Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 236
Filter
1.
Int J Mol Sci ; 25(19)2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39409130

ABSTRACT

Thyroid hormone binds to specific nuclear receptors, regulating the expression of target genes, with major effects on cardiac function. Triiodothyronine (T3) increases the expression of key proteins related to calcium homeostasis, such as the sarcoplasmic reticulum calcium ATPase pump, but the detailed mechanism of gene regulation by T3 in cardiac voltage-gated calcium (Cav1.2) channels remains incompletely explored. Furthermore, the effects of T3 on Cav1.2 auxiliary subunits have not been investigated. We conducted quantitative reverse transcriptase polymerase chain reaction, Western blot, and immunofluorescence experiments in H9c2 cells derived from rat ventricular tissue, examining the effects of T3 on the expression of α1c, the principal subunit of Cav1.2 channels, and Cavß4, an auxiliary Cav1.2 subunit that regulates gene expression. The translocation of phosphorylated cyclic adenosine monophosphate response element-binding protein (pCREB) by T3 was also examined. We found that T3 has opposite effects on these channel proteins, upregulating α1c and downregulating Cavß4, and that it increases the nuclear translocation of pCREB while decreasing the translocation of Cavß4. Finally, we found that overexpression of Cavß4 represses the mRNA expression of α1c, suggesting that T3 upregulates the expression of the α1c subunit in response to a decrease in Cavß4 subunit expression.


Subject(s)
Calcium Channels, L-Type , Myocytes, Cardiac , Animals , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/genetics , Rats , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Triiodothyronine/pharmacology , Triiodothyronine/metabolism , Down-Regulation/drug effects , Thyroid Hormones/metabolism , Cell Line , Up-Regulation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation/drug effects , Protein Subunits/metabolism , Protein Subunits/genetics
2.
Endocrinology ; 165(7)2024 May 27.
Article in English | MEDLINE | ID: mdl-38862394

ABSTRACT

Alloxan-induced diabetic rats present with hypothyroidism. When treated with triiodothyronine (T3), glycemia and proinflammatory cytokine expression are downregulated, improving insulin sensitivity. The effectiveness of associating T3 with insulin (replacement dose [6 U] and [3 U]) in controlling glycemia was investigated in this experimental model. Male Wistar rats were made diabetic by alloxan injection and sorted into groups treated or not with insulin (3 or 6 U) associated or not with T3 (1.5 µg 100 g-1 BW) for 28 days. Nondiabetic rats constituted the control group. Fasting glycemia, glucose decay rate, and thyrotropin (TSH) were measured in the blood/serum of all animals. Immunoblotting was used to assess total GLUT4 expression in skeletal muscles and epididymal white adipose tissue. Cytokine and nuclear factor-κB (NF-κB) expression were measured in these tissues and liver. Diabetic rats presented with increased fasting glycemia, inflammatory cytokines, and NF-κB expression, TSH levels, and insulin resistance. In diabetic rats treated with T3 and/or insulin, these parameters were decreased, whereas GLUT4 and anti-inflammatory cytokine expression were increased. T3 combined with 3-U insulin restored the parameters to values of the control group and was more effective at controlling glycemia than 6-U insulin. Thus, a combination of T3 and insulin might represent a promising strategy for diabetes management since it reduces the insulin requirement by half and improves glycemic control of diabetic rats, which could postpone insulin resistance that develops with chronic insulin administration. These findings open a perspective for using thyroid analogues that provide tissue-specific effects, which might result in a potentially more effective treatment of diabetes.


Subject(s)
Blood Glucose , Diabetes Mellitus, Experimental , Glucose Transporter Type 4 , Insulin , NF-kappa B , Rats, Wistar , Triiodothyronine , Animals , Male , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Triiodothyronine/blood , Triiodothyronine/pharmacology , Rats , Glucose Transporter Type 4/metabolism , Blood Glucose/metabolism , Blood Glucose/drug effects , NF-kappa B/metabolism , Insulin Resistance , Alloxan , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Thyrotropin/blood , Cytokines/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use
3.
Arq Bras Cardiol ; 121(4): e20230236, 2024 Apr.
Article in Portuguese, English | MEDLINE | ID: mdl-38695407

ABSTRACT

BACKGROUND: Vascular dysfunction constitutes the etiology of many diseases, such as myocardial infarction and hypertension, with the disruption of redox homeostasis playing a role in the imbalance of the vasomotor control mechanism. Our group previously has shown that thyroid hormones exert protective effects on the aortic tissue of infarcted rats by improving angiogenesis signaling. OBJECTIVE: Investigate the role of triiodothyronine (T3) on vascular response, exploring its effects on isolated aortas and whether there is an involvement of vascular redox mechanisms. METHODS: Isolated aortic rings (intact- and denuded-endothelium) precontracted with phenylephrine were incubated with T3 (10-8, 10-7, 10-6, 10-5, and 10-4 M), and tension was recorded using a force-displacement transducer coupled with an acquisition system. To assess the involvement of oxidative stress, aortic rings were preincubated with T3 and subsequently submitted to an in vitro reactive oxygen species (ROS) generation system. The level of significance adopted in the statistical analysis was 5%. RESULTS: T3 (10-4 M) promoted vasorelaxation of phenylephrine precontracted aortic rings in both intact- and denuded-endothelium conditions. Aortic rings preincubated in the presence of T3 (10-4 M) also showed decreased vasoconstriction elicited by phenylephrine (1 µM) in intact-endothelium preparations. Moreover, T3 (10-4 M) vasorelaxation effect persisted in aortic rings preincubated with NG-nitro-L-arginine methylester (L-NAME, 10 µM), a nonspecific NO synthase (NOS) inhibitor. Finally, T3 (10-4 M) exhibited, in vitro, an antioxidant role by reducing NADPH oxidase activity and increasing SOD activity in the aorta's homogenates. CONCLUSION: T3 exerts dependent- and independent-endothelium vasodilation effects, which may be related to its role in maintaining redox homeostasis.


