Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Endocrinology ; 163(3)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35038735

RESUMEN

CONTEXT: 3,5,3'-L-triiodothyronine (T3) is a potent inducer of hepatocyte proliferation via the Wnt/ß-catenin signaling pathway. Previous studies suggested the involvement of rapid noncanonical thyroid hormone receptor (TR) ß signaling, directly activating hepatic Wnt/ß-catenin signaling independent from TRß DNA binding. However, the mechanism by which T3 increases Wnt/ß-catenin signaling in hepatocytes has not yet been determined. OBJECTIVE: We aimed to determine whether DNA binding of TRß is required for stimulation of hepatocyte proliferation by T3. METHODS: Wild-type (WT) mice, TRß knockout mice (TRß KO), and TRß mutant mice with either specifically abrogated DNA binding (TRß GS) or abrogated direct phosphatidylinositol 3 kinase activation (TRß 147F) were treated with T3 for 6 hours or 7 days. Hepatocyte proliferation was assessed by Kiel-67 (Ki67) staining and apoptosis by terminal deoxynucleotidyl transferase dUTP nick-end labeling assay. Activation of ß-catenin signaling was measured in primary murine hepatocytes. Gene expression was analyzed by microarray, gene set enrichment analysis (GSEA), and quantitative reverse transcription polymerase chain reaction. RESULTS: T3 induced hepatocyte proliferation with an increased number of Ki67-positive cells in WT and TRß 147F mice (9.2% ±â€…6.5% and 10.1% ±â€…2.9%, respectively) compared to TRß KO and TRß GS mice (1.2% ±â€…1.1% and 1.5% ±â€…0.9%, respectively). Microarray analysis and GSEA showed that genes of the Wnt/ß-catenin pathway-among them, Fzd8 (frizzled receptor 8) and Ctnnb1 (ß-catenin)-were positively enriched only in T3-treated WT and TRß 147F mice while B-cell translocation gene anti-proliferation factor 2 was repressed. Consequently, expression of Ccnd1 (CyclinD1) was induced. CONCLUSIONS: Instead of directly activating Wnt signaling, T3 and TRß induce key genes of the Wnt/ß-catenin pathway, ultimately stimulating hepatocyte proliferation via CyclinD1. Thus, canonical transcriptional TRß action is necessary for T3-mediated stimulation of hepatocyte proliferation.


Asunto(s)
Proliferación Celular/fisiología , Hepatocitos/fisiología , Receptores beta de Hormona Tiroidea/fisiología , Triyodotironina/farmacología , Animales , Sitios de Unión/genética , Proliferación Celular/efectos de los fármacos , Ciclina D1/fisiología , ADN/metabolismo , Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hipotiroidismo , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Mutación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Receptores beta de Hormona Tiroidea/genética , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
2.
Invest Ophthalmol Vis Sci ; 59(15): 6089-6101, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30592497

RESUMEN

Purpose: Replacing cone photoreceptors, the units of the retina necessary for daytime vision, depends upon the successful production of a full variety of new cones from, for example, stem cells. Using genetic experiments in a model organism with high cone diversity, zebrafish, we map the intersecting effects of cone development factors gdf6a, tbx2b, and thrß. Methods: We investigated these genes of interest by using genetic combinations of mutants, gene knockdown, and dominant negative gene expression, and then quantified cone subtype outcomes (which normally develop in tightly regulated ratios). Results: Gdf6a mutants have reduced blue cones and, discovered here, reduced red cones. In combined gdf6a/tbx2b disruption, the loss of gdf6a in heterozygous tbx2b mutants reduced UV cones. Intriguingly, when we disrupted thrß in gdf6a mutants by using a thrß morpholino, their combined early disruption revealed a lamination phenotype. Disrupting thrß activity via expression of a dominant negative thrß (dnthrß) at either early or late retinal development had differential outcomes on red cones (reduced abundance), versus UV and blue cones (increased abundance). By using dnthrß in gdf6a mutants, we revealed that disrupting thrß activity did not change gdf6a mutant cone phenotypes. Conclusions: Gdf6a loss directly affects blue and red cones and indirectly affects UV cones by increasing sensitivity to additional disruption, such as reduced tbx2b, resulting in fewer UV cones. The effects of thrß change through photoreceptor development, first promoting red cones and restricting UV cones, and later restricting UV and blue cones. The effects of gdf6a on UV, blue, and red cone development overlap with, but likely supersede, those of thrß.


