Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 903
Filtrar
3.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38609183

RESUMEN

Elevated ER stress has been linked to the pathogenesis of several disease conditions including neurodegeneration. In this study, we have holistically determined the differential expression of all the nuclear receptors (NRs) in the presence of classical ER stress inducers. Activation of Nr1h4 and Thrb by their cognate ligands (GW4064 and T3) ameliorates the tunicamycin (TM)-induced expression of ER stress genes. A combination of both ligands is effective in mitigating cell death induced by TM. Further exploration of their protective effects in the Parkinson's disease (PD) model shows that they reduce MPP+-induced dissipation of mitochondrial membrane potential and ROS generation in an in vitro PD model in neuronal cells. Furthermore, the generation of an experimental murine PD model reveals that simultaneous treatment of GW4064 and T3 protects mice from ER stress, dopaminergic cell death, and functional deficits in the MPTP mouse model of PD. Thus, activation of Nr1h4 and Thrb by their respective ligands plays an indispensable role in ER stress amelioration and mounts protective effects in the MPTP mouse model of PD.


Asunto(s)
Enfermedad de Parkinson , Animales , Ratones , Muerte Celular , Modelos Animales de Enfermedad , Dopamina , Neuronas Dopaminérgicas , Receptores beta de Hormona Tiroidea
4.
Exp Cell Res ; 437(2): 114017, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38555013

RESUMEN

Thyroid hormone receptor ß (THRß) is a member of the nuclear receptor superfamily of ligand-modulated transcription factors. Upon ligand binding, THRß sequentially recruits the components of transcriptional machinery to modulate target gene expression. In addition to regulating diverse physiological processes, THRß plays a crucial role in hypothalamus-pituitary-thyroid axis feedback regulation. Anomalies in THRß gene/protein structure are associated with onset of diverse disease states. In this study, we investigated disease-inflicting truncated variants of THRß using in-silico analysis and cell-based assays. We examined the THRß truncated variants on multiple test parameters, including subcellular localization, ligand-receptor interactions, transcriptional functions, interaction with heterodimeric partner RXR, and receptor-chromatin interactions. Moreover, molecular dynamic simulation approaches predicted that shortened THRß-LBD due to point mutations contributes proportionally to the loss of structural integrity and receptor stability. Deviant subcellular localization and compromised transcriptional function were apparent with these truncated variants. Present study shows that 'mitotic bookmarking' property of some THRß variants is also affected. The study highlights that structural and conformational attributes of THRß are necessary for normal receptor functioning, and any deviations may contribute to the underlying cause of the inflicted diseases. We anticipate that insights derived herein may contribute to improved mechanistic understanding to assess disease predisposition.


Asunto(s)
Receptores beta de Hormona Tiroidea , Factores de Transcripción , Receptores beta de Hormona Tiroidea/genética , Ligandos , Factores de Transcripción/genética , Mutación Puntual , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo
6.
N Engl J Med ; 390(6): 497-509, 2024 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-38324483

