Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.428
Filter
1.
Toxicology ; 501: 153713, 2024 01.
Article in English | MEDLINE | ID: mdl-38135142

ABSTRACT

Bis (2-ethylhexyl) tetrabromophthalate (TBPH) is a new type of brominated flame retardant. Some studies suggest that TBPH exposure may be associated with thyroid damage. However, there is a paucity of research on the authentic exposure-related effects and molecular mechanisms in animals or cells. In this study, we used male Sprague-Dawley (SD) rats and the Nthy ori3-1 cell line (the human thyroid follicular epithelial cell) to explore the potential effects of TBPH (5, 50, 500 mg/kg and 1, 10, 100 nM) on the thyroid. The genes and their proteins of cytokines and thyroid-specific proteins, thyroglobulin (TG), thyroid peroxidase (TPO), and sodium iodide cotransporter (NIS) were examined to investigate the possible mechanisms. At the end of the experiment, it was found that 50 and 500 mg/kg TBPH could increase the levels of total thyroxine (TT4) and free thyroxine (FT4) significantly. The messenger RNAs (mRNAs) of Tg, Tpo, Interleukin-6 (Il6), and Interleukin-10 (Il10) in the thyroid tissues from the rats treated with 500 mg/kg were enhanced clearly. Meanwhile, the mRNAs of TG, TPO, IL6, and IL10 were elevated in Nthy ori3-1 cells treated with 100 nM TBPH as well. The mRNAs of TG and TPO were elevated after the knockdown of IL6. To our surprise, after the knockdown of IL10 or the treatment of anti-IL-10-receptor (anti-IL-10-R) antibody, the mRNAs of TG and TPO were significantly reduced, and the effects of TBPH were diminished. In conclusion, our results suggested that the IL-10-IL-10R-TG/TPO-T4 axis is one important target of TBPH in the thyroid.


Subject(s)
Thyroglobulin , Thyroid Gland , Male , Humans , Rats , Animals , Thyroglobulin/genetics , Thyroglobulin/metabolism , Thyroglobulin/pharmacology , Interleukin-10/genetics , Thyroxine , Interleukin-6/metabolism , Rats, Sprague-Dawley , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , RNA, Messenger/metabolism
2.
Environ Sci Technol ; 58(1): 99-109, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38117130

ABSTRACT

Identifying endocrine disrupting chemicals in order to limit their usage is a priority and required according to the European Regulation. There are no Organization for Economic Co-operation and Development (OECD) test guidelines based on fish available for the detection of Thyroid axis Active Chemicals (TACs). This study aimed to fill this gap by developing an assay at eleuthero-embryonic life stages in a novel medaka (Oryzias latipes) transgenic line. This transgenic line expresses green fluorescent protein (GFP) in thyrocytes, under the control of the medaka thyroglobulin gene promoter. The fluorescence expressed in the thyrocytes is inversely proportional to the thyroid axis activity. When exposed for 72 h to activators (triiodothyronine (T3) and thyroxine (T4)) or inhibitors (6-N-propylthiouracil (PTU), Tetrabromobisphenol A (TBBPA)) of the thyroid axis, the thyrocytes can change their size and express lower or higher levels of fluorescence, respectively. This reflects the regulation of thyroglobulin by the negative feedback loop of the Hypothalamic-Pituitary-Thyroid axis. T3, T4, PTU, and TBBPA induced fluorescence changes with the lowest observable effect concentrations (LOECs) of 5 µg/L, 1 µg/L, 8 mg/L, and 5 mg/L, respectively. This promising tool could be used as a rapid screening assay and also to help decipher the mechanisms by which TACs can disrupt the thyroid axis in medaka.


Subject(s)
Oryzias , Thyroid Gland , Animals , Thyroid Gland/physiology , Oryzias/physiology , Thyroglobulin/metabolism , Thyroglobulin/pharmacology , Triiodothyronine/metabolism , Triiodothyronine/pharmacology
3.
Protein Sci ; 32(11): e4784, 2023 11.
Article in English | MEDLINE | ID: mdl-37717261

ABSTRACT

Thyroglobulin must pass endoplasmic reticulum (ER) quality control to become secreted for thyroid hormone synthesis. Defective thyroglobulin, blocked in trafficking, can cause hypothyroidism. Thyroglobulin is a large protein (~2750 residues) spanning regions I-II-III plus a C-terminal cholinesterase-like domain. The cholinesterase-like domain functions as an intramolecular chaperone for regions I-II-III, but the folding pathway leading to successful thyroglobulin trafficking remains largely unknown. Here, informed by the recent three-dimensional structure of thyroglobulin as determined by cryo-electron microscopy, we have bioengineered three novel classes of mutants yielding three entirely distinct quality control phenotypes. Specifically, upon expressing recombinant thyroglobulin, we find that first, mutations eliminating a disulfide bond enclosing a 200-amino acid loop in region I have surprisingly little impact on the ability of thyroglobulin to fold to a secretion-competent state. Next, we have identified a mutation on the surface of the cholinesterase-like domain that has no discernible effect on regional folding yet affects contact between distinct regions and thereby triggers impairment in the trafficking of full-length thyroglobulin. Finally, we have probed a conserved disulfide in the cholinesterase-like domain that interferes dramatically with local folding, and this defect then impacts on global folding, blocking the entire thyroglobulin in the ER. These data highlight variants with distinct effects on ER quality control, inhibiting domain-specific folding; folding via regional contact; neither; or both.