Asunto(s)
Diferenciación Celular/fisiología , Factor 6 de Diferenciación de Crecimiento/fisiología , Retina/embriología , Células Fotorreceptoras Retinianas Conos/citología , Células Madre/fisiología , Proteínas de Dominio T Box/fisiología , Receptores beta de Hormona Tiroidea/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Embrión no Mamífero , Regulación de la Expresión Génica/fisiología , Silenciador del Gen , Técnicas de Genotipaje , Inmunohistoquímica , Modelos Animales , Células Fotorreceptoras de Vertebrados/fisiología , Polimorfismo de Longitud del Fragmento de Restricción , Pez Cebra
3.
Sci Signal ; 11(536)2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29945885

RESUMEN

Thyroid hormone receptor ß1 (THRB1) and estrogen-related receptor α (ESRRA; also known as ERRα) both play important roles in mitochondrial activity. To understand their potential interactions, we performed transcriptome and ChIP-seq analyses and found that many genes that were co-regulated by both THRB1 and ESRRA were involved in mitochondrial metabolic pathways. These included oxidative phosphorylation (OXPHOS), the tricarboxylic acid (TCA) cycle, and ß-oxidation of fatty acids. TH increased ESRRA expression and activity in a THRB1-dependent manner through the induction of the transcriptional coactivator PPARGC1A (also known as PGC1α). Moreover, TH induced mitochondrial biogenesis, fission, and mitophagy in an ESRRA-dependent manner. TH also induced the expression of the autophagy-regulating kinase ULK1 through ESRRA, which then promoted DRP1-mediated mitochondrial fission. In addition, ULK1 activated the docking receptor protein FUNDC1 and its interaction with the autophagosomal protein MAP1LC3B-II to induce mitophagy. siRNA knockdown of ESRRA, ULK1, DRP1, or FUNDC1 inhibited TH-induced autophagic clearance of mitochondria through mitophagy and decreased OXPHOS. These findings show that many of the mitochondrial actions of TH are mediated through stimulation of ESRRA expression and activity, and co-regulation of mitochondrial turnover through the PPARGC1A-ESRRA-ULK1 pathway is mediated by their regulation of mitochondrial fission and mitophagy. Hormonal or pharmacologic induction of ESRRA expression or activity could improve mitochondrial quality in metabolic disorders.


Asunto(s)
Autofagia , Mitocondrias/fisiología , Dinámicas Mitocondriales , Mitofagia , Receptores de Estrógenos/metabolismo , Receptores beta de Hormona Tiroidea/fisiología , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Células Cultivadas , Dinaminas/genética , Dinaminas/metabolismo , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosforilación , Receptores de Estrógenos/genética , Receptor Relacionado con Estrógeno ERRalfa
4.
Endocrinology ; 159(6): 2484-2494, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29750276

RESUMEN

Thyroid hormone receptor ß (TRß) suppresses tumor growth through regulation of gene expression, yet the associated TRß-mediated changes in chromatin assembly are not known. The chromatin ATPase brahma-related gene 1 (BRG1; SMARCA4), a key component of chromatin-remodeling complexes, is altered in many cancers, but its role in thyroid tumorigenesis and TRß-mediated gene expression is unknown. We previously identified the oncogene runt-related transcription factor 2 (RUNX2) as a repressive target of TRß. Here, we report differential expression of BRG1 in nonmalignant and malignant thyroid cells concordant with TRß. BRG1 and TRß have similar nuclear distribution patterns and significant colocalization. BRG1 interacts with TRß, and together, they are part of the regulatory complex at the RUNX2 promoter. Loss of BRG1 increases RUNX2 levels, whereas reintroduction of TRß and BRG1 synergistically decreases RUNX2 expression. RUNX2 promoter accessibility corresponded to RUNX2 expression levels. Inhibition of BRG1 activity increased accessibility of the RUNX2 promoter and corresponding expression. Our results reveal a mechanism of TRß repression of oncogenic gene expression: TRß recruitment of BRG1 induces chromatin compaction and diminishes RUNX2 expression. Therefore, BRG1-mediated chromatin remodeling may be obligatory for TRß transcriptional repression and tumor suppressor function in thyroid tumorigenesis.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , ADN Helicasas/fisiología , Proteínas Nucleares/fisiología , Receptores beta de Hormona Tiroidea/fisiología , Factores de Transcripción/fisiología , Carcinogénesis/genética , Células Cultivadas , Regulación hacia Abajo/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología
5.
Cancer Res ; 77(24): 6838-6850, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-28972075

RESUMEN

Germline RB1 mutations strongly predispose humans to cone precursor-derived retinoblastomas and strongly predispose mice to pituitary tumors, yet shared cell type-specific circuitry that sensitizes these different cell types to the loss of RB1 has not been defined. Here we show that the cell type-restricted thyroid hormone receptor isoform TRß2 sensitizes to RB1 loss in both settings by antagonizing the widely expressed and tumor-suppressive TRß1. TRß2 promoted expression of the E3 ubiquitin ligase SKP2, a critical factor for RB1-mutant tumors, by enabling EMI1/FBXO5-dependent inhibition of SKP2 degradation. In RB1 wild-type neuroblastoma cells, endogenous Rb or ectopic TRß2 was required to sustain SKP2 expression as well as cell viability and proliferation. These results suggest that in certain contexts, Rb loss enables TRß1-dependent suppression of SKP2 as a safeguard against RB1-deficient tumorigenesis. TRß2 counteracts TRß1, thus disrupting this safeguard and promoting development of RB1-deficient malignancies. Cancer Res; 77(24); 6838-50. ©2017 AACR.


