Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Clin Endocrinol (Oxf) ; 79(5): 595-605, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23834164

RESUMEN

Normal thyroid hormone (TH) metabolism and action require adequate cellular TH signalling. This entails proper function of TH transporters in the plasma membrane, intracellular deiodination of TH and action of the bioactive hormone T3 at its nuclear receptors (TRs). The present review summarizes the discoveries of different syndromes with reduced sensitivity at the cellular level. Mutations in the TH transporter MCT8 cause psychomotor retardation and abnormal thyroid parameters. Mutations in the SBP2 protein, which is required for normal deiodination, give rise to a multisystem disorder including abnormal thyroid function tests. Mutations in TRß1 are a well-known cause of resistance to TH with mostly a mild phenotype, while only recently, patients with mutations in TRα1 were identified. The latter patients have slightly abnormal TH levels, growth retardation and cognitive defects. This review will describe the mechanisms of disease, clinical phenotype, diagnostic testing and suggestions for treatment strategies for each of these syndromes.


Asunto(s)
Hormonas Tiroideas/metabolismo , Membrana Celular/metabolismo , Humanos , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Simportadores , Glándula Tiroides/metabolismo
3.
Endocrinology ; 158(10): 3307-3318, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28938430

RESUMEN

The thyroid hormone (TH) analog eprotirome (KB2115) was developed to lower cholesterol through selective activation of the TH receptor (TR) ß1 in the liver. Interestingly, eprotirome shows low uptake in nonhepatic tissues, explaining its lipid-lowering action without adverse extrahepatic thyromimetic effects. Clinical trials have shown marked decreases in serum cholesterol levels. We explored the transport of eprotirome across the plasma membrane by members of three TH transporter families: monocarboxylate transporters MCT8 and MCT10; Na-independent organic anion transporters 1A2, 1B1, 1B3, 1C1, 2A1, and 2B1; and Na-dependent organic anion transporters SLC10A1 to SLC10A7. Cellular transport was studied in transfected COS1 cells using [14C]eprotirome and [125I]TH analogs. Of the 15 transporters tested initially, the liver-specific bile acid transporter SLC10A1 showed the highest eprotirome uptake (greater than a sevenfold induction after 60 minutes) as well as TRß1-mediated transcriptional activity. Uptake of eprotirome by SLC10A1 was Na+ dependent and saturable with a Michaelis constant of 8 µM. Eprotirome transport was inhibited by known substrates for SLC10A1 (e.g., cholate and taurocholate), and by TH analogs such as triiodothyropropionic acid and triiodothyroacetic acid. However, no significant SLC10A1-mediated transport was observed of these [125I]TH analogs. We also studied the plasma disappearance and biliary excretion of [14C]eprotirome injected in control and Slc10a1 knockout mice. Although eprotirome is also transported by mouse Slc10a1, the pharmacokinetics of eprotirome were not affected by Slc10a1 deficiency. In conclusion, we have demonstrated that the liver-specific bile acid transporter SLC10A1 effectively transports eprotirome. However, Slc10a1 does not appear to be critical for the liver targeting of this TH analog in mice. Therefore, the importance of SLC10A1 for liver uptake of eprotirome in humans remains to be elucidated.


Asunto(s)
Anilidas/farmacología , Anilidas/farmacocinética , Anticolesterolemiantes , Hígado/efectos de los fármacos , Transportadores de Anión Orgánico Sodio-Dependiente/fisiología , Simportadores/fisiología , Animales , Transporte Biológico , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Humanos , Hígado/metabolismo , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/deficiencia , Transportadores de Anión Orgánico Sodio-Dependiente/genética , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sodio/farmacología , Simportadores/deficiencia , Simportadores/genética , Hormonas Tiroideas/metabolismo , Transfección
4.
Mol Cell Endocrinol ; 437: 252-260, 2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27492966