FUNDAMENTO: A disfunção vascular constitui a etiologia de diversas doenças, incluindo infarto do miocárdio e hipertensão, diante da ruptura da homeostase oxi-redutiva ("redox"), desempenhando um papel no desequilíbrio do mecanismo de controle vasomotor. Nosso grupo demonstrou anteriormente que os hormônios tireoidianos melhoram a sinalização da angiogênese, exercendo efeitos protetores sobre o tecido aórtico de ratos infartados. OBJETIVOS: Investigar o papel da triiodotironina (T3) na resposta vascular, explorando seus efeitos em aortas isoladas e a presença de mecanismos redox vasculares. MÉTODOS: Anéis aórticos isolados (endotélio intacto e desnudado) pré-contraídos com fenilefrina foram incubados com T3 (10-8, 10-7, 10-6, 10-5 e 10-4 M) e a tensão foi registrada usando um transdutor de deslocamento de força acoplado a um sistema de coleta. Para avaliar o envolvimento do estresse oxidativo, os anéis aórticos foram pré-incubados com T3 e posteriormente submetidos a um sistema de geração de espécies reativas de oxigênio (ROS) in vitro. O nível de significância adotado na análise estatística foi de 5%. RESULTADOS: A T3 (10-4 M) promoveu o vasorrelaxamento dos anéis aórticos pré-contraídos com fenilefrina em endotélio intacto e desnudado. Os anéis aórticos pré-incubados na presença de T3 (10-4 M) também mostraram diminuição da vasoconstrição provocada pela fenilefrina (1 µM) em preparações de endotélio intacto. Além disso, o efeito vasorrelaxante da T3 (10-4 M) persistiu em anéis aórticos pré-incubados com éster metílico de NG-nitro-L-arginina (L-NAME, 10 µM), um inibidor inespecífico da NO sintase (NOS). Por fim, a T3 (10-4 M) exibiu, in vitro, um papel antioxidante ao reduzir a atividade da NADPH oxidase e aumentar a atividade da SOD nos homogenatos aórticos. CONCLUSÃO: A T3 exerce efeitos dependentes e independentes de endotélio, o que pode estar relacionado ao seu papel na manutenção da homeostase redox.


Subject(s)
Oxidation-Reduction , Oxidative Stress , Rats, Wistar , Reactive Oxygen Species , Triiodothyronine , Vasodilation , Animals , Vasodilation/drug effects , Vasodilation/physiology , Male , Triiodothyronine/pharmacology , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Oxidative Stress/drug effects , Phenylephrine/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Rats , Reproducibility of Results , Vasoconstrictor Agents/pharmacology , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , In Vitro Techniques , Vasoconstriction/drug effects , Vasoconstriction/physiology
4.
Article in English | MEDLINE | ID: mdl-36642213

ABSTRACT

The anti-obesity thyroid hormone, triiodothyronine (T3), and irisin, an exercise- and/or cold-induced myokine, stimulate thermogenesis and energy consumption while decreasing lipid accumulation. The involvement of ATP signaling in adipocyte cell function and obesity has attracted increasing attention, but the crosstalk between the purinergic signaling cascade and anti-obesity hormones lacks experimental evidence. In this study, we investigated the effects of T3 and irisin in the transcriptomics of membrane-bound purinoceptors, ectonucleotidase enzymes and nucleoside transporters participating in the purinergic signaling in cultured human adipocytes. The RNA-seq analysis revealed that differentiated adipocytes express high amounts of ADORA1, P2RY11, P2RY12, and P2RX6 gene transcripts, along with abundant levels of transcriptional products encoding to purine metabolizing enzymes (ENPP2, ENPP1, NT5E, ADA and ADK) and transporters (SLC29A1, SCL29A2). The transcriptomics of purinergic signaling markers changed in parallel to the upsurge of "browning" adipocyte markers, like UCP1 and P2RX5, after treatment with T3 and irisin. Upregulation of ADORA1, ADORA2A and P2RX4 gene transcription was obtained with irisin, whereas T3 preferentially upregulated NT5E, SLC29A2 and P2RY11 genes. Irisin was more powerful than T3 towards inhibition of the leptin gene transcription, the SCL29A1 gene encoding for the ENT1 transporter, the E-NPP2 (autotaxin) gene, and genes that encode for two ADP-sensitive P2Y receptors, P2RY1 and P2RY12. These findings indicate that anti-obesity irisin and T3 hormones differentially affect the purinergic signaling transcriptomics, which might point towards new directions for the treatment of obesity and related metabolic disorders that are worth to be pursued in future functional studies.