Asunto(s)
Proliferación Celular/genética , Proteína de Retinoblastoma/fisiología , Proteínas Quinasas Asociadas a Fase-S/genética , Receptores beta de Hormona Tiroidea/fisiología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Mutación de Línea Germinal , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Proteína de Retinoblastoma/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Activación Transcripcional/genética , Células Tumorales Cultivadas
6.
Proc Natl Acad Sci U S A ; 114(40): E8458-E8467, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28923959

RESUMEN

Nuclear receptor corepressor 1 (NCoR1) is considered to be the major corepressor that mediates ligand-independent actions of the thyroid hormone receptor (TR) during development and in hypothyroidism. We tested this by expressing a hypomorphic NCoR1 allele (NCoR1ΔID), which cannot interact with the TR, in Pax8-KO mice, which make no thyroid hormone. Surprisingly, abrogation of NCoR1 function did not reverse the ligand-independent action of the TR on many gene targets and did not fully rescue the high mortality rate due to congenital hypothyroidism in these mice. To further examine NCoR1's role in repression by the unliganded TR, we deleted NCoR1 in the livers of euthyroid and hypothyroid mice and examined the effects on gene expression and enhancer activity measured by histone 3 lysine 27 (H3K27) acetylation. Even in the absence of NCoR1 function, we observed strong repression of more than 43% of positive T3 (3,3',5-triiodothyronine) targets in hypothyroid mice. Regulation of approximately half of those genes correlated with decreased H3K27 acetylation, and nearly 80% of these regions with affected H3K27 acetylation contained a bona fide TRß1-binding site. Moreover, using liver-specific TRß1-KO mice, we demonstrate that hypothyroidism-associated changes in gene expression and histone acetylation require TRß1. Thus, many of the genomic changes mediated by the TR in hypothyroidism are independent of NCoR1, suggesting a role for additional signaling modulators in hypothyroidism.


Asunto(s)
Hipotiroidismo/patología , Hígado/patología , Mutación , Co-Represor 1 de Receptor Nuclear/fisiología , Receptores beta de Hormona Tiroidea/fisiología , Hormonas Tiroideas/metabolismo , Acetilación , Animales , Células Cultivadas , Regulación de la Expresión Génica , Histonas/metabolismo , Hipotiroidismo/genética , Hipotiroidismo/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Transducción de Señal
7.
Anim Sci J ; 88(12): 1943-1954, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28782242

RESUMEN

Thyroid hormones are important in the development and regulation of testes. This study was conducted to determine the effects of hyper- and hypothyroidism on testicular development in prepubertal rats aged 20-70 days. Weaning male rats (20 days old) until day 70 age were randomly divided into four groups: control, hyperthyroid (hyper-T), hypothyroid (hypo-T) and hypothyroid treated with thyroxine (T4) (hypo-T+T4). The results indicated that thyroid hormones caused a significant effect in body and testis weights, and food and water consumption. In addition there were changes in serum concentrations of tri-iodothyronine, T4, thyroid stimulating hormone (TSH) and testosterone. Histomorphology showed a significant decrease in seminiferous tubule diameter in hyper-T compared to the other groups. Leydig cell numbers showed a significant elevation in hyper-T but not in hypo-T groups. Immunostaining indicated that TSH receptor (TSHR), thyroid hormone receptors α/ß (TRαß) and proliferating cell nuclear antigen (PCNA) have the roles in testicular development. Our findings suggest that hyper- and hypo-thyroidism regulate testicular cell proliferation and spermatogenesis in prepubertal rats, indicating that expression of TSHR, TRαß and PCNA may be regulated by thyroid hormones that are involved in testicular development; and that the administration of T4 to the hypo-T+T4 group leads to an improvement in the testicular condition.


Asunto(s)
Proliferación Celular , Hipertiroidismo/fisiopatología , Hipotiroidismo/fisiopatología , Pubertad/fisiología , Testículo/citología , Testículo/crecimiento & desarrollo , Hormonas Tiroideas/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Hipertiroidismo/patología , Hipotiroidismo/tratamiento farmacológico , Hipotiroidismo/patología , Células Intersticiales del Testículo/citología , Masculino , Antígeno Nuclear de Célula en Proliferación/metabolismo , Antígeno Nuclear de Célula en Proliferación/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Tirotropina/metabolismo , Espermatogénesis , Testículo/patología , Testosterona/sangre , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores alfa de Hormona Tiroidea/fisiología , Receptores beta de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/fisiología , Hormonas Tiroideas/sangre , Tiroxina/administración & dosificación , Tiroxina/farmacología
8.
Medicine (Baltimore) ; 95(33): e4415, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27537566

RESUMEN

BACKGROUND: Thyroid hormone resistance syndrome (THRS) is a rare disorder with increased concentrations of free thyroxine (FT4) and triiodothyronine (FT3), but normal or slightly increased thyroid-stimulating hormone (TSH). The mutations in the thyroid hormone receptor ß (THRß) gene are thought to be the main pathogenesis. OBJECTIVES: The aims of this study were to present 1 pedigree of Chinese THRS, summarize their clinical characteristics, and analyze the gene mutation. METHODS: The clinical characteristics and thyroid function of the proband and his family members were collected. Gene mutations were analyzed by DNA sequencing. RESULTS: The proband and his mother exhibited symptoms of hyperthyroidism, such as palpitations, heat intolerance, and perspiration. The mother also had atrial fibrillation. The rest of the kindred did not display clinical manifestations of hyper- or hypothyroidism. DNA sequencing revealed a heterozygous G>A missense mutation at position 949 in Exon 9 of THRß both in the patient and his mother, which led to the transition from alanine to threonine at position 317 of THRß protein (A317T), whereas the rest of the kindred did not share this mutation. The proband and his mother were diagnosed with pituitary resistance to thyroid hormone. Oral administration of methimazole was stopped and ß-receptor blockers were administrated. CONCLUSIONS: We present 1 pedigree of THRS with heterozygous A317T mutation in THRß gene in the proband and his mother, which is the first reported mutation in Chinese and provides a comprehensive review of available literature.


