. MAJOR CONCLUSIONS: Mutations in human TRα1 mediate RTH with features of hypothyroidism in particular tissues (e.g. skeleton, gastrointestinal tract), but are not associated with a markedly dysregulated pituitary-thyroid axis. GENERAL SIGNIFICANCE: Human THRA mutations could be more common but may have eluded discovery due to the absence of overt thyroid dysfunction. Nevertheless, in the appropriate clinical context, a thyroid biochemical signature (low T4/T3 ratio, subnormal reverse T3 levels), may enable future identification of cases. This article is part of a Special Issue entitled Thyroid hormone signalling.


Subject(s)
Thyroid Hormone Receptors alpha/physiology , Thyroid Hormone Resistance Syndrome/physiopathology , Thyroid Hormones/physiology , Humans , Mutation , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormone Resistance Syndrome/metabolism , Thyroid Hormones/genetics , Thyroid Hormones/metabolism
7.
Biochim Biophys Acta ; 1830(7): 3979-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22634735

ABSTRACT

BACKGROUND: Thyroid hormones regulate skeletal development, acquisition of peak bone mass and adult bone maintenance. Abnormal thyroid status during childhood disrupts bone maturation and linear growth, while in adulthood it results in altered bone remodeling and an increased risk of fracture SCOPE OF REVIEW: This review considers the cellular effects and molecular mechanisms of thyroid hormone action in the skeleton. Human clinical and population data are discussed in relation to the skeletal phenotypes of a series of genetically modified mouse models of disrupted thyroid hormone signaling. MAJOR CONCLUSIONS: Euthyroid status is essential for normal bone development and maintenance. Major thyroid hormone actions in skeletal cells are mediated by thyroid hormone receptor α (TRα) and result in anabolic responses during growth and development but catabolic effects in adulthood. These homeostatic responses to thyroid hormone are locally regulated in individual skeletal cell types by the relative activities of the type 2 and 3 iodothyronine deiodinases, which control the supply of the active thyroid hormone 3,5,3'-L-triiodothyronine (T3) to its receptor. GENERAL SIGNIFICANCE: Population studies indicate that both thyroid hormone deficiency and excess are associated with an increased risk of fracture. Understanding the cellular and molecular basis of T3 action in skeletal cells will lead to the identification of new targets to regulate bone turnover and mineralization in the prevention and treatment of osteoporosis. This article is part of a Special Issue entitled Thyroid hormone signaling.


Subject(s)
Bone Development/physiology , Bone and Bones/physiology , Thyroid Hormones/physiology , Animals , Bone Development/genetics , Bone and Bones/metabolism , Humans , Signal Transduction , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormones/genetics , Thyroid Hormones/metabolism
8.
Clin Calcium ; 24(6): 821-7, 2014 Jun.
Article in Japanese | MEDLINE | ID: mdl-24870832

ABSTRACT

The role of the hypothalamic-pituitary-thyroid axis is important in normal skeletal development, gain of bone mass, and regulation of adult bone metabolism. Hypothyroidism in childhood causes delayed bone maturation and growth disturbance and thyroid dysfunction in adult induces altered bone remodeling and an increased risk of bone fracture. Thyroid hormone actions in skeletal cells are mainly mediated by thyroid hormone receptor α (TRα) . The responses to thyroid hormone are regulated by type 2 and 3 iodothyronine deiodinase (DIO2 and DIO3) , which convert prohormone (T4) to active hormone (T3) . Euthyroid status is necessary for the homeostasis of human bone metabolism.


Subject(s)
Bone Development/physiology , Bone and Bones/metabolism , Thyroid Hormones/physiology , Animals , Bone Density , Bone Diseases, Developmental/etiology , Bone Remodeling , Child , Drug Combinations , Fractures, Bone/etiology , Humans , Hypothyroidism/complications , Iodide Peroxidase/physiology , Mice , Signal Transduction , Thyroid Hormone Receptors alpha/physiology , Thyroxine , Triiodothyronine , Iodothyronine Deiodinase Type II
9.
Dev Biol ; 356(2): 350-8, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21621530

ABSTRACT

Thyroid hormone deficiency is known to deeply affect cerebellum post-natal development. We present here a detailed analysis of the phenotype of a recently generated mouse model, expressing a dominant-negative TRα1 mutation. Although hormonal level is not affected, the cerebellum of these mice displays profound alterations in neuronal and glial differentiation, which are reminiscent of congenital hypothyroidism, indicating a predominant function of this receptor isoform in normal cerebellum development. Some of the observed effects might result from the cell autonomous action of the mutation, while others are more likely to result from a reduction in neurotrophic factor production.