Subject(s)
Fibronectins , Transcriptome , Triiodothyronine , Humans , Adipocytes/drug effects , Adipocytes/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Obesity/genetics , Obesity/metabolism , RNA-Seq , Triiodothyronine/pharmacology , Triiodothyronine/metabolism
5.
Metab Brain Dis ; 37(8): 2735-2750, 2022 12.
Article in English | MEDLINE | ID: mdl-35951206

ABSTRACT

Alzheimer disease's (AD) is a neurodegenerative disorder characterized by cognitive and behavioral impairment. The central nervous system is an important target of thyroid hormones (TH). An inverse association between serum triiodothyronine (T3) levels and the risk of AD symptoms and progression has been reported. We investigated the effects of T3 treatment on the depression-like behavior in male transgenic 3xTg-AD mice. Animals were divided into 2 groups treated with daily intraperitoneal injections of 20 ng/g of body weight (b.w.) L-T3 (T3 group) or saline (vehicle, control group). The experimental protocol lasted 21 days, and behavioral tests were conducted on days 18-20. At the end of the experiment, the TH profile and hippocampal gene expression were evaluated. The T3-treated group significantly increased serum T3 and decreased thyroxine (T4) levels. When compared to control hippocampal samples, the T3 group exhibited attenuated glycogen synthase kinase-3 (GSK3), metalloproteinase 10 (ADAM10), amyloid-beta precursor-protein (APP), serotonin transporter (SERT), 5HT1A receptor, monocarboxylate transporter 8 (MCT8) and bone morphogenetic protein 7 (BMP-7) gene expression, whereas augmented superoxide dismutase 2 (SOD2) and Hairless gene expression. T3-treated animals also displayed reduced immobility time in both the tail suspension and forced swim tests, and in the latter presented a higher latency time compared to the control group. Therefore, our findings suggest that in an AD mouse model, T3 supplementation promotes improvements in depression-like behavior, through the modulation of the serotonergic related genes involved in the transmission mediated by 5HT1A receptors and serotonin reuptake, and attenuated disease progression.


Subject(s)
Alzheimer Disease , Triiodothyronine , Animals , Mice , Male , Triiodothyronine/pharmacology , Triiodothyronine/therapeutic use , Alzheimer Disease/metabolism , Depression/drug therapy , Glycogen Synthase Kinase 3 , Mice, Transgenic , Thyroid Hormones/metabolism , Disease Models, Animal
6.
Int Immunopharmacol ; 110: 109016, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35978502

ABSTRACT

Given the ability of dendritic cells (DCs) to modulate other immune players, their manipulation holds great potential for inducing efficient antitumor immunity. However, DC vaccine manufacturing deserve optimization since tumor cell cargo and DC functional state induced by maturation signals influence their in vivo immunogenic potential. We reported that triiodothyronine (T3) stimulates mice DCs' maturation and their ability to promote pro-inflammatory and cytotoxic T-cell responses. This study aimed to evaluate the efficacy of T3-conditioned DC vaccination in a murine model of colon carcinoma, deciphering the molecular players involved; and to examine the effects of T3 on the maturation and activation of human DCs (huDCs). Bone marrow-derived DCs were exposed to T3 and MC38 cancer cells that underwent cell death (MC38-Apo/Nec) by UVB irradiation. Our results showed that MC38-Apo/Nec cells are efficiently uptaken by DCs and that T3 upregulates CD86 expression with increased production of the pro-inflammatory cytokines IL-12, IL-6 and TGF-ß. In a colon cancer model, vaccination with T3-stimulated and tumor antigen-loaded DCs inhibited tumor growth in wild type mice, an effect that was eliminated in IL-17-deficient animals. Notably, secretion of high levels of IFN-γ and IL-17, induction of Th1, Th17 and tumor-specific cytotoxic T lymphocytes characterized the antitumor response upon vaccination. Moreover, our initial findings demonstrated a significant increase in CD86 expression and IL-12 production by huDCs induced by T3. Overall, these results reinforce the adjuvant properties of T3-conditioned DCs to potentiate T-cell-mediated antitumor immunity and support promising advances in the translation process to human oncotherapy.


Subject(s)
Cancer Vaccines , Colonic Neoplasms , Animals , Cancer Vaccines/therapeutic use , Dendritic Cells , Humans , Interleukin-12/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Triiodothyronine/metabolism , Triiodothyronine/pharmacology , Vaccination
7.
J Mol Endocrinol ; 66(3): 207-221, 2021 03.
Article in English | MEDLINE | ID: mdl-33640872

ABSTRACT

Recent studies have provided evidence that triiodothyronine (T3) might play an effective role in the recovery of ischemic myocardium, through the preservation of mitochondrial function and the improvement of energy substrate metabolism. To this respect, it has been suggested that T3 could activate AMP-activated protein kinase (AMPK), the cellular 'fuel-gauge' enzyme, although its role has yet to be elucidated. The aim of the present study was to investigate the effects produced by acute treatment with T3 (60 nM) and the pharmacological inhibition of AMPK by compound C on isolated rat left atria subjected to 75 min simulated ischemia-75 min reperfusion. Results showed that T3 increased AMPK activation during simulated ischemia-reperfusion, while compound C prevented it. At the end of simulated reperfusion, acute T3 treatment increased contractile function recovery and cellular viability conservation. Mitochondrial ultrastructure was better preserved in the presence of T3 as well as mitochondrial ATP production rate and tissue ATP content. Calcium retention capacity, a parameter widely used as an indicator of the resistance of mitochondrial permeability transition pore (MPTP) to opening, and GSK-3ß phosphorylation, a master switch enzyme that limits MPTP opening, were increased by T3 administration. All these beneficial effects exerted by T3 acute treatment were prevented when compound C was co-administrated. The present study provided original evidence that T3 enhances intrinsic activation of AMPK during myocardial ischemia-reperfusion, being this enzyme involved, at least in part, in the protective effects exerted by T3, contributing to mitochondrial structure and function preservation, post-ischemic contractile recovery and conservation of cellular viability.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cardiotonic Agents/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Myocardium/pathology , Triiodothyronine/therapeutic use , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cardiotonic Agents/pharmacology , Cell Survival/drug effects , Diastole/drug effects , Female , Glycogen Synthase Kinase 3 beta/metabolism , Heart Atria/ultrastructure , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/physiopathology , Phosphorylation/drug effects , Rats, Sprague-Dawley , Systole/drug effects , Triiodothyronine/pharmacology
8.
J Cardiovasc Pharmacol ; 76(6): 698-707, 2020 12.
Article in English | MEDLINE | ID: mdl-33105324