Asunto(s)
Receptores beta de Hormona Tiroidea/genética , Síndrome de Resistencia a Hormonas Tiroideas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , China , Femenino , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense/genética , Linaje , Receptores beta de Hormona Tiroidea/fisiología , Adulto Joven
9.
Endocrinology ; 157(8): 3278-92, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27253998

RESUMEN

Dysregulation of the thyroid hormone receptor (TR)ß is common in human cancers. Restoration of functional TRß delays tumor progression in models of thyroid and breast cancers implicating TRß as a tumor suppressor. Conversely, aberrant expression of the runt-related transcription factor 2 (Runx2) is established in the progression and metastasis of thyroid, breast, and other cancers. Silencing of Runx2 diminishes tumor invasive characteristics. With TRß as a tumor suppressor and Runx2 as a tumor promoter, a compelling question is whether there is a functional relationship between these regulatory factors in thyroid tumorigenesis. Here, we demonstrated that these proteins are reciprocally expressed in normal and malignant thyroid cells; TRß is high in normal cells, and Runx2 is high in malignant cells. T3 induced a time- and concentration-dependent decrease in Runx2 expression. Silencing of TRß by small interfering RNA knockdown resulted in a corresponding increase in Runx2 and Runx2-regulated genes, indicating that TRß levels directly impact Runx2 expression and associated epithelial to mesenchymal transition molecules. TRß specifically bound to 3 putative thyroid hormone-response element motifs within the Runx2-P1 promoter ((-)105/(+)133) as detected by EMSA and chromatin immunoprecipitation. TRß suppressed Runx2 transcriptional activities, thus confirming TRß regulation of Runx2 at functional thyroid hormone-response elements. Significantly, these findings indicate that a ratio of the tumor-suppressor TRß and tumor-promoting Runx2 may reflect tumor aggression and serve as biomarkers in biopsy tissues. The discovery of this TRß-Runx2 signaling supports the emerging role of TRß as a tumor suppressor and reveals a novel pathway for intervention.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Receptores beta de Hormona Tiroidea/fisiología , Neoplasias de la Tiroides/genética , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Regiones Promotoras Genéticas/efectos de los fármacos , Elementos de Respuesta , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo , Activación Transcripcional/efectos de los fármacos , Triyodotironina/farmacología
10.
J Endocrinol ; 224(3): 289-301, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25501997

RESUMEN

Thyroid hormone (TH) acts through specific receptors (TRs), which are conditional transcription factors, to induce fibroblast growth factor 21 (FGF21), a peptide hormone that is usually induced by fasting and that influences lipid and carbohydrate metabolism via local hepatic and systemic endocrine effects. While TH and FGF21 display overlapping actions when administered, including reductions in serum lipids, according to the current models these hormones act independently in vivo. In this study, we examined mechanisms of regulation of FGF21 expression by TH and tested the possibility that FGF21 is required for induction of hepatic TH-responsive genes. We confirm that active TH (triiodothyronine (T3)) and the TRß-selective thyromimetic GC1 increase FGF21 transcript and peptide levels in mouse liver and that this effect requires TRß. T3 also induces FGF21 in cultured hepatocytes and this effect involves direct actions of TRß1, which binds a TRE within intron 2 of FGF21. Gene expression profiles of WT and Fgf21-knockout mice are very similar, indicating that FGF21 is dispensable for the majority of hepatic T3 gene responses. A small subset of genes displays diminished T3 response in the absence of FGF21. However, most of these are not obviously directly involved in T3-dependent hepatic metabolic processes. Consistent with these results, T3-dependent effects on serum cholesterol are maintained in the Fgf21(-/-) background and we observe no effect of the Fgf21-knockout background on serum triglycerides and glucose. Our findings indicate that T3 regulates the genes involved in classical hepatic metabolic responses independently of FGF21.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Hígado/metabolismo , Receptores beta de Hormona Tiroidea/fisiología , Animales , Factores de Crecimiento de Fibroblastos/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Elementos de Respuesta , Triyodotironina/farmacología
11.
Cardiovasc Res ; 102(3): 448-59, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24604622