RESUMEN

BACKGROUND: Nonalcoholic steatohepatitis (NASH) is a progressive liver disease with no approved treatment. Resmetirom is an oral, liver-directed, thyroid hormone receptor beta-selective agonist in development for the treatment of NASH with liver fibrosis. METHODS: We are conducting an ongoing phase 3 trial involving adults with biopsy-confirmed NASH and a fibrosis stage of F1B, F2, or F3 (stages range from F0 [no fibrosis] to F4 [cirrhosis]). Patients were randomly assigned in a 1:1:1 ratio to receive once-daily resmetirom at a dose of 80 mg or 100 mg or placebo. The two primary end points at week 52 were NASH resolution (including a reduction in the nonalcoholic fatty liver disease [NAFLD] activity score by ≥2 points; scores range from 0 to 8, with higher scores indicating more severe disease) with no worsening of fibrosis, and an improvement (reduction) in fibrosis by at least one stage with no worsening of the NAFLD activity score. RESULTS: Overall, 966 patients formed the primary analysis population (322 in the 80-mg resmetirom group, 323 in the 100-mg resmetirom group, and 321 in the placebo group). NASH resolution with no worsening of fibrosis was achieved in 25.9% of the patients in the 80-mg resmetirom group and 29.9% of those in the 100-mg resmetirom group, as compared with 9.7% of those in the placebo group (P<0.001 for both comparisons with placebo). Fibrosis improvement by at least one stage with no worsening of the NAFLD activity score was achieved in 24.2% of the patients in the 80-mg resmetirom group and 25.9% of those in the 100-mg resmetirom group, as compared with 14.2% of those in the placebo group (P<0.001 for both comparisons with placebo). The change in low-density lipoprotein cholesterol levels from baseline to week 24 was -13.6% in the 80-mg resmetirom group and -16.3% in the 100-mg resmetirom group, as compared with 0.1% in the placebo group (P<0.001 for both comparisons with placebo). Diarrhea and nausea were more frequent with resmetirom than with placebo. The incidence of serious adverse events was similar across trial groups: 10.9% in the 80-mg resmetirom group, 12.7% in the 100-mg resmetirom group, and 11.5% in the placebo group. CONCLUSIONS: Both the 80-mg dose and the 100-mg dose of resmetirom were superior to placebo with respect to NASH resolution and improvement in liver fibrosis by at least one stage. (Funded by Madrigal Pharmaceuticals; MAESTRO-NASH ClinicalTrials.gov number, NCT03900429.).


Asunto(s)
Cirrosis Hepática , Enfermedad del Hígado Graso no Alcohólico , Piridazinas , Uracilo , Adulto , Humanos , Método Doble Ciego , Hígado/diagnóstico por imagen , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/patología , Piridazinas/uso terapéutico , Resultado del Tratamiento , Uracilo/análogos & derivados , Receptores beta de Hormona Tiroidea/agonistas , Biopsia , Relación Dosis-Respuesta a Droga
8.
Mil Med ; 189(1-2): e439-e442, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-37531455

RESUMEN

Clinicians may confuse an impaired sensitivity to thyroid hormone with hyperthyroidism and offer an inappropriate treatment. We report a diagnosis of resistance to thyroid hormone (RTH) caused by a rare mutation in the thyroid hormone receptor beta gene in a patient previously presumed to have Graves' disease. We have found only one published case of a novel point mutation, c.749T>C (p.Ile250Thr variant) associated with 50% reduction in thyroid hormone receptor binding affinity for triiodothyronine in the I250T mutant; it was found in this patient. A 66-year-old male veteran, with a history of non-ischemic cardiomyopathy and arrhythmias, was referred by a cardiologist with concerns for a possible thyrotropin (TSH) adenoma on account of elevated TSH and free thyroxine (FT4) levels. Pituitary imaging was negative. He was previously treated with radioiodine for presumptive Graves' disease in the civilian sector. Examination revealed a goiter with no nodules. Repeat TSH and FT4 levels were elevated and also free triiodothyronine (FT3) and reverse triiodothyronine. These findings and other test results were consistent with RTH, which was confirmed by genetic testing. Mutation analysis showed the patient to be heterozygous for the p.Ile250Thr variant. He later developed hypothyroidism. Resistance to thyroid hormone can be misdiagnosed as hyperthyroidism with consequent inappropriate treatment. Treatment is not needed in most RTH-beta patients. Thyroid ablation should generally be avoided. Clinicians must be cautious whenever they encounter concurrent elevation of TSH, FT4, and FT3. This RTH-beta patient has a rare I250T mutant of the thyroid hormone receptor beta gene, the second reported case in the literature.