ABSTRACT

Cellular death and survival signaling plays a key role in the progress of adverse cardiac remodeling after acute myocardial infarction (AMI). Therapeutic strategies, such as co-treatment with beta-blocker carvedilol and thyroid hormones (THs), give rise to new approaches that can sustain the cellular homeostasis after AMI. Therefore, we sought to investigate the effects of carvedilol and TH co-administration on apoptosis and survival proteins and on cardiac remodeling after AMI. Male Wistar rats were distributed in 5 groups as follows: sham-operated group (SHAM), infarcted group (MI), infarcted plus carvedilol group (MI+C), infarcted plus TH group (MI+TH), and infarcted plus carvedilol and TH co-treatment group (MI+C+TH). Echocardiographic analysis was performed, and hearts were collected for western blot evaluation. The MI group presented systolic posterior wall thickness loss, an increase in the wall tension index, and an increase in atrial natriuretic peptide tissue levels than the SHAM group. However, in the MI+C+TH group, these parameters were equally to the SHAM group. Moreover, whereas the MI group showed Bax protein expression elevated in relation to the SHAM group, the MI+C+TH group presented Bax reduction and also Akt activation compared with the MI group. In addition, the MI+TH group revealed beta-1 adrenergic receptor (ß1AR) upregulation compared with the MI and MI+C groups, whereas the MI+C+TH group presented lower levels of ß1AR in relation to the SHAM and MI+TH groups. In conclusion, we suggest that carvedilol and TH co-administration may mediate its cardioprotective effects against adverse cardiac remodeling post-AMI through the Bax reduction, Akt activation, and ß1AR decrease.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Carvedilol/pharmacology , Myocardial Infarction/drug therapy , Myocardium/metabolism , Thyroxine/pharmacology , Triiodothyronine/pharmacology , Animals , Disease Models, Animal , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt , Rats, Wistar , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction
9.
J Equine Vet Sci ; 86: 102895, 2020 03.
Article in English | MEDLINE | ID: mdl-32067668

ABSTRACT

The effects of two concentrations of triiodothyronine (T3; 0.01 and 1,000 nM) on the osteogenic and chondrogenic differentiation abilities of equine adipose-derived mesenchymal stem cells (AD-MSCs) were evaluated. The osteogenic study evaluated the effect of T3 using alkaline phosphatase activity (ALP) assay; cell viability and density; and formation of mineralized nodules at Days 7, 14, and 21 in culture. The chondrogenic study tested the effect of T3 through ALP assay, mitochondrial metabolism, cell density, and periodic acid-Schiff-positive (PAS+) matrix percentage at Days 7 and 14. In both experiments, analysis of variance was used to compare averages through the Student-Newman-Keuls test. In the osteogenic study, no differences in any variable were detected between groups at Day 7. At Day 14, 0.01 nM T3 reduced cell density and the number of mineralized nodules despite the increase in ALP activity and mitochondrial metabolism (P < .05). ALP activity increased at 1,000 nM T3 concentration (P < .05). At Day 21, 0.01 nM T3 treatment increased ALP activity compared with control treatment (P < .05). At 1,000 nM concentration, T3 reduced mitochondrial metabolism and cell density (P < .05). In the chondrogenic study, the two T3 concentrations increased cell density compared with control treatment at Day 7. At Day 14, higher T3 concentration reduced mitochondrial metabolism, ALP activity, cell density, and PAS+ chondrogenic matrix percentage compared with control treatment (P < .05). Thus, T3 addition to equine AD-MSC cultures has no enhancement effect on osteogenic or chondrogenic differentiation and may, in fact, negatively affect cell density and matrix synthesis depending on hormone concentration and culture time.


Subject(s)
Adipose Tissue , Triiodothyronine , Animals , Cell Differentiation , Cells, Cultured , Horses , Stem Cells , Triiodothyronine/pharmacology
10.
Thyroid ; 29(8): 1060-1072, 2019 08.
Article in English | MEDLINE | ID: mdl-31264512

ABSTRACT

Background: Thyrotoxicosis increases bone turnover, resulting in net bone loss. Sympathetic nervous system (SNS) activation, via ß2-adrenoceptor (ß2-AR) signaling, also has osteopenic effects. Because thyroid hormones (TH) interact with the SNS to regulate several physiological processes, we hypothesized that this interaction also occurs to regulate bone mass. Previous studies support this hypothesis, as α2-AR knockout (KO) mice are less susceptible to thyrotoxicosis-induced osteopenia. Here, we evaluated whether TH-SNS interactions in bone involve ß2-AR signaling. Methods: Thyrotoxicosis was induced in 120-day-old female and male mice with ß2-AR gene inactivation (ß2-AR-/-) by daily treatment with supraphysiological doses of triiodothyronine (T3) for 12 weeks. The impact of thyrotoxicosis on femoral bone microarchitecture, remodeling, fracture risk, and gene expression of the receptor activator of nuclear factor-kappa-B (RANK)-RANK ligand (RANKL)-osteoprotegerin (OPG) pathway was evaluated. In addition, the effect of the ß2-AR-specific agonist clenbuterol (CL) on cAMP accumulation was determined in osteoblastic (MC3T3-E1) cells treated with T3 and/or 17ß-estradiol (E2). Results: Thyrotoxicosis negatively affected trabecular bone microarchitecture in wild-type (WT) females, but this effect was milder or nonexistent in ß2-AR-/- animals, whereas the opposite was seen in males. T3 treatment increased the femoral RANKL/OPG mRNA ratio and the endosteal perimeter and medullary area of the diaphysis in WT females and males, but not in ß2-AR-/- mice, suggesting that T3 promotes endosteal resorption in cortical bone, in a mechanism that involves ß2-AR signaling. T3 treatment increased endocortical mineral apposition rate only in WT females but not in ß2-AR-/- mice, suggesting that TH also induce bone formation in a ß2-AR signaling-dependent mechanism. T3 treatment decreased femoral resistance to fracture only in WT females, but not in KO mice. E2 and CL similarly increased cAMP accumulation in MC3T3-E1 cells; whereas T3 alone had no effect, but it completely blocked E2-stimulated cAMP accumulation, suggesting that some T3 effects on bone may involve E2/cAMP signaling in osteoblasts. Conclusions: These findings sustain the hypothesis that T3 interacts with the SNS to regulate bone morphophysiology in a ß2-AR signaling-dependent mechanism. The data also reveal sex as an important modifier of skeletal manifestations of thyrotoxicosis, as well as a modifier of the TH-SNS interactions to control bone microarchitecture, remodeling, and resistance to fracture.