RESUMEN

AIMS: Endothelial dysfunction is an initial and vascular smooth muscle cell (VSMC) apoptosis, a later step of atherosclerosis. Hypothyroidism accelerates atherosclerosis. However, the early events responsible for this pro-atherosclerotic effect are unclear. METHODS AND RESULTS: Rats were resistant to induction of atherosclerosis by high cholesterol diet alone, but became susceptible in hypothyroid state achieved by administration of propylthiouracil (PTU) for 6 weeks. VSMC dysfunction and apoptosis were obvious within 1 week after PTU treatment, without signs of endothelial dysfunction. This early VSMC damage was caused by hypothyroidism but not the high cholesterol diet. In ApoE knockout mice, PTU-induced hypothyroidism triggered early VSMC apoptosis, increased oxidative stress, and accelerated atherosclerosis development. Thyroid hormone supplementation (T4, 10, or 50 µg/kg) prevented atherogenic phenotypes in hypothyroid rats and mice. In rats, thyroidectomy caused severe hypothyroidism 5 days after operation, which also led to rapid VSMC dysfunction and apoptosis. In vitro studies did not show a direct toxic effect of PTU on VSMCs. In contrast, thyroid hormone (T3, 0.75 µg/L plus T4, 50 nmol/L) exerted a direct protection against VSMC apoptosis, which was reduced by knockdown of TRα1, rather than TRß1 and TRß2 receptors. TRα1-mediated inhibition of apoptotic signalling of JNKs and caspase-3 contributed to the anti-apoptotic action of thyroid hormone. CONCLUSION: These findings provide an in vivo example for VSMC apoptosis as an early trigger of hypothyroidism-associated atherosclerosis, and reveal activation of TRα1 receptors to prevent VSMC apoptosis as a therapeutic strategy in this disease.


Asunto(s)
Apoptosis , Aterosclerosis/etiología , Hipotiroidismo/complicaciones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Animales , Apolipoproteínas E/fisiología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Propiltiouracilo/toxicidad , Ratas , Ratas Sprague-Dawley , Receptores alfa de Hormona Tiroidea/fisiología , Receptores beta de Hormona Tiroidea/fisiología , Hormonas Tiroideas/deficiencia
12.
Thyroid ; 24(2): 260-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23731250

RESUMEN

BACKGROUND: Previous studies showed a close association between several types of human cancers and somatic mutations of thyroid hormone receptor ß (TRß) and reduced expression of TRß due to epigenetic inactivation and/or deletion of the THRB gene. These observations suggest that TRß could act as a tumor suppressor in carcinogenesis. However, the mechanisms by which TRß could function to inhibit tumorigenesis are less well understood. METHODS: We used the human follicular thyroid cancer cell lines (FTC-133 and FTC-236 cells) to elucidate how functional expression of the THRB gene could affect tumorigenesis. We stably expressed the THRB gene in FTC cells and evaluated the effects of the expressed TRß on cancer cell proliferation, migration, and tumor growth in cell-based studies and xenograft models. RESULTS: Expression of TRß in FTC-133 cells, as compared with control FTC cells without TRß, reduced cancer cell proliferation and impeded migration of tumor cells through inhibition of the AKT-mTOR-p70 S6K pathway. TRß expression in FTC-133 and FTC-236 led to less tumor growth in xenograft models. Importantly, new vessel formation was significantly suppressed in tumors induced by FTC cells expressing TRß compared with control FTC cells without TRß. The decrease in vessel formation was mediated by the downregulation of vascular endothelial growth factor in FTC cells expressing TRß. CONCLUSIONS: These findings indicate that TRß acts as a tumor suppressor through downregulation of the AKT-mTOR-p70 S6K pathway and decreased vascular endothelial growth factor expression in FTC cells. The present results raise the possibility that TRß could be considered as a potential therapeutic target for thyroid cancer.


Asunto(s)
Receptores beta de Hormona Tiroidea/fisiología , Proteínas Supresoras de Tumor/fisiología , Adenocarcinoma Folicular/patología , Inhibidores de la Angiogénesis/fisiología , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Serina-Treonina Quinasas TOR , Receptores beta de Hormona Tiroidea/biosíntesis , Receptores beta de Hormona Tiroidea/genética , Neoplasias de la Tiroides/patología , Triyodotironina/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/biosíntesis
13.
PLoS One ; 8(6): e67954, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840792

RESUMEN

Studies have suggested that the nuclear receptor corepressor 1 (NCOR1) could play an important role in human cancers. However, the detailed molecular mechanisms by which it functions in vivo to affect cancer progression are not clear. The present study elucidated the in vivo actions of NCOR1 in carcinogenesis using a mouse model (Thrb(PV/PV) mice) that spontaneously develops thyroid cancer. Thrb(PV/PV) mice harbor a dominantly negative thyroid hormone receptor ß (TRß) mutant (denoted as PV). We adopted the loss-of-the function approach by crossing Thrb(PV) mice with mice that globally express an NCOR1 mutant protein (NCOR1ΔID) in which the receptor interaction domains have been modified so that it cannot interact with the TRß, or PV, in mice. Remarkably, expression of NCOR1ΔID protein reduced thyroid tumor growth, markedly delayed tumor progression, and prolonged survival of Thrb(PV/PV)Ncor1 (ΔID/ΔID) mice. Tumor cell proliferation was inhibited by increased expression of cyclin-dependent kinase inhibitor 1 (p21(waf1/cip1); Cdkn1A), and apoptosis was activated by elevated expression of pro-apoptotic BCL-Associated X (Bax). Further analyses showed that p53 was recruited to the p53-binding site on the proximal promoter of the Cdkn1A and the Bax gene as a co-repressor complex with PV/NCOR1/histone deacetylas-3 (HDAC-3), leading to repression of the Cdkn1A as well as the Bax gene in thyroids of Thrb(PV/PV) mice. In thyroids of Thrb(PV/PV)Ncor1 (ΔID/ΔID) mice, the p53/PV complex could not recruit NCOR1ΔID and HDAC-3, leading to de-repression of both genes to inhibit cancer progression. The present studies provided direct evidence in vivo that NCOR1 could function as an oncogene via transcription regulation in a mouse model of thyroid cancer.