Asunto(s)
Enfermedad de Graves , Hipertiroidismo , Síndrome de Resistencia a Hormonas Tiroideas , Masculino , Humanos , Anciano , Triyodotironina , Receptores beta de Hormona Tiroidea/genética , Radioisótopos de Yodo , Hormonas Tiroideas , Enfermedad de Graves/diagnóstico , Enfermedad de Graves/genética , Enfermedad de Graves/complicaciones , Tirotropina , Mutación , Hipertiroidismo/genética , Hipertiroidismo/complicaciones , Síndrome de Resistencia a Hormonas Tiroideas/diagnóstico , Síndrome de Resistencia a Hormonas Tiroideas/genética , Síndrome de Resistencia a Hormonas Tiroideas/complicaciones
9.
Stem Cell Res ; 74: 103275, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38100912

RESUMEN

THRB is a nuclear receptor, regulating gene expression dependent on thyroid hormone (TH) binding. The same receptor mediates signaling pathway activation in the cytosol. The challenge is to distinguish which of the two mechanisms is responsible for physiological effects of TH. We established an iPSC cell line with two mutations (E125G_G126S) in the THRB DNA-binding domain, which abrogates nuclear action and, thus, allows to study signaling pathway activation exclusively. We also generated a THRB knockout cell line to abolish all THRB effects. Comparison of WT and these two cell lines allows attribution of thyroid hormone effects to the underlying mechanism.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Hormonas Tiroideas , Transducción de Señal , Mutación/genética , Línea Celular , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo
10.
Commun Biol ; 6(1): 1253, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-38081939

RESUMEN

Thyroid hormone (3,5,3'-triiodothyronine, T3) is a key regulator of pituitary gland function. The response to T3 is thought to hinge crucially on interactions of nuclear T3 receptors with enhancers but these sites in pituitary chromatin remain surprisingly obscure. Here, we investigate genome-wide receptor binding in mice using tagged endogenous thyroid hormone receptor ß (TRß) and analyze T3-regulated open chromatin using an anterior pituitary-specific Cre driver (Thrbb2Cre). Strikingly, T3 regulates histone modifications and chromatin opening primarily at sites that maintain TRß binding regardless of T3 levels rather than at sites where T3 abolishes or induces de novo binding. These sites associate more frequently with T3-activated than T3-suppressed genes. TRß-deficiency blunts T3-regulated gene expression, indicating that TRß confers transcriptional sensitivity. We propose a model of gene activation in which poised receptor-enhancer complexes facilitate adjustable responses to T3 fluctuations, suggesting a genomic basis for T3-dependent pituitary function or pituitary dysfunction in thyroid disorders.


Asunto(s)
Cromatina , Hormonas Tiroideas , Ratones , Animales , Cromatina/genética , Cromatina/metabolismo , Hormonas Tiroideas/metabolismo , Triyodotironina/farmacología , Triyodotironina/metabolismo , Hipófisis/metabolismo , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo
11.
J Cell Biochem ; 124(12): 1948-1960, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37992217

RESUMEN

Thyroid hormones (TH) are important modulators of bone remodeling and thus, thyroid diseases, in particular hyperthyroidism, are able to compromise bone quality and fracture resistance. TH actions on bone are mediated by the thyroid hormone receptors (TR) TRα1 and TRß1, encoded by Thra and Thrb, respectively. Skeletal phenotypes of mice lacking Thra (Thra0/0 ) and Thrb (Thrb-/- ) are well-described and suggest that TRα1 is the predominant mediator of TH actions in bone. Considering that bone cells might be affected by systemic TH changes seen in these mutant mice, here we investigated the effects of TR knockout on osteoblasts exclusively at the cellular level. Primary osteoblasts obtained from Thra0/0 , Thrb-/- , and respective wildtype (WT) mice were analyzed regarding their differentiation potential, activity and TH responsiveness in vitro. Thra, but not Thrb knockout promoted differentiation and activity of early, mature and late osteoblasts as compared to respective WT cells. Interestingly, while mineralization capacity and expression of osteoblast marker genes and TH target gene Klf9 was increased by TH in WT and Thra-deficient osteoblasts, Thrb knockout mitigated the responsiveness of osteoblasts to short (48 h) and long term (10 d) TH treatment. Further, we found a low ratio of Rankl, a potent osteoclast stimulator, over osteoprotegerin, an osteoclast inhibitor, in Thrb-deficient osteoblasts and in line, supernatants obtained from Thrb-/- osteoblasts reduced numbers of primary osteoclasts in vitro. In accordance to the increased Rankl/Opg ratio in TH-treated WT osteoblasts only, supernatants from these cells, but not from TH-treated Thrb-/- osteoblasts increased the expression of Trap and Ctsk in osteoclasts, suggesting that osteoclasts are indirectly stimulated by TH via TRß1 in osteoblasts. In conclusion, our study shows that both Thra and Thrb differentially affect activity, differentiation and TH response of osteoblasts in vitro and emphasizes the importance of TRß1 to mediate TH actions in bone.