Subject(s)
Protein Folding , Thyroglobulin , Thyroglobulin/genetics , Thyroglobulin/chemistry , Thyroglobulin/metabolism , Cryoelectron Microscopy , Thyroid Hormones , Protein Transport , Cholinesterases/chemistry , Cholinesterases/metabolism , Disulfides
4.
Int J Mol Sci ; 24(11)2023 May 26.
Article in English | MEDLINE | ID: mdl-37298246

ABSTRACT

In the thyroid gland, cysteine cathepsins are secreted upon thyrotropin stimulation for thyroglobulin processing, and they are present at the primary cilia of thyroid epithelial cells. Treatment with protease inhibitors resulted in the loss of cilia from rodent thyrocytes and caused redistribution of the thyroid co-regulating G protein-coupled receptor Taar1 to the endoplasmic reticulum. These findings suggest that ciliary cysteine cathepsins are important to maintain sensory and signaling properties for the proper regulation and homeostasis of thyroid follicles. Therefore, it is important to better understand how cilia structure and frequencies are maintained in human thyroid epithelial cells. Hence, we aimed to investigate the potential role of cysteine cathepsins for the maintenance of primary cilia in the normal human Nthy-ori 3-1 thyroid cell line. This was approached by determining cilia lengths and frequencies in cysteine peptidase inhibition conditions in Nthy-ori 3-1 cell cultures. Cilia lengths were shortened upon 5 h of cysteine peptidase inhibition with cell-impermeable E64. Likewise, cilia lengths and frequencies were decreased upon additional overnight treatment with the cysteine peptidase-targeting, activity-based probe DCG-04. The results suggest that cysteine cathepsin activity is required for the maintenance of the cellular protrusions not only in rodents, but also in human thyrocytes. Hence, thyrotropin stimulation was used to simulate physiological conditions that eventually lead to cathepsin-mediated thyroglobulin proteolysis, which is initiated in the thyroid follicle lumen. Immunoblotting revealed that thyrotropin stimulation conditions result in the secretion of little procathepsin L and some pro- and mature cathepsin S but no cathepsin B from the human Nthy-ori 3-1 cells. Unexpectedly, however, 24 h incubation periods with thyrotropin shortened the cilia although higher amounts of cysteine cathepsins were present in the conditioned media. These data point to the necessity of further studies to delineate which of the cysteine cathepsins plays the most prominent role in cilia shortening and/or elongation. Collectively, the results of our study provide corroboration for the hypothesis of thyroid autoregulation by local mechanisms that our group previously proposed.


Subject(s)
Thyroglobulin , Thyrotropin , Humans , Thyroglobulin/metabolism , Thyrotropin/pharmacology , Thyrotropin/metabolism , Cilia/metabolism , Cysteine/metabolism , Thyroid Gland/metabolism
5.
Mol Cell Endocrinol ; 572: 111948, 2023 07 15.
Article in English | MEDLINE | ID: mdl-37164149

ABSTRACT

Congenital hypothyroidism (CH) due to thyroglobulin (TG) variants causes very low serum TG levels with normal or enlarged thyroid glands, depending on the severity of the defect, and with autosomal recessive inheritance. The purpose of this study was to functionally characterize p.Cys1281Tyr variant in the TG gene in order to increase our knowledge of the molecular mechanisms associated with CH. In order to find evidence that support the hypothesis that the p.Cys1281Tyr variant would affect the TG folding were performed amino acid prediction, 3D modeling and transient expression analysis in HEK293T cells. 18 of the 21″in silico" algorithms predict a deleterious effect of the p.Cys1281Tyr variant. The full-length 3D model p.Cys1281Tyr TG showed disulfide bond cleavage between the cysteines at positions 1249 and 1281 and rearrangement of the TG structure, while transient expression analysis indicated that p.Cys1281Tyr causes retention of the protein inside the cell. Consequently, these results show that this pathogenic variant makes it impossible for TG to fulfill its function in the biosynthesis process of thyroid hormones, causing CH. In conclusion, our results confirm the pathophysiological importance of misfolding of TG as a consequence of p.Cys1281Tyr variant located in the hinge module/flap region of TG.