Asunto(s)
Proliferación Celular , Modelos Animales de Enfermedad , Co-Represor 1 de Receptor Nuclear/fisiología , Receptores beta de Hormona Tiroidea/fisiología , Neoplasias de la Tiroides/patología , Animales , Apoptosis , Ratones , Ratones Noqueados , Transducción de Señal , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo
14.
Endocrinology ; 154(5): 1940-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23493375

RESUMEN

The effects of thyroid hormones (THs) on brain development and function are largely mediated by the control of gene expression. This is achieved by the binding of the genomically active T3 to transcriptionally active nuclear TH receptors (TRs). T3 and the TRs can either induce or repress transcription. In hypothyroidism, the reduction of T3 lowers the expression of a set of genes, the positively regulated genes, and increases the expression of negatively regulated genes. Two mechanisms may account for the effect of hypothyroidism on genes regulated directly by T3: first, the loss of T3 signaling and TR transactivation, and second, an intrinsic activity of the unliganded TRs directly responsible for repression of positive genes and enhancement of negative genes. To analyze the contribution of the TR subtypes α and ß, we have measured by RT-PCR the expression of a set of positive and negative genes in the cerebral cortex and the striatum of TR-knockout male and female mice. The results indicate that TRα1 exerts a predominant but not exclusive role in the regulation of positive and negative genes. However, a fraction of the genes analyzed are not or only mildly affected by the total absence of TRs. Furthermore, hypothyroidism has a mild effect on these genes in the absence of TRα1, in agreement with a role of unliganded TRα1 in the effects of hypothyroidism.


Asunto(s)
Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Expresión Génica , Receptores alfa de Hormona Tiroidea/fisiología , Receptores beta de Hormona Tiroidea/fisiología , Animales , Animales Recién Nacidos , Corteza Cerebral/crecimiento & desarrollo , Cuerpo Estriado/crecimiento & desarrollo , Femenino , Expresión Génica/genética , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis por Micromatrices , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Receptores alfa de Hormona Tiroidea/genética , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo
15.
Thyroid ; 22(12): 1275-82, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22985455

RESUMEN

BACKGROUND: The iodine-containing drug amiodarone (Amio) and its noniodine containing analogue dronedarone (Dron) are potent antiarrhythmic drugs. Previous in vivo and in vitro studies have shown that the major metabolite of Amio, desethylamiodarone, acts as a thyroid hormone receptor (TR) α(1) and ß(1) antagonist, whereas the major metabolite of Dron debutyldronedarone acts as a selective TRα(1) antagonist. In the present study, Amio and Dron were used as tools to discriminate between TRα(1) or TRß(1) regulated genes in central and peripheral thyroid hormone metabolism. METHODS: Three groups of male rats received either Amio, Dron, or vehicle by daily intragastric administration for 2 weeks. We assessed the effects of treatment on triiodothyronine (T(3)) and thyroxine (T(4)) plasma and tissue concentrations, deiodinase type 1, 2, and 3 mRNA expressions and activities, and thyroid hormone transporters monocarboxylate transporter 8 (MCT8), monocarboxylate transporter 10 (MCT10), and organic anion transporter 1C1 (OATP1C1). RESULTS: Amio treatment decreased serum T(3), while serum T(4) and thyrotropin (TSH) increased compared to Dron-treated and control rats. At the central level of the hypothalamus-pituitary-thyroid axis, Amio treatment decreased hypothalamic thyrotropin releasing hormone (TRH) expression, while increasing pituitary TSHß and MCT10 mRNA expression. Amio decreased the pituitary D2 activity. By contrast, Dron treatment resulted in decreased hypothalamic TRH mRNA expression only. Upon Amio treatment, liver T(3) concentration decreased substantially compared to Dron and control rats (50%, p<0.01), but liver T(4) concentration was unaffected. In addition, liver D1, mRNA, and activity decreased, while the D3 activity and mRNA increased. Liver MCT8, MCT10, and OATP1C1 mRNA expression were similar between groups. CONCLUSION: Our results suggest an important role for TRα1 in the regulation of hypothalamic TRH mRNA expression, whereas TRß plays a dominant role in pituitary and liver thyroid hormone metabolism.