Asunto(s)
Receptores de Hormona Tiroidea , Receptores alfa de Hormona Tiroidea , Ratones , Animales , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Receptores alfa de Hormona Tiroidea/genética , Receptores alfa de Hormona Tiroidea/metabolismo , Hormonas Tiroideas/metabolismo , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Biología , Ligando RANK/metabolismo , Ratones Noqueados
12.
Nat Commun ; 14(1): 6742, 2023 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-37875497

RESUMEN

It is well established that maternal thyroid hormones play an important role for the developing fetus; however, the consequences of maternal hyperthyroidism for the offspring remain poorly understood. Here we show in mice that maternal 3,3',5-triiodothyronine (T3) treatment during pregnancy leads to improved glucose tolerance in the adult male offspring and hyperactivity of brown adipose tissue (BAT) thermogenesis in both sexes starting early after birth. The activated BAT provides advantages upon cold exposure, reducing the strain on other thermogenic organs like muscle. This maternal BAT programming requires intact maternal thyroid hormone receptor ß (TRß) signaling, as offspring of mothers lacking this receptor display the opposite phenotype. On the molecular level, we identify distinct T3 induced alterations in maternal serum metabolites, including choline, a key metabolite for healthy pregnancy. Taken together, our results connect maternal TRß activation to the fetal programming of a thermoregulatory phenotype in the offspring.


Asunto(s)
Tejido Adiposo Pardo , Receptores beta de Hormona Tiroidea , Embarazo , Femenino , Ratones , Animales , Masculino , Tejido Adiposo Pardo/metabolismo , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo , Triyodotironina/metabolismo , Termogénesis/fisiología , Hormonas Tiroideas/metabolismo
13.
Environ Int ; 181: 108256, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37862862

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) are a group of xenobiotics that are widely distributed throughout the aquatic environment. Many PFAS are possible thyroid hormone (TH) system disrupting compounds, because they have the capacity to -amongst other- inhibit the TH thyroxine (T4) from binding to its transport protein transthyretin (TTR). This study investigated the occurrence of TH-displacing activity in the Dutch water cycle, and more specifically, the contribution of PFAS to this effect. Over one year of monitoring data of 29 PFAS (linear and branched) showed the continuous presence of PFAS in drinking waters and their surface water sources. Secondly, the FITC-T4 and TTR-TRß-CALUX bioassays were mutually compared using positive (HPLC-grade water spiked with PFOA) and negative control samples (HPLC-grade water), as well as relative potency factors (RPFs) of up to 20 PFAS congeners. Both assays were found to be suitable for measuring TH-displacing activity in water samples. As a third aim, a field study was performed in the Dutch water cycle that was comprised of samples from drinking water, surface water, PFAS contaminated sites, and 2 wastewater treatment plants. All samples were analyzed with 1. chemical analysis for 29 PFAS, 2. the FITC-T4 bioassay, and 3. the TTR-TRß-CALUX bioassay. The bioassays mutually showed good correlation (R2 0.85). Bioanalytical equivalent concentrations (BEQ) based on chemically-determined concentrations and RPFs (BEQchem) revealed that analyzed PFAS only explained ≤4.1 % of their activity in water extracts measured by both bioassays (BEQbio). This indicated that as yet unknown compounds contribute to the majority of the measured TH-displacing activity. Moreover, water treatment processes (e.g. DW production from SW) showed a larger contribution of target PFAS to the BEQbio. This could be a first lead to identify unknown compounds that contribute to this activity, and as such, enable the assessment of possible risks associated by the occurrence of TH-displacing activity in water.