Subject(s)
Bone Diseases, Metabolic/metabolism , Femur/metabolism , Receptors, Adrenergic, beta-2/metabolism , Thyrotoxicosis/metabolism , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Biomechanical Phenomena , Bone Diseases, Metabolic/etiology , Bone Diseases, Metabolic/pathology , Bone Diseases, Metabolic/physiopathology , Bone Remodeling , Cell Line , Clenbuterol/pharmacology , Cyclic AMP/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Female , Femur/diagnostic imaging , Femur/pathology , Femur/physiopathology , Gene Expression , Male , Mice , Mice, Knockout , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Adrenergic, beta-2/genetics , Signal Transduction , Sympathetic Nervous System/metabolism , Thyrotoxicosis/chemically induced , Thyrotoxicosis/complications , Triiodothyronine/pharmacology , Triiodothyronine/toxicity , X-Ray Microtomography
11.
Arch. endocrinol. metab. (Online) ; 63(2): 142-147, Mar.-Apr. 2019. graf
Article in English | LILACS | ID: biblio-1001213

ABSTRACT

ABSTRACT Objective: To verify the physiological action of triiodothyronine T3 on the expression of transforming growth factor α (TGFA) mRNA in MCF7 cells by inhibition of RNA Polymerase II and the MAPK/ERK pathway Materials and methods: The cell line was treated with T3 at a physiological dose (10−9M) for 10 minutes, 1 and 4 hour (h) in the presence or absence of the inhibitors, α-amanitin (RNA polymerase II inhibitor) and PD98059 (MAPK/ERK pathway inhibitor). TGFA mRNA expression was analyzed by RT-PCR. For data analysis, we used ANOVA, complemented with the Tukey test and Student t-test, with a minimum significance of 5%. Results: T3 increases the expression of TGFA mRNA in MCF7 cells in 4 h of treatment. Inhibition of RNA polymerase II modulates the effect of T3 treatment on the expression of TGFA in MCF7 cells. Activation of the MAPK/ERK pathway is not required for T3 to affect the expression of TGFA mRNA. Conclusion: Treatment with a physiological concentration of T3 after RNA polymerase II inhibition altered the expression of TGFA. Inhibition of the MAPK/ERK pathway after T3 treatment does not interfere with the TGFA gene expression in a breast adenocarcinoma cell line.


Subject(s)
Humans , Female , Triiodothyronine/genetics , Breast Neoplasms/genetics , Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic/genetics , Transforming Growth Factor alpha/genetics , MAP Kinase Signaling System/genetics , Triiodothyronine/metabolism , Triiodothyronine/pharmacology , Proto-Oncogenes/genetics , Breast Neoplasms/metabolism , RNA, Messenger/genetics , Adenocarcinoma/metabolism , Transforming Growth Factor alpha/drug effects , Transforming Growth Factor alpha/metabolism , Cell Line, Tumor/metabolism , MCF-7 Cells/metabolism
12.
Lipids ; 54(2-3): 133-140, 2019 02.
Article in English | MEDLINE | ID: mdl-30891787

ABSTRACT

Adipose tissue (AT), an endocrine organ that modulates several physiological functions by synthesizing and releasing adipokines such as adiponectin, is a metabolic target of triiodothyronine (T3). T3 and adiponectin play important roles in controlling normal metabolic functions such as stimulation of fatty acid oxidation and increase in thermogenesis. The phosphatidylinositol 3-kinase (PI3K) pathway is important for the differentiation of preadipocytes into adipocytes and can be activated by T3 for the transcription of specific genes, such as adiponectin. We examined the role of PI3K in adiponectin modulation by T3 action in murine adipocytes (3T3-L1). The 3T3-L1 adipocytes were treated with 1000 nM T3 for 1 h in the presence or absence of 50 µM LY294002 (LY), a PI3K inhibitor. Then, we assessed the expression of adiponectin and the phosphorylated serine/threonine kinase Akt (pAkt), a PI3K signaling protein, in the adipocytes. Adiponectin and pAKT levels were higher in the T3-adipocyte cells, whereas in the LY group adiponectin was elevated and pAKT was decreased compared to the control (C). PI3K pathway inhibition for 1 h and posterior treatment with T3, in LY + T3, reduced the adiponectin level and increased pAKT levels compared to those in LY. T3 stimulated adiponectin levels by PI3K pathway activation and T3 can compensate alteration in the PI3K pathway, because with inhibition of the pathway it is able to maintain the basal levels of adiponectin and pAKT.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/pharmacology , Chromones/pharmacology , Morpholines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Triiodothyronine/pharmacology , 3T3-L1 Cells , Animals , Cell Differentiation/drug effects , Mice , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects
13.
Cell Physiol Biochem ; 52(2): 354-367, 2019.
Article in English | MEDLINE | ID: mdl-30816679