Subject(s)
Congenital Hypothyroidism , Goiter , Humans , Congenital Hypothyroidism/genetics , Thyroglobulin/genetics , Thyroglobulin/metabolism , HEK293 Cells , Goiter/genetics , Thyroid Hormones
6.
Thyroid ; 33(4): 407-419, 2023 04.
Article in English | MEDLINE | ID: mdl-37037032

ABSTRACT

Background: This review presents a timeline showing how technical advances made over the last seven decades have impacted the development of laboratory thyroid tests. Summary: Thyroid tests have evolved from time-consuming manual procedures using isotopically labeled iodine as signals (131I and later 125I) performed in nuclear medicine laboratories, to automated nonisotopic tests performed on multianalyte instruments in routine clinical chemistry laboratories. The development of isotopic radioimmunoassay techniques around 1960, followed by the advent of monoclonal antibody technology in the mid-1970s, led to the development of a nonisotopic immunometric assay methodology that forms the backbone of present-day thyroid testing. This review discusses the development of methods for measuring total thyroxine and triiodothyronine, direct and indirect free thyroid hormone measurements and estimates (free thyroxine and free triiodothyronine), thyrotropin (TSH), thyroid autoantibodies (thyroperoxidase, thyroglobulin [Tg] and TSH receptor autoantibodies), and Tg protein. Despite progressive improvements made in sensitivity and specificity, current thyroid tests remain limited by between-method differences in the numeric values they report, as well as nonspecific interferences with test reagents and interferences from analyte autoantibodies. Conclusions: Thyroid disease affects ∼10% of the U.S. population and is mostly managed on an outpatient basis, generating 60% of endocrine laboratory tests. In future, it is hoped that interferences will be eliminated, and the standardization/harmonization of tests will facilitate the establishment of universal test reference ranges.


Subject(s)
Thyroid Gland , Triiodothyronine , Humans , Thyroid Gland/metabolism , Triiodothyronine/metabolism , Thyroxine/metabolism , Laboratories , Thyroid Function Tests , Thyroglobulin/metabolism , Thyrotropin/metabolism , Autoantibodies/metabolism
7.
Environ Pollut ; 322: 121117, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36690294

ABSTRACT

MC-LR can interfere with thyroid function in fish, but the underlying mechanism is still unclear. Current study focuses to study the intergenerational inheritance of MC-LR-induced thyroid toxicity in zebrafish and in rat thyroid cells. In vivo experiments, adult female zebrafish (F0) were exposed to MC-LR (0, 5, and 25 µg/L) for 90 days and mated with male zebrafish without MC-LR exposure to generate F1 generation. F1 embryos were allowed to develop normally to 7 days post-fertilization (dpf) in clear water. In the F0 generation, MC-LR induced disturbance of the hypothalamic-pituitary-thyroid (HPT) axis, leading to a decrease in the production of thyroid hormones. Maternal MC-LR exposure also induced growth inhibition by altering thyroid hormones (THs) homeostasis and interfering with thyroid metabolism and development in F1 offspring. Mechanistically, MC-LR caused excessive accumulation of ROS and induced ER stress that further lead to activation of UPR in the F0 and F1 offspring of zebrafish. Interestingly, our findings suggested that MC-LR exposure hampered thyroglobulin turnover by triggering IRE1 and PERK pathway in zebrafish and FRTL-5 thyroid cells, thus disturbing the thyroid endocrine system and contributing to the thyroid toxicity from maternal to its F1 offspring of zebrafish. Particularly, inhibition of the IRE1 pathway by siRNA could alleviate thyroid development injury induced by MC-LR in FRTL-5 cells. In addition, MC-LR induced thyroid cell apoptosis by triggering ER stress. Taken together, our results demonstrated that maternal MC-LR exposure causes thyroid endocrine disruption by ER stress contributing to transgenerational effects in zebrafish offspring.


Subject(s)
Endoplasmic Reticulum Stress , Microcystins , Thyroid Gland , Animals , Female , Male , Apoptosis , Microcystins/toxicity , Microcystins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/pharmacology , Thyroglobulin/metabolism , Thyroglobulin/pharmacology , Thyroid Gland/metabolism , Thyroid Gland/pathology , Thyroid Hormones/metabolism , Water Pollutants, Chemical/metabolism , Zebrafish/metabolism
8.
Endocrine ; 80(1): 47-53, 2023 04.
Article in English | MEDLINE | ID: mdl-36547798