Subject(s)
Cerebellum/growth & development , Mutation , Thyroid Hormone Receptors alpha/physiology , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Congenital Hypothyroidism/etiology , Mice , Mice, Transgenic , Myelin Sheath/physiology , Neuroglia/physiology , Oligodendroglia/cytology , PAX2 Transcription Factor/analysis , Protein Isoforms/genetics , Protein Isoforms/physiology , Purkinje Cells/cytology , Synapses/physiology , Thyroid Hormone Receptors alpha/genetics , gamma-Aminobutyric Acid/physiology
10.
J Biol Chem ; 285(32): 24477-86, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20529852

ABSTRACT

Failure of the functional pancreatic beta-cell mass to expand in response to increased metabolic demand is a hallmark of type 2 diabetes. Lineage tracing studies indicate that replication of existing beta-cells is important for beta-cell proliferation in adult animals. In rat pancreatic beta-cell lines (RIN5F), treatment with 100 nM thyroid hormone (triiodothyronine, T(3)) enhances cell proliferation. This result suggests that T(3) is required for beta-cell proliferation or replication. To identify the role of thyroid hormone receptor alpha (TR(alpha)) in the processes of beta-cell growth and cell cycle regulation, we constructed a recombinant adenovirus vector, AdTR(alpha). Infection with AdTR(alpha) to RIN5F cells increased the expression of cyclin D1 mRNA and protein. Overexpression of the cyclin D1 protein in AdTR(alpha)-infected cells led to activation of the cyclin D1/cyclin-dependent kinase/retinoblastoma protein/E2F pathway, along with cell cycle progression and cell proliferation following treatment with 100 nM T(3). Conversely, lowering cellular cyclin D1 by small interfering RNA knockdown in AdTR(alpha)-infected cells led to down-regulation of the cyclin D1/CDK/Rb/E2F pathway and inhibited cell proliferation. Furthermore, in immunodeficient mice with streptozotocin-induced diabetes, intrapancreatic injection of AdTR(alpha) led to the restoration of islet function and to an increase in the beta-cell mass. These results support the hypothesis that liganded TR(alpha) plays a critical role in beta-cell replication and in expansion of the beta-cell mass during postnatal development. Thus, liganded TR(alpha) may be a target for therapeutic strategies that can induce the expansion and regeneration of beta-cells.


Subject(s)
Insulin-Secreting Cells/cytology , Thyroid Hormone Receptors alpha/physiology , Animals , Cell Line , Cell Nucleus/metabolism , Cell Proliferation , Cyclin D1/metabolism , Gene Expression Regulation , Humans , Ligands , Mice , Mice, SCID , Models, Biological , RNA, Small Interfering/metabolism , Retinoblastoma Protein/metabolism , Thyroid Hormone Receptors alpha/metabolism
11.
EMBO J ; 26(21): 4535-45, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17932484

ABSTRACT

Thyroid hormone, via its nuclear receptors TRalpha and TRbeta, controls metabolism by acting locally in peripheral tissues and centrally by regulating sympathetic signaling. We have defined aporeceptor regulation of metabolism by using mice heterozygous for a mutant TRalpha1 with low affinity to T3. The animals were hypermetabolic, showing strongly reduced fat depots, hyperphagia and resistance to diet-induced obesity accompanied by induction of genes involved in glucose handling and fatty acid metabolism in liver and adipose tissues. Increased lipid mobilization and beta-oxidation occurred in adipose tissues, whereas blockade of sympathetic signaling to brown adipose tissue normalized the metabolic phenotype despite a continued perturbed hormone signaling in this cell type. The results define a novel and important role for the TRalpha1 aporeceptor in governing metabolic homeostasis. Furthermore, the data demonstrate that a nuclear hormone receptor affecting sympathetic signaling can override its autonomous effects in peripheral tissues.