ABSTRACT

BACKGROUND/AIMS: Although a cross-talk between immune and endocrine systems has been well established, the precise pathways by which these signals co-regulate pro- and antiinflammatory responses on antigen-presenting cells remain poorly understood. In this work we investigated the mechanisms by which triiodothyronine (T3) controls T cell activity via dendritic cell (DC) modulation. METHODS: DCs from wild-type (WT) and IL-6-deficient mice were pulsed with T3. Cytokine production and programmed death protein ligands (PD-L) 1 and 2 expression were assayed by flow cytometry and ELISA. Interferon-regulatory factor-4 (IRF4) expression was evaluated by RT-qPCR and flow cytometry. The ability of DCs to stimulate allogenic splenocytes was assessed in a mixed lymphocyte reaction and the different profile markers were analyzed by flow cytometry and ELISA. For in vivo experiments, DCs treated with ovalbumin and T3 were injected into OTII mice. Proliferation, cytokine production, frequency of FoxP3+ regulatory T (Treg) cells and PD-1+ cells were determined by MTT assay, ELISA and flow cytometry, respectively. RESULTS: T3 endows DCs with pro-inflammatory potential capable of generating IL-17-dominant responses and down-modulating expression of PD-L1 and 2. T3-stimulated WT-DCs increased the proportion of IL-17-producing splenocytes, an effect which was eliminated when splenocytes were incubated with T3-treated DCs derived from IL-6-deficient mice. Enhanced IL-17 expression was recorded in both, CD4- and CD4+ populations and involved the IRF-4 pathway. Particularly, γδ-T cells but not natural killer (NK), NKT, B lymphocytes nor CD8+ T cells were the major source of IL-17-production from CD4- cells. Moreover, T3-conditioned DCs promoted a decrease of the FoxP3+ Treg population. Furthermore, T3 down-modulated PD-1 expression on CD4- cells thereby limiting inhibitory signals driven by this co-inhibitory pathway. Thus, T3 acts at the DC level to drive proinflammatory responses in vitro. Accordingly, we found that T3 induces IL-17 and IFNγ-dominant antigen-specific responses in vivo. CONCLUSION: These results emphasize the relevance of T3 as an additional immune-endocrine checkpoint and a novel therapeutic target to modulate IL-17-mediated pro-inflammatory responses.


Subject(s)
Dendritic Cells/immunology , Interleukin-17/immunology , Signal Transduction/drug effects , Triiodothyronine/pharmacology , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Dendritic Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interleukin-17/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Mice , Mice, Knockout , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Signal Transduction/immunology
14.
Arch Endocrinol Metab ; 63(2): 142-147, 2019.
Article in English | MEDLINE | ID: mdl-30916164

ABSTRACT

OBJECTIVE: To verify the physiological action of triiodothyronine T3 on the expression of transforming growth factor α (TGFA) mRNA in MCF7 cells by inhibition of RNA Polymerase II and the MAPK/ERK pathway. MATERIALS AND METHODS: The cell line was treated with T3 at a physiological dose (10-9M) for 10 minutes, 1 and 4 hour (h) in the presence or absence of the inhibitors, α-amanitin (RNA polymerase II inhibitor) and PD98059 (MAPK/ERK pathway inhibitor). TGFA mRNA expression was analyzed by RT-PCR. For data analysis, we used ANOVA, complemented with the Tukey test and Student t-test, with a minimum significance of 5%. RESULTS: T3 increases the expression of TGFA mRNA in MCF7 cells in 4 h of treatment. Inhibition of RNA polymerase II modulates the effect of T3 treatment on the expression of TGFA in MCF7 cells. Activation of the MAPK/ERK pathway is not required for T3 to affect the expression of TGFA mRNA. CONCLUSION: Treatment with a physiological concentration of T3 after RNA polymerase II inhibition altered the expression of TGFA. Inhibition of the MAPK/ERK pathway after T3 treatment does not interfere with the TGFA gene expression in a breast adenocarcinoma cell line.


Subject(s)
Adenocarcinoma/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , MAP Kinase Signaling System/genetics , Transforming Growth Factor alpha/genetics , Triiodothyronine/genetics , Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor/metabolism , Female , Humans , MCF-7 Cells/metabolism , Proto-Oncogene Mas , Proto-Oncogenes/genetics , RNA, Messenger/genetics , Transforming Growth Factor alpha/drug effects , Transforming Growth Factor alpha/metabolism , Triiodothyronine/metabolism , Triiodothyronine/pharmacology
15.
Mol Cell Endocrinol ; 478: 151-167, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30142372

ABSTRACT

We therefore investigated whether there is synergism between triiodothyronine (T3) hormone and trophic molecules released from mechanically-stressed endothelial cells (EC-enriched medium) in osteogenic phenotype by mapping classical repertory of genes. Although there are studies reporting the efficiency of T3 hormone on bone cells, it is scarce considering their effect in conjunction with other physiologically active molecules, such as those released by the active endothelial cells. To address this issue, human bone marrow-derived mesenchymal stem cells (hBMSCs) were treated with EC-enriched medium subjected to shear-stress up to 72 h in vitro, in conjunction or not with T3 hormone. Although our results found an important synergism considering these parameters on modulating key bone-related gene markers, such as on the alkaline phosphatase (ALP) behavior (at both mRNA and protein content), contributing for osteoblast differentiation, important genes such as OSTERIX and RUNX2 were significantly down-expressed, while a over-expression of RANKL was found when the conjunction effect of T3 and endothelial paracrine signaling was considered. In addition, T3 hormone over expressed both OCT4 and NANOG genes in a DNA epigenetic-independent manner. However, we observed a dynamic reprogramming of DNMT1, DNMT3A, DNMT3B and TET1, important DNA-related epigenetic markers. Specifically, T3 hormone alone up-modulated TET2 transcripts profile. Complimentarily, expression of microRNA (miRs) processing-related genes also was modulated, as well as miR-10b, miR-22, miR-21, miR-143 and miR-145 transcriptional related profiles. Altogether, our results suggested a positive effect of mechanically-stressed endothelial cells-induced paracrine signaling on T3 hormone-obtaining osteogenic phenotype, contributing to understanding the paradoxal effect of T3 hormone on the bone physiology.