RESUMEN

Transport of thyroid hormone (TH) across the plasma membrane is necessary for the genomic action of T3 mediated by its nuclear T3 receptor. MCT8 and MCT10 have been identified as important TH transporters. Mutations in MCT8 result in severe psychomotor retardation. In addition to TH transport into the cell, MCT8 and MCT10 also facilitate TH efflux from cells. Therefore, the aim of this study was to examine if MCT8 and MCT10 increase the availability of T3 for its nuclear receptor rather than generate a rapid equilibrium between cellular and serum T3. T3 action was investigated in JEG3 cells co-transfected with TRß1 and a T3 response element-driven luciferase construct, and T3 metabolism was analyzed in cells transfected with type 3 deiodinase (D3). In addition, cells were transfected with MCT8 or MCT10 and/or the cytoplasmic T3-binding protein mu-crystallin (CRYM). Luciferase signal was markedly stimulated by incubating cells for 24 h with 1 nM T3, but this response was not augmented by MCT8 or MCT10 expression. Limiting the time of T3 exposure to 1-6 h and co-transfection with CRYM allowed for a modest increase in luciferase response to T3. In contrast, T3 metabolism by D3 was potently stimulated by MCT8 or MCT10 expression, but it was not affected by expression of CRYM. These results suggest that MCT8 and MCT10 by virtue of their bidirectional T3 transport have less effect on steady-state nuclear T3 levels than on T3 levels at the cell periphery where D3 is located. CRYM alters the dynamics of cellular TH transport but its exact function in the cellular distribution of TH remains to be determined.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Núcleo Celular/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Triyodotironina/farmacología , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Genes Reporteros , Humanos , Modelos Biológicos , Simportadores , Transfección , Cristalinas mu
5.
Eur Thyroid J ; 3(1): 17-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24847461

RESUMEN

Thyroid hormone (TH) exerts its biological activity via the TH receptors TRα1 and TRß1/2, which are encoded by the THRA and THRB genes. The first patients with mutations in THRB were identified decades ago. These patients had a clinical syndrome of resistance to TH associated with high serum TH and nonsuppressed thyroid-stimulating hormone levels. Until recently, no patients with mutations in THRA had been identified. In an attempt to predict the clinical phenotype of such patients, different TRα1 mutant mouse models have been generated. These mice have a variable phenotype depending on the location and severity of the mutation. Recently, the first humans with mutations in THRA were identified. Their phenotype consists of relatively low serum T4 and high serum T3 levels (and thus an elevated T3/T4 ratio), growth retardation, delayed mental and bone development, and constipation. While, in retrospect, certain features present in humans can also be found in mouse models, the first humans carrying a defect in TRα1 were not suspected of having a THRA gene mutation initially. The current review focuses on the clinical consequences of TRα1 mutations.

6.
Horm Res Paediatr ; 82(4): 261-71, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25247785

RESUMEN

BACKGROUND/AIMS: Monocarboxylate transporter 8 (MCT8) is essential for thyroid hormone (TH) transport in the brain. Mutations in MCT8 are associated with the Allan-Herndon-Dudley syndrome (AHDS), characterized by severe psychomotor retardation and altered serum thyroid parameters. Here we report two novel mutations in MCT8 and discuss the clinical findings. CASE REPORT AND RESULTS: We describe 4 males with AHDS from two unrelated families varying in age from 1.5 to 11 years. All 4 patients presented with typical clinical signs of AHDS, including severe psychomotor retardation, axial hypotonia, lack of speech, diminished muscle mass, increased muscle tone, hyperreflexia, myopathic facies, high T3, low T4 and rT3, and normal/mildly elevated TSH levels. Comparison of patients at different ages suggests the progressive nature of AHDS. Genetic analyses identified a novel missense MCT8 mutation (p.G495A) in family 1 and a 2.8-kb deletion comprising exons 3 and 4 in family 2. Functional analysis of p.G495A revealed impaired TH transport varying from 20 to 85% depending on the cell context. CONCLUSION: Here we report 4 AHDS patients in unrelated Turkish families harboring novel MCT8 mutations. Despite the widely different mutations, the clinical phenotypes are very similar and findings support the progressive nature of AHDS.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X/genética , Transportadores de Ácidos Monocarboxílicos/genética , Hipotonía Muscular/genética , Atrofia Muscular/genética , Niño , Preescolar , Análisis Mutacional de ADN , Eliminación de Gen , Humanos , Lactante , Discapacidad Intelectual/etiología , Discapacidad Intelectual/genética , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/psicología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Hipotonía Muscular/metabolismo , Hipotonía Muscular/psicología , Atrofia Muscular/metabolismo , Atrofia Muscular/psicología , Mutación/genética , Mutación Missense/genética , Linaje , Simportadores , Hormonas Tiroideas/sangre
7.
Mol Endocrinol ; 27(4): 586-97, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23518925