Subject(s)
Thyroid Hormone Receptors alpha/physiology , Adipose Tissue/metabolism , Animals , Fatty Acids/metabolism , Gene Expression Regulation , Glucose/metabolism , Lipids/chemistry , Liver/metabolism , Metabolism , Mice , Mice, Transgenic , Protein Binding , Receptors, Thyroid Hormone/metabolism , Signal Transduction , Sympathetic Nervous System/metabolism , Thyroid Hormone Receptors alpha/metabolism
12.
Sci Rep ; 11(1): 24105, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34916557

ABSTRACT

Thyroid hormone (TH) and thyroid hormone receptor (THR) regulate stem cell proliferation and differentiation during development, as well as during tissue renewal and repair in the adult. THR undergoes posttranslational modification by small ubiquitin-like modifier (SUMO). We generated the THRA (K283Q/K288R)-/- mouse model for in vivo studies and used human primary preadipocytes expressing the THRA sumoylation mutant (K283R/K288R) and isolated preadipocytes from mutant mice for in vitro studies. THRA mutant mice had reduced white adipose stores and reduced adipocyte cell diameter on a chow diet, compared to wild-type, and these differences were further enhanced after a high fat diet. Reduced preadipocyte proliferation in mutant mice, compared to wt, was shown after in vivo labeling of preadipocytes with EdU and in preadipocytes isolated from mice fat stores and studied in vitro. Mice with the desumoylated THRA had disruptions in cell cycle G1/S transition and this was associated with a reduction in the availability of cyclin D2 and cyclin-dependent kinase 2. The genes coding for cyclin D1, cyclin D2, cyclin-dependent kinase 2 and Culin3 are stimulated by cAMP Response Element Binding Protein (CREB) and contain CREB Response Elements (CREs) in their regulatory regions. We demonstrate, by Chromatin Immunoprecipitation (ChIP) assay, that in mice with the THRA K283Q/K288R mutant there was reduced CREB binding to the CRE. Mice with a THRA sumoylation mutant had reduced fat stores on chow and high fat diets and reduced adipocyte diameter.


Subject(s)
Adipose Tissue, White/metabolism , Sumoylation/physiology , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors alpha/physiology , Adipocytes/pathology , Adipocytes/physiology , Adipose Tissue, White/cytology , Animals , CREB-Binding Protein/metabolism , Cell Proliferation , Diet, High-Fat/adverse effects , Humans , Mice , Mice, Mutant Strains , Small Ubiquitin-Related Modifier Proteins/physiology
13.
Endocrinology ; 162(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-33999131

ABSTRACT

CONTEXT: Hypothyroidism impairs cardiovascular health and contributes to endothelial dysfunction with reduced vasodilation. How 3,5,3'-triiodothyronine (T3) and its receptors are involved in the regulation of vasomotion is not yet fully understood. In general, thyroid hormone receptors (TRs) either influence gene expression (canonical action) or rapidly activate intracellular signaling pathways (noncanonical action). OBJECTIVE: Here we aimed to characterize the T3 action underlying the mechanism of arterial vasodilation and blood pressure (BP) regulation. METHODS: Mesenteric arteries were isolated from male rats, wild-type (WT) mice, TRα knockout (TRα 0) mice, and from knockin mice with a mutation in the DNA-binding domain (TRα GS). In this mutant, DNA binding and thus canonical action is abrogated while noncanonical signaling is preserved. In a wire myograph system, the isolated vessels were preconstricted with norepinephrine. The response to T3 was measured, and the resulting vasodilation (Δ force [mN]) was normalized to maximum contraction with norepinephrine and expressed as percentage vasodilation after maximal preconstriction with norepinephrine (%NE). Isolated vessels were treated with T3 (1 × 10-15 to 1 × 10-5 mol/L) alone and in combination with the endothelial nitric oxide-synthase (eNOS) inhibitor L-NG-nitroarginine methyl ester (L-NAME) or the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin. The endothelium was removed to determine the contribution of T3 to endothelium-dependent vasodilation. The physiological relevance of T3-induced vasodilation was determined by in vivo arterial BP measurements in male and female mice. RESULTS: T3 treatment induced vasodilation of mesenteric arteries from WT mice within 2 minutes (by 21.5 ±â€…1.7%NE). This effect was absent in arteries from TRα 0 mice (by 5.3 ±â€…0.6%NE, P < .001 vs WT) but preserved in TRα GS arteries (by 17.2 ±â€…1.1%NE, not significant vs WT). Inhibition of either eNOS or PI3K reduced T3-mediated vasodilation from 52.7 ±â€…4.5%NE to 28.5 ±â€…4.1%NE and 22.7 ±â€…2.9%NE, respectively. Removal of the endothelium abolished the T3-mediated vasodilation in rat mesenteric arteries (by 36.7 ±â€…5.4%NE vs 3.5 ±â€…6.2%NE). In vivo, T3 injection led to a rapid decrease of arterial BP in WT (by 13.9 ±â€…1.9 mm Hg) and TRα GS mice (by 12.4 ±â€…1.9 mm Hg), but not in TRα 0 mice (by 4.1 ±â€…1.9 mm Hg). CONCLUSION: These results demonstrate that T3 acting through noncanonical TRα action affects cardiovascular physiology by inducing endothelium-dependent vasodilation within minutes via PI3K and eNOS activation.