Subject(s)
Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Paracrine Communication , Signal Transduction , Stress, Mechanical , Triiodothyronine/pharmacology , DNA Methylation/drug effects , DNA Methylation/genetics , Endothelial Cells/drug effects , Epigenesis, Genetic/drug effects , Extracellular Matrix/metabolism , Fibroblast Growth Factors/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ligands , MicroRNAs/metabolism , Minerals/metabolism , Paracrine Communication/drug effects , Phenotype , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism
16.
Mol Cell Endocrinol ; 478: 45-52, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30031103

ABSTRACT

Rapid actions of triiodothyronine (T3) on thyrotropin (TSH) synthesis and secretion have been described in hypothyroid male rats. However, the molecular mechanisms remain unknown. TαT1 cells, a thyrotroph cell line, was used herein to characterize the possible non-genomic actions of T3 on the expression of alpha (Cga) and Tshb genes, and the posttranscriptional processing and translation of both transcripts. The involvement of αVß3 integrin was also assessed. T3 quickly reduced Tshb mRNA content, poly(A) tail length and its association with ribosomes. The effect of T3 on Tshb gene expression was detected even in the presence of a transcription inhibitor. The decrease in Tshb mRNA content and polyadenylation depend on T3 interaction with αVß3 integrin, while T3 reduced Cga mRNA content by transcriptional action. The translational rate of both transcripts was reduced by a mechanism, which does not depend on T3-αVß3 integrin interaction. Results indicate that, in parallel with the inhibitory transcriptional action in Cga and Tshb gene expression, T3 rapidly triggers additional posttranscriptional mechanisms, reducing the TSH synthesis. These non-genomic actions partially depend on T3-αVß3 integrin interaction at the plasma membrane of thyrotrophs and add new insights to the molecular mechanisms involved in T3 negative feedback loop.


Subject(s)
Feedback, Physiological , Thyrotropin, beta Subunit/genetics , Transcription, Genetic/drug effects , Triiodothyronine/pharmacology , Animals , Cell Line , Cell Survival/drug effects , DNA/metabolism , Glycoprotein Hormones, alpha Subunit/genetics , Glycoprotein Hormones, alpha Subunit/metabolism , Integrin alphaVbeta3/metabolism , Poly A/metabolism , Polyadenylation/drug effects , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism , Thyrotrophs/drug effects , Thyrotrophs/metabolism , Thyrotropin, beta Subunit/metabolism
17.
Life Sci ; 202: 35-43, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29626530

ABSTRACT

AIMS: High fat diet consumes and thyroid hormones (THs) disorders may affect nutrients metabolism, but their impact on the absorptive epithelium, the first place of nutrients access, remains unknown. Our aim was to evaluate the intestinal morphology and nutrients transporters content in mice fed standard (LFD) or high fat (HFD) diets in hypo or hyperthyroidism-induced condition. MATERIAL AND METHODS: C57BL/6 male mice fed LFD or HFD diets for 12 weeks, followed by saline, PTU (antithyroid drug) or T3 treatment up to 30 days. The mice were euthanized and proximal intestine was removed to study GLUT2, GLUT5, PEPT1, FAT-CD36, FATP4, NPC1L1 and NHE3 distribution by Western blotting. Since PPAR-a is activated by fatty acids, which is abundant in the HFD, we also evaluated whether PPAR-a affects nutrients transporters. Thus, mice were treated with fenofibrate, a PPAR-a agonist. KEY FINDINGS: HFD decreased GLUT2, PEPT1, FAT-CD6 and NPC1L1, but increased NHE3, while GLUT5 and FATP4 remained unaltered. THs did not alter distribution of nutrients transporters neither in LFD nor in HFD groups, but they increased villi length and depth crypt in LFD and HFD, respectively. Fenofibrate did not affect content of nutrients transporters, excluding PPAR-a involvement on the HFD-induced changes. SIGNIFICANCE: We assume that chronic HFD consumption reduced most of the nutrients transporters content in the small intestine of mice, which might limit the entrance of nutrients and gain weight. Since NHE3 promotes sodium absorption, and it was increased in HFD group, this finding could contribute to explain the hypertension observed in obesity.


Subject(s)
Diet, High-Fat/adverse effects , Hyperthyroidism/metabolism , Hypothyroidism/metabolism , Intestinal Mucosa/metabolism , Membrane Transport Proteins/metabolism , PPAR alpha/metabolism , Animals , Antithyroid Agents/pharmacology , Fenofibrate/pharmacology , Glucose Tolerance Test , Hyperthyroidism/chemically induced , Hypolipidemic Agents/pharmacology , Hypothyroidism/chemically induced , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestines/drug effects , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , PPAR alpha/antagonists & inhibitors , Propylthiouracil/pharmacology , Sodium-Hydrogen Exchanger 3/metabolism , Thyroid Hormones/metabolism , Triiodothyronine/pharmacology
18.
Clin Sci (Lond) ; 132(11): 1117-1133, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29685981