Asunto(s)
Agua Potable , Fluorocarburos , Contaminantes Químicos del Agua , Fluoresceína-5-Isotiocianato , Hormonas Tiroideas , Glándula Tiroides , Bioensayo , Receptores beta de Hormona Tiroidea , Contaminantes Químicos del Agua/toxicidad
14.
Endocrinology ; 164(10)2023 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-37702560

RESUMEN

Thyroid hormone receptor beta (TRß) is a recognized tumor suppressor in numerous solid cancers. The molecular signaling of TRß has been elucidated in several cancer types through re-expression models. Remarkably, the potential impact of selective activation of endogenous TRß on tumor progression remains largely unexplored. We used cell-based and in vivo assays to evaluate the effects of the TRß agonist sobetirome (GC-1) on a particularly aggressive and dedifferentiated cancer, anaplastic thyroid cancer (ATC). Here we report that GC-1 reduced the tumorigenic phenotype, decreased cancer stem-like cell populations, and induced redifferentiation of the ATC cell lines with different mutational backgrounds. Of note, this selective activation of TRß amplified the effects of therapeutic agents in blunting the aggressive cell phenotype and stem cell growth. In xenograft assays, GC-1 alone inhibited tumor growth and was as effective as the kinase inhibitor, sorafenib. These results indicate that selective activation of TRß not only induces a tumor suppression program de novo but enhances the effectiveness of anticancer agents, revealing potential novel combination therapies for ATC and other aggressive solid tumors.


Asunto(s)
Carcinoma Anaplásico de Tiroides , Neoplasias de la Tiroides , Femenino , Humanos , Animales , Ratones , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Receptores beta de Hormona Tiroidea , Agresión , Neoplasias de la Tiroides/tratamiento farmacológico
16.
Thyroid ; 33(10): 1140-1149, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37594753

RESUMEN

Background: Thyroid hormone (TH) has actions in every tissue of the body and is essential for normal development, as well as having important actions in the adult. The earliest markers of TH action that were identified and monitored clinically, even before TH could be measured in serum, included oxygen consumption, basal metabolic rate, serum cholesterol, and deep tendon reflex time. Cellular, rodent, amphibian, zebrafish, and human models have been used to study TH action. Summary: Early studies of the mechanism of TH action focused on saturable-specific triiodothyronine (T3) nuclear binding and direct actions of T3 that altered protein expression. Additional effects of TH were recognized on mitochondria, stimulation of ion transport, especially the sodium potassium ATPase, augmentation of adrenergic signaling, role as a neurotransmitter, and direct plasma membrane effects. The cloning of the thyroid hormone receptor (THR) genes in 1986 and report of the THR crystal structure in 1995 produced rapid progress in understanding the mechanism of TH nuclear action, as well as the development of modified THR ligands. These findings revealed nuances of TH signaling, including the role of nuclear receptor coactivators and corepressors, repression of positively stimulated genes by the unliganded receptor, THR isoform-specific actions of TRα (THRA) and TRß (THRB), and THR binding DNA as a heterodimer with retinoid-x-receptor (RXR) for genes positively regulated by TH. The identification of genetic disorders of TH transport and signaling, especially Resistance to Thyroid Hormone (RTH) and monocarboxylate transporter 8 (Mct8) defects, has been highly informative with respect to the mechanism of TH action. Conclusions: The impact of THR isoform, post-translational modifications, receptor cofactors, DNA response element, and selective TH tissue uptake, on TH action, have clinical implications for diagnosing and treating thyroid disease. Additionally, these findings have led to the development of novel TH and TH analogue therapies for metabolic, neurological, and cardiovascular diseases.


Asunto(s)
Receptores beta de Hormona Tiroidea , Pez Cebra , Animales , Adulto , Humanos , Pez Cebra/genética , Receptores beta de Hormona Tiroidea/genética , Hormonas Tiroideas/metabolismo , Triyodotironina/farmacología , ADN , Isoformas de Proteínas , Receptores de Hormona Tiroidea/genética
17.
J Clin Endocrinol Metab ; 108(10): e944-e948, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37149816

RESUMEN

CONTEXT: Pregnant women with mutations in the thyroid hormone receptor beta (THRB) gene expose their fetuses to high thyroid hormone (TH) levels shown to be detrimental to a normal fetus (NlFe) but not to an affected fetus (AfFe). However, no information is available about differences in placental TH regulators. OBJECTIVE: To investigate whether there are differences in placentas associated with a NlFe compared with an AfFe, we had the unique opportunity to study placentas from 2 pregnancies of the same woman with THRB mutation G307D. One placenta supported a NlFe while the other an AfFe. METHODS: Sections of placentas were collected and frozen at -80 °C after term delivery of a NlFe and an AfFe. Two placentas from healthy women of similar gestational age were also obtained. The fetal origin of the placental tissues was established by gDNA quantitation of genes on the X and Y chromosomes and THRB gene. Expression and enzymatic activity of deiodinases 2 and 3 were measured. Expression of following genes was also quantitated: MCT10, MCT8, LAT1, LAT2, THRB, THRA. RESULTS: The placenta carrying the AfFe exhibited a significant reduction of deiodinase 2 and 3 activities as well as the expression of the TH transporters MCT10, LAT1 and LAT2, and THRA. CONCLUSION: We present the first study of the effect of the fetal THRB genotype on the placenta. Though limited by virtue of the rarity of THRB mutations and sample availability, we show that the fetal THRB genotype influences the levels of TH regulators in the placenta.


Asunto(s)
Genes erbA , Placenta , Femenino , Embarazo , Humanos , Placenta/metabolismo , Receptores beta de Hormona Tiroidea , Hormonas Tiroideas/metabolismo , Feto/metabolismo , Genotipo
18.
Environ Pollut ; 330: 121745, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37127237

RESUMEN

Thyroid-disrupting chemicals (TDCs) have received increasing concerns because of their negative health impacts on both wildlife and humans. This study aimed to develop in vitro screening assays for TDCs based on thyroid hormone receptor ß (TRß) and transthyretin (TTR) proteins. Firstly, the recombinant ligand-binding domain of TRß (TRß-LBD) and TTR proteins of zebrafish were produced by eukaryotic expression system and then used as bio-recognition components to construct electrochemical biosensors. In the biosensors, the supported bilayer lipid membrane (s-BLM) was used as a matrix to immobilize proteins, and gold nanoflowers (AuNFs) were used to improve the sensitivity by increasing electroactive surface area. Under the optimizing conditions, the zfTRß-LBD/AuNFs/s-BLM/GCE biosensor had a detection range of 0.23 nM-1.92 µM and a detection limit of 0.07 nM for triiodothyronine (T3), while the zfTTR/AuNFs/s-BLM/GCE biosensor had a detection range of 0.46 nM-3.84 µM, with a detection limit of 0.13 nM. Based on the constructed biosensors, the order of T3 equivalent concentrations of bisphenols was BPA ≈ BPS > BPF > BPAF ≈ BPAP > BPZ, which was similar to the results of recombinant TRß two-hybrid yeast assay. Furthermore, the reliability of the biosensors was validated by molecular docking, in which BPA and BPS showed higher binding affinity to zfTRß-LBD. Therefore, this study provided a valuable tool for efficiently screening TDCs.


Asunto(s)
Glándula Tiroides , Pez Cebra , Animales , Humanos , Glándula Tiroides/metabolismo , Pez Cebra/metabolismo , Receptores beta de Hormona Tiroidea/metabolismo , Prealbúmina/metabolismo , Simulación del Acoplamiento Molecular , Reproducibilidad de los Resultados
19.
Front Endocrinol (Lausanne) ; 14: 1174600, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033230

RESUMEN

The retina is increasingly recognized as a target of thyroid hormone. We previously reported critical functions for thyroid hormone receptor TRß2, encoded by Thrb, in cones, the photoreceptors that mediate color vision. TRß1, another Thrb receptor isoform, is widely expressed in other tissues but little studied in the retina. Here, we investigate these N-terminal isoforms by RNA-sequencing analysis and reveal a striking biphasic profile for TRß1 in mouse and human retina. In contrast to the early TRß2 peak, TRß1 peaks later during retinal maturation or later differentiation of human retinal organoids. This switch in receptor expression profiles was confirmed using lacZ reporter mice. TRß1 localized in cones, amacrine cells and ganglion cells in contrast to the restricted expression of TRß2 in cones. Intriguingly, TRß1 was also detected in the retinal pigmented epithelium and in anterior structures in the ciliary margin zone, ciliary body and iris, suggesting novel functions in non-retinal eye tissues. Although TRß1 was detected in cones, TRß1-knockout mice displayed only minor changes in opsin photopigment expression and normal electroretinogram responses. Our results suggest that strikingly different temporal and cell-specific controls over TRß1 and TRß2 expression may underlie thyroid hormone actions in a range of ocular cell types. The TRß1 expression pattern suggests novel functions in retinal and non-neural ocular tissues.


Asunto(s)
Receptores de Hormona Tiroidea , Retina , Ratones , Humanos , Animales , Retina/metabolismo , Hormonas Tiroideas/metabolismo , Receptores beta de Hormona Tiroidea/genética , Ratones Noqueados , Mamíferos/metabolismo
20.
Medicine (Baltimore) ; 102(17): e33587, 2023 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-37115071

RESUMEN

INTRODUCTION: Thyroid hormone resistance (RTH) (mim # 188570) is a rare autosomal dominant genetic disorder characterized by reduced thyroid hormone response in target tissues. The clinical manifestations of RTH vary from no symptoms to symptoms of thyroid hormone deficiency to symptoms of thyroid hormone excess. PATIENT CONCERN AND CLINICAL FINDINGS: A 24-month-old girl presented with growth retardation, tachycardia, and persistently elevated thyroid hormones despite antithyroid treatment. DIAGNOSIS/INTERVENTION/OUTCOMES: The patient was diagnosed with RTH, after whole exon gene sequencing, found a de novo missense mutation (c.1375T > G,p.Phe459Val) in a novel locus of the thyroid hormone receptor beta gene. She had only mild growth retardation, so the decision was made to monitor her development without intervention. At her last follow-up at 5 years and 8 months of age, she continued to show growth retardation (-2 standard deviation below age-appropriate levels), in addition to delayed language development. Her comprehension ability and heart rate have remained normal. CONCLUSIONS: We report a mild case of RTH caused by a novel thyroid hormone receptor beta gene mutation. RTH should be considered in the differential diagnosis of abnormal serum thyroxine levels during neonatal screening.


Asunto(s)
Genes erbA , Síndrome de Resistencia a Hormonas Tiroideas , Preescolar , Femenino , Humanos , Pueblos del Este de Asia , Trastornos del Crecimiento/genética , Mutación , Receptores beta de Hormona Tiroidea/genética , Síndrome de Resistencia a Hormonas Tiroideas/diagnóstico , Síndrome de Resistencia a Hormonas Tiroideas/genética , Hormonas Tiroideas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...