Subject(s)
Mesenteric Arteries/physiology , Thyroid Hormone Receptors alpha/physiology , Vasodilation/physiology , Animals , Binding Sites/genetics , Blood Pressure/drug effects , Blood Pressure/physiology , DNA/metabolism , Female , Gene Knock-In Techniques , Male , Mice , Mice, Knockout , Mutation , Nitric Oxide Synthase Type III/physiology , Norepinephrine/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Rats , Signal Transduction/physiology , Thyroid Hormone Receptors alpha/chemistry , Thyroid Hormone Receptors alpha/genetics , Triiodothyronine/pharmacology , Vasodilation/drug effects
14.
Dev Biol ; 326(1): 155-68, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19056375

ABSTRACT

Thyroid hormone (T(3)) influences cell proliferation, death and differentiation during development of the central nervous system (CNS). Hormone action is mediated by T(3) receptors (TR) of which there are two subtypes, TRalpha and TRbeta. Specific roles for TR subtypes in CNS development are poorly understood. We analyzed involvement of TRalpha and TRbeta in neural cell proliferation during metamorphosis of Xenopus laevis. Cell proliferation in the ventricular/subventricular neurogenic zones of the tadpole brain increased dramatically during metamorphosis. This increase was dependent on T(3) until mid-prometamorphosis, after which cell proliferation decreased and became refractory to T(3). Using double labeling fluorescent histochemistry with confocal microscopy we found TRalpha expressed throughout the tadpole brain, with strongest expression in proliferating cells. By contrast, TRbeta was expressed predominantly outside of neurogenic zones. To corroborate the histochemical results we transfected living tadpole brain with a Xenopus TRbeta promoter-EGFP plasmid and found that most EGFP expressing cells were not dividing. Lastly, treatment with the TRalpha selective agonist CO23 increased brain cell proliferation; whereas, treatment with the TRbeta-selective agonists GC1 or GC24 did not. Our findings support the view that T(3) acts to induce cell proliferation in the tadpole brain predominantly, if not exclusively, via TRalpha.


Subject(s)
Metamorphosis, Biological , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormone Receptors beta/physiology , Triiodothyronine/physiology , Xenopus laevis/growth & development , Acetates/pharmacology , Animals , Benzhydryl Compounds/pharmacology , Brain/cytology , Brain/growth & development , Cell Proliferation , Green Fluorescent Proteins/metabolism , Hydantoins/pharmacology , Larva/cytology , Larva/growth & development , Neurogenesis/physiology , Phenols/pharmacology , Thyroid Hormone Receptors alpha/antagonists & inhibitors , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/antagonists & inhibitors , Thyroid Hormone Receptors beta/genetics , Triiodothyronine/pharmacology , Xenopus laevis/metabolism
15.
Gen Comp Endocrinol ; 165(1): 11-8, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19481082