Asunto(s)
Receptores alfa de Hormona Tiroidea/fisiología , Receptores beta de Hormona Tiroidea/fisiología , Hormonas Tiroideas/metabolismo , Animales , Hipotálamo/metabolismo , Yoduro Peroxidasa/genética , Hígado/metabolismo , Masculino , Hipófisis/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Wistar , Receptores alfa de Hormona Tiroidea/antagonistas & inhibidores , Receptores beta de Hormona Tiroidea/antagonistas & inhibidores
16.
Endocrinology ; 153(3): 1548-60, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22253431

RESUMEN

Thyroid hormone is critical for auditory development and has well-known actions in the inner ear. However, less is known of thyroid hormone functions in the middle ear, which contains the ossicles (malleus, incus, stapes) that relay mechanical sound vibrations from the outer ear to the inner ear. During the later stages of middle ear development, prior to the onset of hearing, middle ear cavitation occurs, involving clearance of mesenchyme from the middle ear cavity while the immature cartilaginous ossicles attain appropriate size and ossify. Using in situ hybridization, we detected expression of Thra and Thrb genes encoding thyroid hormone receptors α1 and ß (TRα1 and TRß, respectively) in the immature ossicles, surrounding mesenchyme and tympanic membrane in the mouse. Thra(+/PV) mice that express a dominant-negative TRα1 protein exhibited deafness with elevated auditory thresholds and a range of middle ear abnormalities including chronic persistence of mesenchyme in the middle ear into adulthood, markedly enlarged ossicles, and delayed ossification of the ossicles. Congenitally hypothyroid Tshr(-/-) mice and TR-deficient Thra1(-/-);Thrb(-/-) mice displayed similar abnormalities. These findings demonstrate that middle ear maturation is TR dependent and suggest that the middle ear is a sensitive target for thyroid hormone in development.


Asunto(s)
Oído Medio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Receptores de Hormona Tiroidea/fisiología , Animales , Femenino , Audición , Hibridación in Situ , Yunque/embriología , Masculino , Martillo/embriología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Receptores de Tirotropina/fisiología , Estribo/embriología , Receptores alfa de Hormona Tiroidea/fisiología , Receptores beta de Hormona Tiroidea/fisiología
17.
Proc Natl Acad Sci U S A ; 108(42): 17462-7, 2011 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-21987803

RESUMEN

Mutations in the ligand-binding domain of the thyroid hormone receptor ß (TRß) lead to resistance to thyroid hormone (RTH). These TRß mutants function in a dominant-negative fashion to interfere with the transcription activity of wild-type thyroid hormone receptors (TRs), leading to dysregulation of the pituitary-thyroid axis and resistance in peripheral tissues. The molecular mechanism by which TRß mutants cause RTH has been postulated to be an inability of the mutants to properly release the nuclear corepressors (NCORs), thereby inhibiting thyroid hormone (TH)-mediated transcription activity. To test this hypothesis in vivo, we crossed Thrb(PV) mice (a model of RTH) expressing a human TRß mutant (PV) with mice expressing a mutant Ncor1 allele (Ncor1(ΔID) mice) that cannot recruit a TR or a PV mutant. Remarkably, in the presence of NCOR1ΔID, the abnormally elevated thyroid-stimulating hormone and TH levels found in Thrb(PV) mice were modestly but significantly corrected. Furthermore, thyroid hyperplasia, weight loss, and other hallmarks of RTH were also partially reverted in mice expressing NCOR1ΔID. Taken together, these data suggest that the aberrant recruitment of NCOR1 by RTH TRß mutants leads to clinical RTH in humans. The present study suggests that therapies aimed at the TR-NCOR1 interaction or its downstream actions could be tested as potential targets in treating RTH.


Asunto(s)
Co-Represor 1 de Receptor Nuclear/fisiología , Síndrome de Resistencia a Hormonas Tiroideas/genética , Síndrome de Resistencia a Hormonas Tiroideas/fisiopatología , Animales , Modelos Animales de Enfermedad , Genes erbA , Humanos , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Mutación , Co-Represor 1 de Receptor Nuclear/química , Co-Represor 1 de Receptor Nuclear/genética , Estructura Terciaria de Proteína , Eliminación de Secuencia , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/fisiología , Síndrome de Resistencia a Hormonas Tiroideas/patología , Hormonas Tiroideas/sangre , Hormonas Tiroideas/fisiología
18.
Med Clin (Barc) ; 137(12): 551-4, 2011 Nov 12.
Artículo en Español | MEDLINE | ID: mdl-21703645

RESUMEN

BACKGROUND AND OBJECTIVE: Resistance to thyroid hormone (RTH) is a syndrome mostly caused by mutations in thyroid hormone receptor beta gen (THRB). We present five families with RTH phenotype. PATIENTS AND METHODS: THRB gene sequencing. In vitro studies to evaluate the mutants response to thyroid hormones and their dominant negative effect. Mechanism of resistance in patients with RTH without THRB mutations quantifying expression of regulator of calcineurin 2 (ZAKI4) and Kruppel-like factor 9 (BTEB) genes in patients fibroblast cultures. RESULTS: THRB mutations were found in three cases: R243Q, R320C, R429Q. Mutants showed a decreased response to T3. R243Q and R320C had a strong dominant negative effect. One subject without THRB mutation showed changes in ZAKI4 and BTEB expression similar to R320C and the other showed expression levels higher than normal controls. CONCLUSIONS: Three cases of RTH were caused by THRB heterozygous mutations but in two cases mutations were not found. THRB mutation carriers and one of the patients without mutations share a similar mechanism of resistance and in the other subject RTH is TRß independent.