ABSTRACT

Patients with hyperthyroidism exhibit increased risk of development and progression of cardiac diseases. The activation of the renin-angiotensin system (RAS) has been indirectly implicated in these cardiac effects observed in hyperthyroidism. Angiotensin-(1-7) (Ang-(1-7)) has previously been shown to counterbalance pathological effects of angiotensin II (Ang II). The aim of the present study was to investigate the effects of elevated circulating Ang-(1-7) levels on cardiac effects promoted by hyperthyroidism in a transgenic rat (TG) model that constitutively overexpresses an Ang-(1-7)-producing fusion protein [TGR(A1-7)3292]. TG and wild-type (WT) rats received daily injections (i.p.) of triiodothyronine (T3; 7 µg/100 g of body weight (BW)) or vehicle for 14 days. In contrast with WT rats, the TG rats did not develop cardiac hypertrophy after T3 treatment. Indeed, TG rats displayed reduced systolic blood pressure (SBP) and cardiac hyperdynamic condition induced by hyperthyroidism. Moreover, increased plasma levels of Ang II observed in hyperthyroid WT rats were prevented in TG rats. TG rats were protected from glycogen synthase kinase 3ß (GSK3ß) inactivation and nuclear factor of activated T cells (NFAT) nuclear accumulation induced by T3. In vitro studies evidenced that Ang-(1-7) prevented cardiomyocyte hypertrophy and GSK3ß inactivation induced by T3. Taken together, these data reveal an important cardioprotective action of Ang-(1-7) in experimental model of hyperthyroidism.


Subject(s)
Angiotensin I/physiology , Cardiomegaly/etiology , Glycogen Synthase Kinase 3 beta/physiology , Hyperthyroidism/complications , NFATC Transcription Factors/physiology , Peptide Fragments/physiology , Angiotensin I/blood , Angiotensin I/pharmacology , Animals , Cardiomegaly/blood , Cardiomegaly/diagnostic imaging , Cells, Cultured , Echocardiography , Hyperthyroidism/blood , Hyperthyroidism/chemically induced , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Peptide Fragments/blood , Peptide Fragments/pharmacology , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Wistar , Renin-Angiotensin System/physiology , Signal Transduction/physiology , Triiodothyronine/pharmacology
19.
Physiol Rep ; 6(3)2018 02.
Article in English | MEDLINE | ID: mdl-29388360

ABSTRACT

Thyroid hormones play an important role in glucose metabolism and there is evidence of increased prevalence of thyroid dysfunction in obese and diabetic patients. This study aimed at evaluating the thyroid function and the effects of the triiodothyronine (T3) treatment on glycemia control, insulin sensitivity and subclinical inflammation in cafeteria-diet-induced obesity in rats. Obesity was induced in male Wistar rats by offering a cafeteria diet and a subset of the obese rats was treated with T3 (1.5 µg per 100 g of body weight) for a 28-day period. The pituitary-thyroid axis was evaluated by molecular and biochemical parameters. Cytokine content was measured in the serum as well as in the mesenteric and epididymal white adipose tissue. Obese rats exhibited impairment of glycemia control, increased content of inflammatory cytokines in mesenteric white adipose tissue, decreased serum thyrotropin (TSH) concentration and increased sodium/iodide symporter (NIS) and TSH receptor (TSHR) protein content in thyroid gland. T3 treatment improved insulin sensitivity, glucose tolerance, and reduced inflammatory cytokine content in mesenteric white adipose tissue. In the thyroid gland NIS, TSHR, and thyroperoxidase (TPO) content were reduced while thyroglobulin (TG) content was increased by T3. The thyrotrophic response to negative feedback exerted by T3 was preserved in obese rats. The present data reinforce the beneficial effects of T3 treatment of obese rats on the improvement of insulin sensitivity and on the negative modulation of inflammatory cytokine expression in adipose tissue. Moreover, we have evidenced that the pituitary-thyroid axis is affected in obese rats, as illustrated by the impaired TSH secretion.


Subject(s)
Adipose Tissue/drug effects , Cytokines/blood , Insulin Resistance , Obesity/metabolism , Triiodothyronine/pharmacology , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Cytokines/metabolism , Feedback, Physiological , Male , Rats , Rats, Wistar , Receptors, Thyrotropin/metabolism , Symporters/metabolism , Thyroglobulin/metabolism , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyrotropin/blood
20.
Endocrine ; 59(1): 191-202, 2018 01.
Article in English | MEDLINE | ID: mdl-29210006

ABSTRACT

Hypothyroidism and thyrotoxicosis produce adverse effects in male reproduction by unknown mechanisms. We investigated whether triiodothyronine (T3) modulates luteinizing hormone (LH) and follicle stimulating hormone (FSH) synthesis/secretion, by inducing different thyroid states. In hypothyroidism, the content of Lhb and Fshb mRNAs was increased, while their association to ribosomes and the protein content were reduced and the serum LH and FSH concentrations were augmented and decreased, respectively. Thyrotoxicosis reduced Lhb mRNA and LH serum concentration, and increased Lhb mRNA translational rate. The Fshb mRNA content and its association to ribosomes were also increased, whereas FSH serum concentrations were comparable to euthyroid levels. Acute T3 treatment decreased the total content of Lhb and Fshb mRNAs, and increased their association to ribosomes, as well as the LHB and FSHB contents in secretory granules. This study shows that T3 acts on gonadotrophs, resulting in direct effects on LH and FSH synthesis/secretion of male rats, suggesting that some reproductive disorders observed in men may be associated with thyroid hormone imbalances.


Subject(s)
Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Triiodothyronine/pharmacology , Animals , Gene Expression/drug effects , Hypothyroidism/genetics , Hypothyroidism/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar , Thyroidectomy
SELECTION OF CITATIONS
SEARCH DETAIL