ABSTRACT

We cloned the thyroid hormone receptor alpha (TRalpha) and beta (TRbeta) cDNAs from the ovaries of the protandrous black porgy and compared the expression levels of TRalpha and TRbeta mRNA during the sex change in black porgy. We observed that the TRalpha mRNA by quantitative real-time PCR and protein levels by Western blot were highest in the mature ovaries. Additionally, TRbeta mRNA levels were only expressed highly in the mature ovaries when compared to any other gonadal stages. Then, we injected gonadotropin-releasing hormone analogue (GnRHa) to know the effects on TRs mRNA in immature black porgy. Injection with GnRHa resulted in a significant increase in TRalpha level while significantly reducing TRbeta level after 12h. We concluded that TRalpha was related in testicular development as well as ovarian development and TRbeta was only affect to ovarian development in black porgy. These results will provide a framework for better understanding of the role of TRs during sex change processes in this fish.


Subject(s)
Fish Proteins/metabolism , Perciformes/metabolism , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormone Receptors beta/metabolism , Animals , Blotting, Western , Female , Fish Proteins/genetics , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/pharmacology , Male , Ovary/drug effects , Ovary/metabolism , Perciformes/genetics , Polymerase Chain Reaction , Sexual Maturation/drug effects , Sexual Maturation/genetics , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/genetics
16.
J Neurosci ; 28(8): 1904-15, 2008 Feb 20.
Article in English | MEDLINE | ID: mdl-18287507

ABSTRACT

Thyroid hormone (TH) deficiency during development causes severe and permanent neuronal damage, but the primary insult at the tissue level has remained unsolved. We have defined locomotor deficiencies in mice caused by a mutant thyroid hormone receptor alpha1 (TR alpha1) with potent aporeceptor activity attributable to reduced affinity to TH. This allowed identification of distinct functions that required either maternal supply of TH during early embryonic development or sufficient innate levels of hormone during late fetal development. In both instances, continued exposure to high levels of TH after birth and throughout life was needed. The hormonal dependencies correlated with severely delayed appearance of parvalbumin-immunoreactive GABAergic interneurons and increased numbers of calretinin-immunoreactive cells in the neocortex. This resulted in reduced numbers of fast spiking interneurons and defects in cortical network activity. The identification of locomotor deficiencies caused by insufficient supply of TH during fetal/perinatal development and their correlation with subtype-specific interneurons suggest a previously unknown basis for the neuronal consequences of endemic cretinism and untreated congenital hypothyroidism, and specifies TR alpha1 as the receptor isoform mediating these effects.


Subject(s)
Interneurons/metabolism , Motor Skills Disorders/genetics , Motor Skills Disorders/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Animals , Female , Fetal Development/genetics , Interneurons/cytology , Interneurons/physiology , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Motor Activity/genetics , Motor Skills Disorders/pathology , Pregnancy , Thyroid Hormone Receptors alpha/biosynthesis , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormones/metabolism , Thyroid Hormones/physiology
17.
J Physiol ; 587(Pt 6): 1319-29, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19171649