RESUMEN

The impact of thyroid hormone (TH) on metabolism and energy expenditure is well established, but the role of TH in regulating nutritional sensing, particularly in the central nervous system, is only poorly defined. Here, we studied the consequences of hypothyroidism on leptin production as well as leptin sensing in congenital hypothyroid TRH receptor 1 knockout (Trhr1 ko) mice and euthyroid control animals. Hypothyroid mice exhibited decreased circulating leptin levels due to a decrease in fat mass and reduced leptin expression in white adipose tissue. In neurons of the hypothalamic arcuate nucleus, hypothyroid mice showed increased leptin receptor Ob-R expression and decreased suppressor of cytokine signaling 3 transcript levels. In order to monitor putative changes in central leptin sensing, we generated hypothyroid and leptin-deficient animals by crossing hypothyroid Trhr1 ko mice with the leptin-deficient ob/ob mice. Hypothyroid Trhr1/ob double knockout mice showed a blunted response to leptin treatment with respect to body weight and food intake and exhibited a decreased activation of phospho-signal transducer and activator of transcription 3 as well as a up-regulation of suppressor of cytokine signaling 3 upon leptin treatment, particularly in the arcuate nucleus. These data indicate alterations in the intracellular processing of the leptin signal under hypothyroid conditions and thereby unravel a novel mode of action by which TH affects energy metabolism.


Asunto(s)
Hipotálamo/metabolismo , Hipotiroidismo/metabolismo , Leptina/metabolismo , Transducción de Señal , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/patología , Hipotálamo/fisiopatología , Hipotiroidismo/sangre , Hipotiroidismo/fisiopatología , Leptina/deficiencia , Leptina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno/efectos de los fármacos , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Leptina/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Hormonas Tiroideas/sangre
8.
J Clin Endocrinol Metab ; 98(7): 3029-38, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23633213

RESUMEN

CONTEXT: Recently the first patients with inactivating mutations in T3 receptor (TR)-α1 have been identified. These patients have low free T4, low T4, high T3, low rT3, and normal TSH serum levels, in combination with growth retardation, delayed bone development, and constipation. OBJECTIVE: The aim of the current study was to report the effects of levothyroxine (LT4) treatment on the clinical phenotype of 2 patients (father and daughter) with a heterozygous inactivating mutation in TRα1. SETTING AND PARTICIPANTS: Both patients were treated with LT4 for the last 5 years. To evaluate the effect of LT4 treatment, LT4 was withdrawn for 35 days and subsequently reinitiated. Data were collected from medical records, by reanalysis of serum collected over the last 6 years, and by a detailed clinical evaluation. RESULTS: Treatment with LT4 resulted in a suppression of serum TSH and normalization of serum free T4 and rT3, whereas T3 levels remained elevated in both patients. In addition, there was a normalization of the dyslipidemia as well as a response in serum IGF-I, SHBG, and creatine kinase in the index patient. All these parameters returned to pretreatment values when LT4 was briefly stopped. LT4 also resulted in an improvement of certain clinical features, such as constipation and nerve conductance. However, cognitive and fine motor skill defects remained. CONCLUSION: This study reports the consequences of LT4 treatment over a prolonged period of time in 2 of the first patients with a heterozygous mutation in TRα1. LT4 therapy leads to an improvement of certain but not all features of the clinical phenotype.