Asunto(s)
Receptores beta de Hormona Tiroidea/genética , Síndrome de Resistencia a Hormonas Tiroideas/genética , Adulto , Células Cultivadas , Niño , Electroforesis en Gel de Poliacrilamida , Femenino , Fibroblastos/metabolismo , Genes Dominantes , Bocio/sangre , Bocio/genética , Heterocigoto , Humanos , Hipertiroidismo/sangre , Hipertiroidismo/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Musculares/metabolismo , Mutación Puntual , Receptores beta de Hormona Tiroidea/fisiología , Síndrome de Resistencia a Hormonas Tiroideas/sangre , Hormonas Tiroideas/sangre
19.
Exp Clin Endocrinol Diabetes ; 119(2): 81-5, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20827662

RESUMEN

CONTEXT: Thyroid hormone (TH) mediated changes in gene expression were thought to be primarily initiated by the nuclear TH receptor (TR) binding to a thyroid hormone response element in the promoter of target genes. A recently described extranuclear mechanism of TH action consists of the association of TH-liganded TRß with phosphatidylinositol 3-kinase (PI3K) in the cytosol and subsequent activation of the PI3K pathway. OBJECTIVE: The aim of this study was to examine the effect of TH, TRß and PI3K on stanniocalcin 1 (STC1) expression in human cells. DESIGN: We treated human skin fibroblasts with triiodothyronine (T3) in the absence or presence of the PI3K inhibitor LY294002, a dominant negative PI3K subunit, Δp85α, and the protein synthesis inhibitor cycloheximide (CHX). The role of the TRß was studied in cells from patients with resistance to thyroid hormone (RTH). STC-1 mRNA expression was measured by real-time PCR. RESULTS: We found an induction of STC1 by T3 in normal cells, but less in cells from subjects with RTH (2.7 ± 0.2 vs. 1.6 ± 0.04, P < 0.01). The effect of T3 was completely abrogated by blocking PI3K with LY294002 (3.9 ± 0.5 vs. 0.85 ± 0.5; P < 0.05) and greatly reduced after transfection of a dominant negative PI3K subunit, demonstrating dependency on the PI3K pathway. CONCLUSION: These results establish STC1 as a TH target gene in humans. Furthermore, we show that STC1 induction by TH depends on both TRß and PI3K activation.


Asunto(s)
Glicoproteínas/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Receptores beta de Hormona Tiroidea/agonistas , Triyodotironina/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/metabolismo , Humanos , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/fisiología , Síndrome de Resistencia a Hormonas Tiroideas/genética , Síndrome de Resistencia a Hormonas Tiroideas/metabolismo , Síndrome de Resistencia a Hormonas Tiroideas/patología , Activación Transcripcional/efectos de los fármacos
20.
Endocrinology ; 151(10): 5074-83, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20719854

RESUMEN

Brown adipose tissue (BAT) thermogenesis increases when uncoupling protein-1 (UCP1) is activated adrenergically and requires T3. In humans, UCP1 activation in BAT seems involved in body weight maintenance. BAT type 2 deiodinase (D2) increases in response to adrenergic agents, producing the T3 required for UCP1 expression. T3 actions are mediated by thyroid hormone nuclear T3 receptors (TR), TRα and TRß. Studies in mice suggest that TRß is required for UCP1 induction, whereas TRα regulates body temperature and adrenergic sensitivity. In the present study, we compare the effects of T3 vs. specific TRß1 and TRα1 agonists [GC-1 and CO23] on the adrenergic induction of UCP1 and D2 in cultured rat brown adipocytes. T3 and GC-1 produced similar increases on UCP1, whereas CO23 increased UCP1 only at high doses (50 nm). GC-1 at low doses (0.2-10 nm) was less potent than T3, increasing the adrenergic stimulation of D2 activity and mRNA. At higher doses, GC-1 further stimulated whereas T3 inhibited D2 activity but not D2 mRNA, suggesting posttranscriptional effects. CO23 had no effect on D2 activity but increased D2 mRNA. T3, GC-1, or CO23 by themselves did not increase UCP1 or D2 mRNA. High T3 doses shortened D2 half-life and increased D2 turnover via proteasome, whereas GC-1 did not change D2 stability. The α1- and α2-adrenergic D2 responses increased using high T3 doses. In summary, T3 increases the adrenergic stimulation of UCP1 and D2 expression mostly via the TRß1 isoform, and in brown adipocytes, D2 is protected from degradation by the action of T3 on TRß1.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Yoduro Peroxidasa/genética , Canales Iónicos/genética , Proteínas Mitocondriales/genética , Receptores beta de Hormona Tiroidea/fisiología , Acetatos/farmacología , Tejido Adiposo Pardo/efectos de los fármacos , Adrenérgicos/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Hidantoínas/farmacología , Yoduro Peroxidasa/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Norepinefrina/farmacología , Fenoles/farmacología , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores beta de Hormona Tiroidea/agonistas , Receptores beta de Hormona Tiroidea/metabolismo , Triyodotironina/farmacología , Proteína Desacopladora 1 , Yodotironina Deyodinasa Tipo II
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...