ABSTRACT

The cardiac transient outward current I(to) is regulated by thyroid hormone (T3). However, it remains unclear whether T3 directly modulates underlying gene transcription and which thyroid receptor (TR) isoform might be responsible for gene transactivation. To clarify this situation, we analysed the role of T3 and its receptors alpha1 (TRalpha1) and beta1 (TRbeta1) in regulation of KCNA4, KCND2, KCND3 and KCNIP2 transcription in rat cardiomyocytes. Initial results demonstrated a T3-mediated increase of I(to) current density. T3 stimulation enhanced KCND2 and KCND3 expression and decreased KCNA4 transcription, while KCNIP2 remained unaffected. To dissect the role of TRalpha1 and TRbeta1 in T3-dependent I(to) modulation, TRalpha1 and TRbeta1 were overexpressed in cardiomyocytes by adenovirus-mediated gene transfer. TRalpha1 increased I(to), while TRbeta1 significantly reduced I(to) in size, which was associated with TRalpha1-mediated increase and TRbeta1-mediated reduction of KCND2/3 transcription. To further evaluate a possible direct interaction of TRalpha1 and TRbeta1 with the KCND3 promoter, TR expression vectors were cotransfected with a construct containing 2335 bp of the KCND3 5'-flanking sequence linked to a luciferase reporter into ventricular myocytes. While the TRalpha1 aporeceptor enhanced KCND3 transcription, the TRbeta1 aporeceptor suppressed KCND3 expression, with both effects exhibiting ligand-dependent amplification upon T3 stimulation. Deletion of the KCND3 5'-flanking region localized the suppressible promoter sequence for TRbeta1 to within -293 bp and the activating promoter sequence for TRalpha1 to within -2335 to -1654 bp of the transcription start site. Disruption of putative TR binding sites by mutagenesis abolished the TRalpha1- (G-1651T) and TRbeta1- (G-73T) mediated effects, indicating that TRalpha1 and TRbeta1 response elements map to different regions of the KCND3 promoter. Thus, I(to) is modulated by diverse T3-dependent regulation of underlying gene transcription. TRalpha1 and TRbeta1 exhibit distinct effects on KCND3 transactivation with TRalpha1 enhancing and TRbeta1 suppressing KCND3 transcription.


Subject(s)
Gene Expression Regulation , Myocytes, Cardiac/physiology , Shal Potassium Channels/genetics , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormone Receptors beta/physiology , Animals , Animals, Newborn , Cells, Cultured , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/genetics , Gene Expression/drug effects , Kv Channel-Interacting Proteins/genetics , Kv1.4 Potassium Channel/genetics , Mutation/physiology , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Promoter Regions, Genetic/genetics , Rats , Rats, Inbred Strains , Response Elements/genetics , Transfection , Triiodothyronine/pharmacology
18.
Pflugers Arch ; 458(6): 1061-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19629520

ABSTRACT

Thyroid hormone (TH) markedly modulates cardiovascular function and heart rate. The pacemaker current I(f) and encoding hyperpolarization-activated cation (HCN) genes have been identified as TH targets. To analyze the specific contribution and functional significance of thyroid receptor isoforms responsible for HCN gene transactivation, we generated transgenic neonatal rat cardiomyocytes with adenovirus-mediated overexpression of the thyroid receptors alpha1 (TR alpha 1) and beta1 (TR beta 1), and analyzed native I(f) current and expression levels of the underlying molecular components HCN2 and HCN4. Initial results revealed that spontaneous beating activity was higher in TR alpha 1- and lower in TR beta 1-expressing cardiomyocytes. This was associated with accelerated depolarization velocity and abbreviated action potential duration in cells overexpressing TR alpha 1, while TR beta 1 suppressed phase 4 depolarization and prolonged action potentials. Consistently, TR alpha 1-infected myocytes exhibited larger I(f) current densities along with increased HCN2 and HCN4 mRNA and protein levels. In contrast, HCN2 gene expression was not significantly affected by TR beta 1. TR beta 1 exclusively suppressed HCN4 transcription. T3 application led to significant effects only in controls and TR alpha 1-infected cardiomyocytes; whereas, no ligand-dependent actions were observed in TR beta 1-expressing neonatal cardiomyocytes. Our results demonstrate that TR alpha 1 and TR beta 1 divergently regulate cardiac pacing activity. TH-induced positive chronotropic effects are likely to be mediated by TR alpha 1 through enhanced expression of I(f) pacemaker current and its underlying genes.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Heart/physiology , Ion Channels/genetics , Muscle Proteins/genetics , Potassium Channels/genetics , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormone Receptors beta/physiology , Action Potentials/drug effects , Animals , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Electrophysiological Phenomena , Heart Rate/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/biosynthesis , Muscle Proteins/biosynthesis , Myocytes, Cardiac/metabolism , Potassium Channels/biosynthesis , Rats
19.
Mol Endocrinol ; 22(1): 47-55, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17872380