Asunto(s)
Mutación del Sistema de Lectura , Receptores alfa de Hormona Tiroidea/genética , Síndrome de Resistencia a Hormonas Tiroideas/tratamiento farmacológico , Síndrome de Resistencia a Hormonas Tiroideas/genética , Tiroxina/uso terapéutico , Niño , Estreñimiento/etiología , Estreñimiento/prevención & control , Resistencia a Medicamentos , Dislipidemias/etiología , Dislipidemias/prevención & control , Femenino , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Conducción Nerviosa/efectos de los fármacos , Receptores alfa de Hormona Tiroidea/metabolismo , Síndrome de Resistencia a Hormonas Tiroideas/metabolismo , Síndrome de Resistencia a Hormonas Tiroideas/fisiopatología , Hormonas Tiroideas/sangre , Resultado del Tratamiento
9.
Mol Cell Endocrinol ; 337(1-2): 96-100, 2011 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-21315799

RESUMEN

Thyroid hormone (TH) transporter proteins mediate transport of TH across the plasma membrane, thereby facilitating its intracellular bioavailability. As only a few transporters have been identified which are relatively specific for TH, including monocarboxylate transporter (MCT) 8 and MCT10, the need for identification of novel specific TH transporters is obvious. A possible strategy to identify TH transporters is their modification with a ligand-derived affinity-label and subsequent identification by mass spectrometry. Previously, N-bromoacetyl (BrAc)-iodothyronines have been reported as useful affinity-labels for human (h) MCT8. In the present study we reinvestigated possible BrAc[(125)I]T3-labeling of hMCT8 and hMCT10. The present study demonstrates that hMCT8 and hMCT10 both facilitate BrAc[(125)I]T3 transport, but are not labeled by BrAc[(125)I]T3. We provide evidence that human protein disulfide isomerase, which molecular mass is similar to hMCT8, is labeled by BrAc[(125)I]T3. In addition, differential inhibitory effects were observed of iodothyronines derivatives with different side chains on T3 transport by hMCT8 and hMCT10. In conclusion, we demonstrated that not hMCT8 and hMCT10, but human protein disulfide isomerase, is labeled by BrAc[(125)I]T3. The usefulness of BrAc[(125)I]T3 as a tool for the identification of novel TH transporters remains to be explored.


Asunto(s)
Marcadores de Afinidad/farmacología , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Recombinantes/metabolismo , Triyodotironina/análogos & derivados , Marcadores de Afinidad/farmacocinética , Animales , Células COS , Chlorocebus aethiops , Humanos , Yoduro Peroxidasa/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Ratas , Simportadores , Triyodotironina/farmacocinética , Triyodotironina/farmacología
10.
Mol Cell Endocrinol ; 315(1-2): 138-45, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19682536

RESUMEN

Transport of (sulfated) iodothyronines across the plasma membrane is required for their intracellular metabolism. Rat Na(+)/taurocholate cotransporting polypeptide (Ntcp; Slc10a1) has been identified as an important transporter protein. We demonstrate that among the 7 members of the solute carrier family SLC10, only human SLC10A1 mediates sodium-dependent transport of the iodothyronine T4 and iodothyronine sulfates T3S and T4S. In contrast to SLC10A2-7, cells co-expressing SLC10A1 and the deiodinase D1 demonstrate a dramatic increase in T3S and T4S metabolism. The SLC10A1 substrates taurocholate, DHEAS and E3S inhibit T3S and T4S transport. Furthermore, co-transfection of SLC10A1 with CRYM, a well-known intracellular iodothyronine-binding protein, results in an enhanced intracellular accumulation of T3S and T4S, indicating that CRYM binds iodothyronine sulfates. The present findings indicate that the liver-specific transporter SLC10A1 transports (sulfated) iodothyronines, thereby increasing their intracellular availability. Therefore, SLC10A1 may fulfill a critical step in providing liver D1 with iodothyronine sulfates for rapid degradation.


Asunto(s)
Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Simportadores/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Transporte Biológico/fisiología , Células COS , Línea Celular , Chlorocebus aethiops , Cristalinas/genética , Cristalinas/metabolismo , Sulfato de Deshidroepiandrosterona/metabolismo , Humanos , Yoduro Peroxidasa/metabolismo , Radioisótopos de Yodo/química , Radioisótopos de Yodo/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Simportadores/genética , Hormonas Tiroideas/química , Cristalinas mu
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA