Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
Neuroendocrinology ; 113(4): 470-478, 2023.
Article in English | MEDLINE | ID: mdl-36442463

ABSTRACT

INTRODUCTION: Thyrotropin receptor-stimulating antibody (TSAb) is a pathogenic antibody in the serum of patients with Graves' disease. The binding of TSAb to thyroid-stimulating hormone receptor (TSHR) in non-thyroid tissue may be associated with the occurrence and development of Graves' disease-related complications. However, only few studies have been conducted on the effects of TSAb on the brain, and the pathogenesis of acute hyperthyroidism myopathy (ATM) is unclear. Therefore, this study aimed to explore the effect of TSAb on the polarization of BV-2 cells in the brain and its possible mechanism and provide a basic experimental basis for ATM. METHODS: BV-2 cells were treated with different concentrations of TSAb. The relative survival rate of BV-2 cells was determined using the CCK-8 assay; the migration ability of BV-2 cells was detected using the Transwell migration assay; and the expression levels of M1/M2 polarization markers (CD86, inducible nitric oxide synthase [iNOS], CD206, and arginase 1 [Arg-1]), TSHR, tumor necrosis factor-alpha (TNF-α), and nuclear factor-kappa B (NF-κB) protein in BV-2 cells were measured using WB. RESULTS: Compared with the negative control group, the proliferative activity of BV-2 cells was significantly increased in the 20, 50, and 100 ng/mL TSAb groups, and the migration ability of BV-2 cells was significantly enhanced in the 50 and 100 ng/mL TSAb groups. The expression levels of M1 polarization markers (CD86 and iNOS), TSHR, TNF-α, and NF-κB protein in BV-2 cells treated with 50 and 100 ng/mL TSAb for 24 h were significantly upregulated, whereas those of M2 polarization markers (CD206 and Arg-1) significantly decreased. CONCLUSIONS: TSAb can induce abnormal activation of microglia, polarize to the M1 phenotype, and promote the inflammatory cascade reaction, in which TSHR plays a key role in NF-κB activation and proinflammatory cytokine release.


Subject(s)
Graves Disease , NF-kappa B , Humans , Long-Acting Thyroid Stimulator/pharmacology , Microglia , Tumor Necrosis Factor-alpha , Immunoglobulins, Thyroid-Stimulating/pharmacology , Receptors, Thyrotropin/physiology , Graves Disease/etiology , Inflammation , Signal Transduction
2.
Front Endocrinol (Lausanne) ; 12: 706101, 2021.
Article in English | MEDLINE | ID: mdl-34276566

ABSTRACT

The synergistic activation of transcription factors can lead to thyroid progenitor cell speciation. We have previously shown in vitro that mouse or human stem cells, expressing the transcription factors NKx2-1 and Pax8, can differentiate into thyroid neo-follicular structures (TFS). We now show that syngeneic mouse TFS when implanted into hypothyroid TSH receptor knockout (TSHR-KO) mice can ameliorate the hypothyroid state for an extended period. ES cells derived from heterozygous TSHR-KO blastocysts were stably transfected with Nkx2-1-GFP and Pax8-mcherry constructs and purified into 91.8% double positive cells by flow cytometry. After 5 days of activin A treatment these double positive cells were then induced to differentiate into neo-follicles in Matrigel for 21 days in the presence of 500µU/mL of TSH. Differentiated TFS expressing thyroglobulin mRNA were implanted under the kidney capsule of 4-6 weeks old TSHR-KO mice (n=5) as well as hind limb muscle (n=2) and anterior chamber of one eye (n=2). Five of the mice tested after 4 weeks were all rendered euthyroid and all mice remained euthyroid at 20 weeks post implantation. The serum T4 fully recovered (pre-bleed 0.62 ± 0.03 to 8.40 ± 0.57 µg/dL) and the previously elevated TSH became normal or suppressed (pre-bleed 391 ± 7.6 to 4.34 ± 1.25 ng/dL) at the end of the 20 week observation period. The final histology obtained from the implanted kidney tissues showed only rudimentary thyroid follicular structures but which stained positive for thyroglobulin expression. The presence of only rudimentary structures at the site of implant on these extended animals suggested possible migration of cells from the site of implant or an inability of TFCs to maintain proper follicular morphology in these external sites for extended periods. However, there were no signs of tumor formation and no immune infiltration. These preliminary studies show that TSHR-KO mice are a useful model for orthotropic implantation of functional thyroid cells without the need for thyroidectomy, radioiodine ablation or anti thyroid drug control of thyroid function. This approach is also proof of principle that thyroid cells derived from mouse ES cells are capable of surviving as functional neo-follicles in vivo for an extended period of 20 weeks.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Hypothyroidism/therapy , Receptors, Thyrotropin/physiology , Stem Cell Transplantation/methods , Stem Cells/cytology , Thyroid Gland/cytology , Animals , Female , Hypothyroidism/etiology , Hypothyroidism/metabolism , Hypothyroidism/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Thyroid Function Tests
3.
Commun Biol ; 4(1): 795, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172814

ABSTRACT

The underlying molecular mechanisms that determine long day versus short day breeders remain unknown in any organism. Atlantic herring provides a unique opportunity to examine the molecular mechanisms involved in reproduction timing, because both spring and autumn spawners exist within the same species. Although our previous whole genome comparisons revealed a strong association of TSHR alleles with spawning seasons, the functional consequences of these variants remain unknown. Here we examined the functional significance of six candidate TSHR mutations strongly associated with herring reproductive seasonality. We show that the L471M missense mutation in the spring-allele causes enhanced cAMP signaling. The best candidate non-coding mutation is a 5.2 kb retrotransposon insertion upstream of the TSHR transcription start site, near an open chromatin region, which is likely to affect TSHR expression. The insertion occurred prior to the split between Pacific and Atlantic herring and was lost in the autumn-allele. Our study shows that strongly associated coding and non-coding variants at the TSHR locus may both contribute to the regulation of seasonal reproduction in herring.


Subject(s)
Fishes/physiology , Receptors, Thyrotropin/genetics , Alleles , Animals , Atlantic Ocean , Conserved Sequence , Haplotypes , Mutation , Receptors, Thyrotropin/physiology , Reproduction/physiology , Seasons , Signal Transduction , Thyrotropin, beta Subunit/genetics
4.
Invest Ophthalmol Vis Sci ; 60(13): 4336-4345, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31622470

ABSTRACT

Purpose: To investigate the molecular pathways that drive thyroid stimulating hormone receptor (TSHR)-induced cellular proliferation in orbital fibroblasts (OFs) from thyroid eye disease (TED) patients. Methods: Orbital fibroblasts from TED and non-TED patients were treated with TSH and changes in gene expression and proliferation were measured. To determine the role of TSHR, TSHR-specific siRNA was used to deplete TSHR levels. Proliferation was measured by bromodeoxyuridine (BrdU) incorporation. PI3K/Akt activation was analyzed by Western blot. The PI3K inhibitor LY294002 was used to investigate PI3K/Akt signaling in OF proliferation. Expression of TSHR, inflammatory cytokines, proliferation related genes and miR-146a and miR-155 were measured by qPCR. Results: Orbital fibroblasts from TED patients proliferate significantly more than non-TED OFs in response to TSH. TSH-induced proliferation was dependent upon TSHR expression and required the PI3K/Akt signaling cascade. TSHR activation stimulated miR-146a and miR-155 expression. TED OFs produced significantly more miR-146a and miR-155 than non-TED OFs. MiR-146a and miR-155 targets, ZNRF3 and PTEN, which both limit cell proliferation, were decreased in TSH treated OFs. Conclusions: These data reveal that TSHR signaling in TED OFs stimulates proliferation directly through PI3K/Akt signaling and indirectly through induction of miR-146a and miR-155. MiR-146a and miR-155 enhance TED OF proliferation by reducing expression of target genes that normally block cell proliferation. TSHR-dependent expression of miR-146a and miR-155 may explain part of the fibroproliferative pathology observed in TED.


Subject(s)
Graves Ophthalmopathy/metabolism , MicroRNAs/genetics , Orbit/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Thyrotropin/physiology , Adult , Aged , Blotting, Western , Bromodeoxyuridine/metabolism , Cell Proliferation , Cells, Cultured , Fibroblasts/metabolism , Gene Expression , Humans , Middle Aged , Orbit/cytology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology
5.
Best Pract Res Clin Endocrinol Metab ; 32(3): 241-256, 2018 06.
Article in English | MEDLINE | ID: mdl-29779579

ABSTRACT

The thyroid gland produces thyroid hormones (TH), which are essential regulators for growth, development and metabolism. The thyroid is mainly controlled by the thyroid-stimulating hormone (TSH) that binds to its receptor (TSHR) on thyrocytes and mediates its action via different G protein-mediated signaling pathways. TSH primarily activates the Gs-pathway, and at higher concentrations also the Gq/11-pathway, leading to an increase of intracellular cAMP and Ca2+, respectively. To date, the physiological importance of other G protein-mediated signaling pathways in thyrocytes is unclear. Congenital hypothyroidism (CH) is defined as the lack of TH at birth. In familial cases, high-throughput sequencing methods have facilitated the identification of novel mutations. Nevertheless, the precise etiology of CH yet remains unraveled in a proportion of cases. Genetically modified mouse models can reveal new pathophysiological mechanisms of thyroid diseases. Here, we will present an overview of genetic mouse models for thyroid diseases, which have provided crucial insights into thyroid gland development, function, and growth with a special focus on TSHR and microRNA signaling.


Subject(s)
Mice, Transgenic , Thyroid Gland/embryology , Thyroid Gland/growth & development , Thyroid Gland/physiology , Animals , Congenital Hypothyroidism/genetics , Congenital Hypothyroidism/pathology , Humans , Mice , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/physiology , Signal Transduction/physiology , Thyroid Diseases/genetics , Thyroid Diseases/pathology , Thyroid Hormones/physiology , Thyrotropin/genetics , Thyrotropin/physiology
6.
Best Pract Res Clin Endocrinol Metab ; 32(2): 155-164, 2018 04.
Article in English | MEDLINE | ID: mdl-29678283

ABSTRACT

In the absence of crystal available for the full-length thyrotropin receptor, knowledge of its structure and functioning has benefitted from the identification and characterization of mutations in patients with various thyroid dysfunctions. The characterization of activating mutations has contributed to the elaboration of a model involving the extracellular domain of the receptor as an inverse tethered agonist which, upon binding of the ligand, relieves the transmembrane domain from an inhibiting interaction and activates it. The models derived from comparisons with other receptors, enriched with the information provided by the study of mutations, have proven useful for the design of small-molecule agonists and antagonists that may be used in the future to treat thyroid dysfunctions. In this review, extrathyroidal expression of the thyrotropin receptor is described, the role of which is still poorly defined.


Subject(s)
Receptors, Thyrotropin/physiology , Thyroid Diseases/etiology , Animals , Humans , Mutation/physiology , Receptors, Thyrotropin/genetics , Signal Transduction/drug effects , Thyroid Diseases/genetics , Thyroid Diseases/metabolism , Thyrotropin/metabolism
7.
J Pharmacol Exp Ther ; 364(1): 38-45, 2018 01.
Article in English | MEDLINE | ID: mdl-29089368

ABSTRACT

Recently, we showed that TSH-enhanced differentiation of a human preosteoblast-like cell model involved a ß-arrestin 1 (ß-Arr 1)-mediated pathway. To study this pathway in more detail, we sought to discover a small molecule ligand that was functionally selective toward human TSH receptor (TSHR) activation of ß-Arr 1. High-throughput screening using a cell line stably expressing mutated TSHRs and mutated ß-Arr 1 (DiscoverX1 cells) led to the discovery of agonists that stimulated translocation of ß-Arr 1 to the TSHR, but did not activate Gs-mediated signaling pathways, i.e., cAMP production. D3-ßArr (NCGC00379308) was selected. In DiscoverX1 cells, D3-ßArr stimulated ß-Arr 1 translocation with a 5.1-fold greater efficacy than TSH and therefore potentiated the effect of TSH in stimulating ß-Arr 1 translocation. In human U2OS-TSHR cells expressing wild-type TSHRs, which is a model of human preosteoblast-like cells, TSH upregulated the osteoblast-specific genes osteopontin (OPN) and alkaline phosphatase (ALPL). D3-ßArr alone had only a weak effect to upregulate these bone markers, but D3-ßArr potentiated TSH-induced upregulation of ALPL and OPN mRNA levels 1.6-fold and 5.5-fold, respectively, at the maximum dose of ligands. Furthermore, the positive allosteric modulator effect of D3-ßArr resulted in an increase of TSH-induced secretion of OPN protein. In summary, we have discovered the first small molecule positive allosteric modulator of TSHR. As D3-ßArr potentiates the effect of TSH to enhance differentiation of a human preosteoblast in an in vitro model, it will allow a novel experimental approach for probing the role of TSH-induced ß-Arr 1 signaling in osteoblast differentiation.


Subject(s)
Cell Differentiation/drug effects , Drug Discovery/methods , Osteoblasts/drug effects , Receptors, Thyrotropin/agonists , Thyrotropin/pharmacology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , CHO Cells , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Cricetinae , Cricetulus , High-Throughput Screening Assays/methods , Humans , Osteoblasts/physiology , Receptors, Thyrotropin/physiology , Thyroid Epithelial Cells/drug effects , Thyroid Epithelial Cells/metabolism , Thyrotropin/analogs & derivatives
8.
Endocr Regul ; 51(2): 117-128, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28609287

ABSTRACT

Thyrostimulin is a glycoprotein heterodimer of GPA2 and GPB5, first described in 2002. It is involved in the physiological function of several tissues. Moreover, evidence points towards the ability of thyrostimulin's individual monomers to induce a biological effect, which could denote the circulatory/systemic effects of the molecule when found in higher concentrations. From the evolutionary point of view, thyrostimulin shares a binding epitope with the thyroid-stimulating hormone for the thyroid stimulating hormone receptor, whilst possessing affinity for another unique binding site on the same receptor. Although thyrostimulin can be involved in the hypothalamicpituitary- thyroid axis, its presence in various tissues in an eclectic array of different species renders it multifunctional. From weight loss via increasing metabolic rate to progression of cancer in human ovaries, it is certainly not a signaling molecule to overlook. Furthermore, thyrostimulin has been implicated in bone metabolism, acute illness, and reproductive function. In summary, to our knowledge, this is the first review dealing with the physiological role of thyrostimulin and its potential applications in the clinical practice.


Subject(s)
Glycoproteins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Receptors, Thyrotropin/metabolism , Acute Disease , Animals , Carcinoma, Ovarian Epithelial , Female , Glycoproteins/chemistry , Glycoproteins/physiology , Humans , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiology , Mice , Neoplasms, Glandular and Epithelial/physiopathology , Ovarian Neoplasms/physiopathology , Receptors, Thyrotropin/physiology , Reproduction/physiology , Thyroid Gland/metabolism , Thyroid Gland/physiology , Weight Loss/physiology
9.
Endocr Rev ; 2016(1): 23-42, 2016 02.
Article in English | MEDLINE | ID: mdl-27454362

ABSTRACT

The TSH receptor (TSHR) on the surface of thyrocytes is unique among the glycoprotein hormone receptors in comprising two subunits: an extracellular A-subunit, and a largely transmembrane and cytosolic B-subunit. Unlike its ligand TSH, whose subunits are encoded by two genes, the TSHR is expressed as a single polypeptide that subsequently undergoes intramolecular cleavage into disulfide-linked subunits. Cleavage is associated with removal of a C-peptide region, a mechanism similar in some respects to insulin cleavage into disulfide linked A- and B-subunits with lossofaC-peptideregion. The potential pathophysiological importance of TSHR cleavage into A-and B-subunits is that some A-subunits are shed from the cell surface. Considerable experimental evidence supports the concept that A-subunit shedding in genetically susceptible individuals is a factor contributing to the induction and/or affinity maturation of pathogenic thyroid-stimulating autoantibodies, the direct cause of Graves' disease. The noncleaving gonadotropin receptors are not associated with autoantibodies that induce a "Graves' disease of the gonads." We also review herein current information on the location of the cleavage sites, the enzyme(s) responsible for cleavage, the mechanism by which A-subunits are shed, and the effects of cleavage on receptor signaling. (Endocrine Reviews 37: 114-134, 2016).


Subject(s)
Graves Disease/metabolism , Protein Processing, Post-Translational , Receptors, Thyrotropin/metabolism , Thyroid Epithelial Cells/metabolism , Animals , Autoantibodies/immunology , Graves Disease/immunology , Graves Disease/pathology , Humans , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/physiology
10.
Brain Res ; 1642: 278-286, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27059392

ABSTRACT

AIMS: Previous studies have shown that brain-derived thyroid-stimulating hormone (TSH) and its receptor (TSHr) are present in hypothalamic extracts. No studies investigating both the anatomical location and functional significance of putative TSHr proteins in specific central nervous system (CNS) nuclei involved in feeding controls have yet been conducted. The aim was thus to determine whether TSHr are present in nuclei associated with feeding behavior, and if such receptors may be functional. METHODS: Brain tissue from adult rats was analyzed for gene expression and receptor protein expression was investigated with immunohistochemistry and western blotting. To investigate whether putative TSHr may be functional, we evaluated food intake of rats given intraparenchymal nanoinjections of TSH into the nucleus of the solitary tract (NTS). RESULTS: RT-qPCR confirmed previous reports that TSHr mRNA is expressed in CNS tissues of the adult rat. Immunohistochemistry showed TSHr-immunoreactivity in the arcuate, the ventromedial, the dorsomedial, and the paraventricular hypothalamic nuclei. We also found TSHr-ir in the dorsal hindbrain to be localized to the area postrema, NTS, dorsal motor nucleus of the vagus, and the hypoglossal motor nucleus. Further protein analysis with western blotting showed 120kDa TSHr-ir proteins present in the hypothalamus and brainstem. Injections of TSH into the NTS reduced food intake similar to the positive control, urocortin. CONCLUSIONS: These data suggest that functional TSHr are present in the caudal brainstem and hypothalamic nuclei of relevance for feeding control as a possibly uncleaved holoreceptor, and highlights a hindbrain component to central TSH inhibition of food intake.


Subject(s)
Brain Stem/physiology , Eating , Hypothalamus/metabolism , Hypothalamus/physiology , Receptors, Thyrotropin/metabolism , Receptors, Thyrotropin/physiology , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Eating/drug effects , Hypothalamus/drug effects , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Thyrotropin/agonists , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Solitary Nucleus/physiology , Thyrotropin/administration & dosage
11.
Tumour Biol ; 37(3): 3105-13, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26427660

ABSTRACT

The purposes of this study were to investigate the potential roles of long noncoding RNA (lncRNA) PVT1 in thyroid cancer cell proliferation and to explore their possible mechanisms. A total of 84 patients who were diagnosed as having thyroid cancer (papillary thyroid carcinoma (PTC), follicular thyroid carcinoma (FTC), and anaplastic thyroid carcinoma (ATC)) in Renji Hospital were enrolled in this study. Expressions of lncRNA PVT1 in thyroid cancer tissues and cell lines (IHH-4, FTC-133, and 8505C) were analyzed using RT-polymerase chain reaction (PCR) and western blotting analysis. The effects of lncRNA PVT1 expression on thyroid cancer cell proliferation and cell cycle were analyzed using flow cytometry. Furthermore, the effects of lncRNA expression on thyroid-stimulating hormone receptor (TSHR) expression and polycomb enhancer of zeste homolog 2 (EZH2) were also analyzed using RNA immunoprecipitation (RIP) assay and chromatin immunoprecipitation (ChIP) assay, respectively. Compared to the controls, lncRNA PVT1 was significantly up-regulated in thyroid tissues, as well as in three kinds of tumor cell lines (P < 0.05). Silenced PVT1 significantly inhibited thyroid cell line IHH-4, FTC-133, and 8505C cell proliferation and arrested cell cycle at G0/G1 stage and significantly decreased cyclin D1 and TSHR expressions (P < 0.05). Moreover, lncRNA PVT1 could be enriched by EZH2, and silencing PVT1 resulted in the decreased recruitment of EZH2. This study suggested that lncRNA PVT1 may contribute to tumorigenesis of thyroid cancer through recruiting EZH2 and regulating TSHR expression.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/physiology , RNA, Long Noncoding/physiology , Receptors, Thyrotropin/physiology , Thyroid Neoplasms/pathology , Adult , Aged , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Middle Aged , RNA, Long Noncoding/analysis , Thyroid Neoplasms/etiology
12.
Endocr J ; 62(5): 393-8, 2015.
Article in English | MEDLINE | ID: mdl-25797365

ABSTRACT

Genetic defects of hormone receptors are the most common form of end-organ hormone resistance. One example of such defects is TSH resistance, which is caused by biallelic inactivating mutations in the TSH receptor gene (TSHR). TSH, a master regulator of thyroid functions, affects virtually all cellular processes involving thyroid hormone production, including thyroidal iodine uptake, thyroglobulin iodination, reuptake of iodinated thyroglobulin and thyroid cell growth. Resistance to TSH results in defective thyroid hormone production from the neonatal period, namely congenital hypothyroidism. Classically, clinical phenotypes of TSH resistance due to inactivating TSHR mutations were thought to vary depending on the residual mutant receptor activity. Nonfunctional mutations in the two alleles produce severe thyroid hypoplasia with overt hypothyroidism (uncompensated TSH resistance), while hypomorphic mutations in at least one allele produce normal-sized thyroid gland with preserved hormone-producing capacity (compensated TSH resistance). More recently, a new subgroup of TSH resistance (nonclassic TSH resistance) that is characterized by paradoxically high thyroidal iodine uptake has been reported. In this article, the pathophysiology and clinical features of TSH resistance due to inactivating TSHR mutations are reviewed, with particular attention to the nonclassic form.


Subject(s)
Thyroid Hormone Resistance Syndrome , Thyrotropin , Alleles , Congenital Hypothyroidism/genetics , Genotype , Humans , Iodine/metabolism , Mutation , Phenotype , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/physiology , Thyroid Dysgenesis/genetics , Thyroid Gland/pathology , Thyroid Gland/physiopathology , Thyroid Hormone Resistance Syndrome/complications , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormones/biosynthesis
13.
J Clin Endocrinol Metab ; 100(3): 1071-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25485727

ABSTRACT

CONTEXT: There is no pathogenetically linked medical therapy for Graves' ophthalmopathy (GO). Lack of animal models and conflicting in vitro studies have hindered the development of such therapy. Recent reports propose that Graves' Igs bind to and activate thyrotropin receptors (TSHRs) and IGF-1 receptors (IGF-1Rs) on cells in orbital fat, stimulating hyaluronan (HA) secretion, a component of GO. OBJECTIVE: The objective of the study was to investigate potential cross talk between TSHRs and IGF-1Rs in the pathogenesis of GO using a sensitive HA assay. DESIGN/SETTING/PARTICIPANTS: Orbital fibroblasts from GO patients were collected in an academic clinical practice and cultured in a research laboratory. Cells were treated with TSH, IGF-1, and a monoclonal Graves' Ig M22. MAIN OUTCOME MEASURES: HA was measured by a modified ELISA. RESULTS: Simultaneous activation by TSH and IGF-1 synergistically increased HA secretion from 320 ± 52 for TSH and 430 ± 65 µg/mL for IGF-1 alone, to 1300 ± 95 µg/mL. IGF-1 shifted the TSH EC50 19-fold to higher potency. The dose response to M22 was biphasic. An IGF-1R antagonist inhibited the higher potency phase but had no effect on the lower potency phase. M22 did not cause IGF-1R autophosphorylation. A TSHR antagonist abolished both phases of M22-stimulated HA secretion. CONCLUSIONS: M22 stimulation of HA secretion by GO fibroblasts/preadipocytes involves cross talk between TSHR and IGF-1R. This cross talk relies on TSHR activation rather than direct activation of IGF-1R and leads to synergistic stimulation of HA secretion. These data propose a model for GO pathogenesis that explains previous contradictory results and argues for TSHR as the primary therapeutic target for GO.


Subject(s)
Graves Disease/immunology , Hyaluronic Acid/metabolism , Immunoglobulins/pharmacology , Receptor Cross-Talk/physiology , Receptor, IGF Type 1/physiology , Receptors, Thyrotropin/physiology , Cells, Cultured , Drug Synergism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Graves Disease/metabolism , Graves Disease/pathology , Humans , Insulin-Like Growth Factor I/pharmacology , Orbit/pathology , Thyrotropin/pharmacology , Up-Regulation/drug effects
14.
Trans Am Ophthalmol Soc ; 112: 26-37, 2014.
Article in English | MEDLINE | ID: mdl-25411513

ABSTRACT

PURPOSE: To better understand the pathogenesis of thyroid-associated orbitopathy (TAO) through elucidating the role of thyrotropin receptor (TSHR) and CD40 in the expression of interleukin-8 (IL-8) in peripheral blood fibrocytes. Fibrocytes infiltrate the orbit of patients with TAO, where they differentiate into fibroblasts. Fibrocyte precursors occur with increased frequency in the peripheral blood expressing TSHR and CD40 in TAO patients. We hypothesize that in vitro derived fibrocytes and peripheral blood fibrocyte precursors express proinflammatory chemoattractant molecules including IL-8 initiated by TSHR and CD40 signaling. Since nearly all TAO patients express activating antibodies to TSHR, this is particularly relevant for activation of peripheral blood fibrocytes. METHODS: TSHR and CD40 expression on peripheral blood fibrocytes was determined by flow cytometry. IL-8 RNA was quantitated by real-time polymerase chain reaction. IL-8 protein production was measured by Luminex and flow cytometry. Thyroid-stimulating hormone and CD40 ligand-stimulated phosphorylation of Akt in peripheral blood fibrocytes was studied by flow cytometry. RESULTS: Both TSHR- and CD40-mediated signaling lead to IL-8 expression in mature fibrocytes. Fibrocyte precursors assayed directly from circulating peripheral blood demonstrate intracellular IL-8 expression with addition of thyroid-stimulating hormone or CD40 ligand. TSHR- and CD40-induced IL-8 production is mediated by Akt phosphorylation. CONCLUSIONS: Peripheral blood TSHR(+) and CD40(+) fibrocytes express IL-8 and may promote the recruitment of inflammatory cells, mitogenesis, and tissue remodeling in TAO. TSHR- and CD40-mediated IL-8 signaling is mediated by Akt. Delineating the molecular mechanisms of fibrocyte immune function may provide potential therapeutic targets for TAO.


Subject(s)
CD40 Antigens/physiology , Fibroblasts/metabolism , Graves Ophthalmopathy/metabolism , Interleukin-8/metabolism , Receptors, Thyrotropin/physiology , Adult , Analysis of Variance , Cells, Cultured , Fibroblasts/drug effects , Flow Cytometry , Humans , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology
15.
FASEB J ; 28(8): 3446-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24723693

ABSTRACT

Thyrotropin (TSH) activation of the TSH receptor (TSHR), a 7-transmembrane-spanning receptor (7TMR), may have osteoprotective properties by direct effects on bone. TSHR activation by TSH phosphorylates protein kinases AKT1, p38α, and ERK1/2 in some cells. We found TSH-induced phosphorylation of these kinases in 2 cell lines engineered to express TSHRs, human embryonic kidney HEK-TSHR cells and human osteoblastic U2OS-TSHR cells. In U2OS-TSHR cells, TSH up-regulated pAKT1 (7.1±0.5-fold), p38α (2.9±0.4-fold), and pERK1/2 (3.1±0.2-fold), whereas small molecule TSHR agonist C2 had no or little effect on pAKT1 (1.8±0.08-fold), p38α (1.2±0.09-fold), and pERK1/2 (1.6±0.19-fold). Furthermore, TSH increased expression of osteoblast marker genes ALPL (8.2±4.6-fold), RANKL (21±5.9-fold), and osteopontin (OPN; 17±5.3-fold), whereas C2 had little effect (ALPL, 1.7±0.5-fold; RANKL, 1.3±0.6-fold; and OPN, 2.2±0.7-fold). ß-Arrestin-1 and -2 can mediate activatory signals by 7TMRs. TSH stimulated translocation of ß-arrestin-1 and -2 to TSHR, whereas C2 failed to translocate either ß-arrestin. Down-regulation of ß-arrestin-1 by siRNA inhibited TSH-stimulated phosphorylation of ERK1/2, p38α, and AKT1, whereas down-regulation of ß-arrestin-2 increased phosphorylation of AKT1 in both cell types and of ERK1/2 in HEK-TSHR cells. Knockdown of ß-arrestin-1 inhibited TSH-stimulated up-regulation of mRNAs for OPN by 87 ± 1.7% and RANKL by 73 ± 2.4%, and OPN secretion by 74 ± 10%. We conclude that TSH enhances osteoblast differentiation in U2OS cells that is, in part, caused by activatory signals mediated by ß-arrestin-1.


Subject(s)
Arrestins/physiology , Osteoblasts/drug effects , Thyrotropin/physiology , Bone Neoplasms/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Neoplasm Proteins/physiology , Osteoblasts/cytology , Osteopontin/metabolism , Osteosarcoma/pathology , Phosphorylation , Protein Kinases/metabolism , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/pharmacology , Receptors, Thyrotropin/physiology , Recombinant Fusion Proteins , Signal Transduction/physiology , Thyrotropin/pharmacology , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
16.
Endocr Rev ; 34(5): 691-724, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23645907

ABSTRACT

The TSH receptor (TSHR) is a member of the glycoprotein hormone receptors, a subfamily of family A G protein-coupled receptors. The TSHR is of great importance for the growth and function of the thyroid gland. The TSHR and its endogenous ligand TSH are pivotal proteins with respect to a variety of physiological functions and malfunctions. The molecular events of TSHR regulation can be summarized as a process of signal transduction, including signal reception, conversion, and amplification. The steps during signal transduction from the extra- to the intracellular sites of the cell are not yet comprehensively understood. However, essential new insights have been achieved in recent years on the interrelated mechanisms at the extracellular region, the transmembrane domain, and intracellular components. This review contains a critical summary of available knowledge of the molecular mechanisms of signal transduction at the TSHR, for example, the key amino acids involved in hormone binding or in the structural conformational changes that lead to G protein activation or signaling regulation. Aspects of TSHR oligomerization, signaling promiscuity, signaling selectivity, phenotypes of genetic variations, and potential extrathyroidal receptor activity are also considered, because these are relevant to an understanding of the overall function of the TSHR, including physiological, pathophysiological, and pharmacological perspectives. Directions for future research are discussed.


Subject(s)
Receptors, Thyrotropin/physiology , Signal Transduction/physiology , Amino Acids/genetics , Binding Sites , Cell Membrane/chemistry , GTP-Binding Proteins/metabolism , Gene Expression , Genetic Variation , Humans , Molecular Structure , Mutation , Phenotype , Protein Conformation , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/genetics , Thyroid Diseases/genetics
17.
Ann Dermatol Venereol ; 140(5): 382-5, 2013 May.
Article in French | MEDLINE | ID: mdl-23663712

ABSTRACT

BACKGROUND: Localized myxoedema is a rare dermopathy in patients with Graves' disease. The pretibial area is the most commonly affected region but herein we present a case of myxoedema of the big toe. PATIENTS AND METHODS: A 44-year-old male with Graves' disease ongoing for seven years presented bilateral ophthalmopathy and myxoedema of the big toes. The myxoedema was treated successfully with intralesional steroids. DISCUSSION: The physiopathology of myxoedema involves fibroblast activation and glycosaminoglycan production. This activation could result from stimulation of TSH receptors at their surface by TSH receptor antibodies (TRAK) or from an inflammatory process. The pretibial topography may be related to the high frequency in this area of microtrauma, with modulation of the cytokine microenvironment. CONCLUSION: The atypical localization seems to correlate with a Koebner phenomenon. Treatment of Graves' disease is generally insufficient to resolve the cutaneous problems. Topical corticosteroid therapy generally results in rapid improvement of recent lesions.


Subject(s)
Foot Dermatoses/pathology , Graves Disease/pathology , Myxedema/pathology , Toes/pathology , Adult , Biopsy , Carbimazole/therapeutic use , Decompression, Surgical , Fibroblasts/metabolism , Fibroblasts/pathology , Foot Dermatoses/drug therapy , Foot Dermatoses/etiology , Foot Dermatoses/physiopathology , Glycosaminoglycans/analysis , Glycosaminoglycans/metabolism , Graves Disease/complications , Graves Disease/drug therapy , Graves Disease/physiopathology , Graves Disease/surgery , Graves Ophthalmopathy/etiology , Graves Ophthalmopathy/surgery , Hormone Replacement Therapy , Humans , Immunoglobulins, Thyroid-Stimulating/immunology , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Injections, Intralesional , Male , Methimazole/therapeutic use , Myxedema/drug therapy , Myxedema/etiology , Myxedema/physiopathology , Receptors, Thyrotropin/immunology , Receptors, Thyrotropin/physiology , Thyroidectomy , Thyroxine/therapeutic use , Triamcinolone/administration & dosage , Triamcinolone/therapeutic use
18.
J Endocrinol Invest ; 36(9): 716-21, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23563316

ABSTRACT

BACKGROUND: TSHR is a G-protein-coupled seven transmembrane domain receptor that activates the two major signal transduction pathways: the Gαs/adenylate cyclase and the Gαq/11/phospholipase C pathways. Inactivating mutations in the TSHR gene have been demonstrated to be responsible for subclinical hypothyroidism, a disorder characterized by elevated serum TSH concentrations despite normal thyroid hormones levels. AIM: We identified in a child a nonsense mutation (W520X) in the third transmembrane domain of the TSHR that causes the lack of the C-terminus portion of the receptor. The functional significance of this variation was assessed in vitro. MATERIAL/SUBJECT AND METHODS: The W520X mutation was introduced into the pSVL vector containing the wild-type sequence of TSHR gene. Wild-type and mutated vectors were expressed in Chinese Hamster Ovary (CHO) cells, and cAMP, inositol phosphate (IP), immunofluorescence and FACS analyses were performed. RESULTS: Transfection with pSVL-TSHR vector induced basal cAMP and IP production in the absence of TSH stimulation, indicating a constitutive activity for the TSHR. An impairment of receptor function was demonstrated by the observation that cells expressing the mutant TSHR exhibited a lower second messenger production with respect to the wild-type, despite a normal expression of the receptor at the cell surface. CONCLUSIONS: The mechanism through which the W520X mutation exerts its effect is more likely haploinsufficiency rather than a dominant-negative effect. This could explain the phenotype of our patient, who has a hormonal pattern in the range of a mild subclinical hypothyroidism, without an overt disease phenotype.


Subject(s)
Hypothyroidism/genetics , Receptors, Thyrotropin/genetics , Animals , CHO Cells , Child , Cricetinae , Cricetulus , Female , Haploinsufficiency , Humans , Male , Receptors, Thyrotropin/physiology
19.
J Clin Endocrinol Metab ; 98(1): E144-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23118423

ABSTRACT

CONTEXT: Genetic factors play a major role in the etiology of autoimmune thyroid disease (AITD) including Graves' disease (GD) and Hashimoto's thyroiditis (HT). We have previously identified three loci on chromosomes 10q, 12q, and 14q that showed strong linkage with AITD, HT, and GD, respectively. OBJECTIVES: The objective of the study was to identify the AITD susceptibility genes at the 10q, 12q, and 14q loci. DESIGN AND PARTICIPANTS: Three hundred forty North American Caucasian AITD patients and 183 healthy controls were studied. The 10q, 12q, and 14q loci were fine mapped by genotyping densely spaced single-nucleotide polymorphisms (SNPs) using the Illumina GoldenGate genotyping platform. Case control association analyses were performed using the UNPHASED computer package. Associated SNPs were reanalyzed in a replication set consisting of 238 AITD patients and 276 controls. RESULTS: Fine mapping of the AITD locus, 10q, showed replicated association of the AITD phenotype (both GD and HT) with SNP rs6479778. This SNP was located within the ARID5B gene recently reported to be associated with rheumatoid arthritis and GD in Japanese. Fine mapping of the GD locus, 14q, revealed replicated association of the GD phenotype with two markers, rs12147587 and rs2284720, located within the NRXN3 and TSHR genes, respectively. CONCLUSIONS: Fine mapping of three linked loci identified novel susceptibility genes for AITD. The discoveries of new AITD susceptibility genes will engender a new understanding of AITD etiology.


Subject(s)
Autoimmune Diseases/genetics , Chromosome Mapping/methods , Genetic Loci/genetics , Genetic Predisposition to Disease , Thyroid Diseases/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Child , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Female , Genetic Association Studies , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Polymorphism, Single Nucleotide/physiology , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/physiology , Thyroiditis, Autoimmune/genetics , Transcription Factors/genetics , Transcription Factors/physiology , Young Adult
20.
J Clin Endocrinol Metab ; 97(12): 4287-92, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23019348

ABSTRACT

The physiological role of the TSH receptor (TSHR) as a major regulator of thyroid function is well understood, but TSHRs are also expressed in multiple normal extrathyroidal tissues, and the physiological roles of TSHRs in these tissues are unclear. Moreover, TSHRs play a major role in several pathological conditions including hyperthyroidism, hypothyroidism, and thyroid tumors. Small molecule, "drug-like" TSHR agonists, neutral antagonists, and inverse agonists may be useful as probes of TSHR function in extrathyroidal tissues and as leads to develop drugs for several diseases of the thyroid. In this Update, we review the most recent findings regarding the development and use of these small molecule TSHR ligands.


Subject(s)
Drug Discovery/trends , Receptors, Thyrotropin/agonists , Receptors, Thyrotropin/antagonists & inhibitors , Animals , Antithyroid Agents/chemistry , Antithyroid Agents/pharmacology , Antithyroid Agents/therapeutic use , Endocrinology/trends , Hormone Antagonists/chemistry , Hormone Antagonists/pharmacology , Humans , Ligands , Models, Biological , Models, Molecular , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/physiology , Thyroid Diseases/drug therapy , Thyroid Diseases/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...