ABSTRACT

The thyroid hormone (TH) controls, via its nuclear receptor, TH receptor-alpha1 (TRalpha1), intestinal crypt cell proliferation in the mouse. In order to understand whether this receptor also plays a role in intestinal regeneration after DNA damage, we applied a protocol of gamma-ray irradiation and monitored cell proliferation and apoptosis at several time points. In wild-type mice, the dose of 8 Gy induced cell cycle arrest and apoptosis in intestinal crypts a few hours after irradiation. This phenomenon reverted 48 h after irradiation. TRalpha(0/0) mutant mice displayed a constant low level of proliferating cells and a high apoptosis rate during the period of study. At the molecular level, in TRalpha(0/0) animals we observed a delay in the p53 phosphorylation induced by DNA damage. In our search for the expression of the protein kinases responsible for p53 phosphorylation upon irradiation, we have focused on DNA-dependent protein kinase catalytic subunit (DNA-PKcs). The number of cells expressing DNA-PKcs in crypts remained high 48 h after irradiation, specifically in TRalpha mutants. Altogether, in TRalpha(0/0) animals the rate of apoptosis in crypt cells remained high, apparently due to an elevated number of cells still presenting DNA damage. In conclusion, the TRalpha gene plays a role in crypt cell homeostasis by regulating the rate of cell renewal and apoptosis induced by DNA damage.


Subject(s)
DNA Damage , Intestine, Small/physiology , Regeneration/physiology , Thyroid Hormone Receptors alpha/physiology , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Blotting, Western , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Differentiation/genetics , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Female , Gamma Rays , Intestine, Small/cytology , Intestine, Small/metabolism , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Thyroid ; 29(9): 1336-1343, 2019 09.
Article in English | MEDLINE | ID: mdl-31303139

ABSTRACT

Background: Inflammation is associated with marked changes in cellular thyroid hormone (TH) metabolism in triiodothyronine (T3) target organs. In the hypothalamus, type 2 deiodinase (D2), the main T3 producing enzyme, increases upon inflammation, leading to an increase in local T3 availability, which in turn decreases thyrotropin releasing hormone expression in the paraventricular nucleus. Type 3 deiodinase (D3), the T3 inactivating enzyme, decreases during inflammation, which might also contribute to the increased T3 availability in the hypothalamus. While it is known that D2 is regulated by nuclear factor κB (NF-κB) during inflammation, the underlying mechanisms of D3 regulation are unknown. Therefore, the aim of the present study was to investigate inflammation-induced D3 regulation using in vivo and in vitro models. Methods: Mice were injected with a sublethal dose of bacterial endotoxin (lipopolysaccharide [LPS]) to induce a systemic acute-phase response. A human neuroblastoma (SK-N-AS) cell line was used to test the involvement of the thyroid hormone receptor alpha 1 (TRα1) as well as the activator protein-1 (AP-1) and NF-κB inflammatory pathways in the inflammation-induced decrease of D3. Results: D3 expression in the hypothalamus was decreased 24 hours after LPS injection in mice. This decrease was similar in mice lacking the TRα. Incubation of SK-N-AS cells with LPS robustly decreased both D3 mRNA expression and activity. This led to increased intracellular T3 concentrations. The D3 decrease was prevented when NF-κB or AP-1 was inhibited. TRα1 mRNA expression decreased in SK-N-AS cells incubated with LPS, but knockdown of the TRα in SK-N-AS cells did not prevent the LPS-induced D3 decrease. Conclusions: We conclude that the inflammation-induced D3 decrease in the hypothalamus is mediated by the inflammatory pathways NF-κB and AP-1, but not TRα1. Furthermore, the observed decrease modulates intracellular T3 concentrations. Our results suggest a concerted action of inflammatory modulators to regulate both hypothalamic D2 and D3 activities to increase the local TH concentrations.


Subject(s)
Hypothalamus/enzymology , Inflammation/metabolism , Iodide Peroxidase/genetics , Animals , Cell Line, Tumor , Down-Regulation , Female , Humans , Iodide Peroxidase/physiology , Lipopolysaccharides , Male , Mice , NF-kappa B/physiology , RNA, Messenger/analysis , Signal Transduction , Thyroid Hormone Receptors alpha/physiology , Transcription Factor AP-1/physiology , Iodothyronine Deiodinase Type II
SELECTION OF CITATIONS
SEARCH DETAIL