ABSTRACT

Thyroglobulin (TG), the predominant glycoprotein of the thyroid gland, functions as matrix protein in thyroid hormonegenesis. TG deficiency results in thyroid dyshormonogenesis. These variants produce a heterogeneous spectrum of congenital goitre, with an autosomal recessive mode of inheritance. The purpose of this study was to identify and functionally characterize new variants in the TG gene in order to increase the understanding of the molecular mechanisms responsible for thyroid dyshormonogenesis. A total of four patients from two non-consanguineous families with marked alteration of TG synthesis were studied. The two families were previously analysed in our laboratory, only one deleterious allele, in each one, was detected after sequencing the TG gene (c.2359 C > T [p.Arg787*], c.5560 G > T [p.Glu1854*]). These findings were confirmed in the present studies by Next-Generation Sequencing. The single nucleotide coding variants of the TG gene were then analyzed to predict the possible variant causing the disease. The p.Pro2232Leu (c.6695 C > T), identified in both families, showing a low frequency population in gnomAD v2.1.1 database and protein homology, amino acid prediction, and 3D modeling analysis predict a potential pathogenic effect of this variant. We also transiently express p.Pro2232Leu in a full-length rat TG cDNA clone and confirmed that this point variant was sufficient to cause intracellular retention of mutant TG in HEK293T cells. Consequently, each family carried a compound heterozygous for p.Arg787*/p.Pro2232Leu or p.Glu1854*/p.Pro2232Leu variants. In conclusion, our results confirm the pathophysiological importance of altered TG folding as a consequence of missense variants located in the ChEL domain of TG.


Subject(s)
Congenital Hypothyroidism , Goiter , Animals , Humans , Rats , Congenital Hypothyroidism/genetics , HEK293 Cells , Thyroglobulin/genetics , Thyroglobulin/metabolism , Protein Transport/genetics
9.
Vet Comp Oncol ; 21(1): 111-122, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36583463

ABSTRACT

Organoid cultures could constitute a valuable in vitro model to explore new treatments for canine (c) medullary thyroid carcinoma (MTC). The study's objectives were to establish and characterize 3D organoid cultures of cMTC using histology and immunohistochemistry (IHC) and to evaluate the effect of antitumor drugs on organoids' viability. Five cMTC tissue samples were used to develop organoid cultures of which one organoid line, named cMTC N°2, could be passaged for an extended period. This cMTC N°2 organoid line was further compared to the primary tumour regarding morphology and IHC expression of thyroid transcription factor-1 (TTF-1), thyroglobulin, calcitonin, synaptophysin, vimentin, Ki-67, cyclooxygenase-2 (COX-2), P-glycoprotein and vascular endothelial growth factor (VEGF). Quality control of the cMTC N°2 organoid line was achieved by a single nucleotide polymorphism (SNP) array of the organoids, primary tumour and healthy blood cells of the same dog. The effect of carboplatin, meloxicam and toceranib phosphate (TOC) on cMTC N°2 organoids' viability was evaluated. The cMTC N°2 organoid line was cultured for 94 days and showed similar histological features with the primary tumour. Immunolabelling for TTF-1, thyroglobulin, calcitonin and VEGF was similar between the primary tumour and cMTC N°2 organoids. Compared to the primary tumour, organoids showed higher immunolabelling for vimentin and Ki-67, and lower immunolabelling for synaptophysin, COX-2 and P-glycoprotein. The SNP genotype was similar for each chromosome between healthy blood cells, primary tumour and cMTC N°2 organoids. Carboplatin, meloxicam and TOC had no effect on cMTC N°2 organoid cell viability within achievable in vivo concentration range. In conclusion, the cMTC N°2 organoid line is a promising first milestone towards an established in vitro organoid model to explore pathophysiology and new treatment modalities in cMTC.


Subject(s)
Dog Diseases , Thyroid Neoplasms , Dogs , Animals , Calcitonin/metabolism , Calcitonin/pharmacology , Thyroglobulin/metabolism , Thyroglobulin/pharmacology , Synaptophysin/metabolism , Synaptophysin/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vimentin/metabolism , Carboplatin/pharmacology , Cyclooxygenase 2/metabolism , Ki-67 Antigen/metabolism , Meloxicam/therapeutic use , Dog Diseases/pathology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/veterinary , Organoids/metabolism , Organoids/pathology , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/pharmacology
10.
Int J Mol Sci ; 23(21)2022 Nov 06.
Article in English | MEDLINE | ID: mdl-36362390

ABSTRACT

The primary functional units of the thyroid gland are follicles of various sizes comprised of a monolayer of epithelial cells (thyrocytes) surrounding an apical extracellular cavity known as the follicle lumen. In the normal thyroid gland, the follicle lumen is filled with secreted protein (referred to as colloid), comprised nearly exclusively of thyroglobulin with a half-life ranging from days to weeks. At the cellular boundary of the follicle lumen, secreted thyroglobulin becomes iodinated, resulting from the coordinated activities of enzymes localized to the thyrocyte apical plasma membrane. Thyroglobulin appearance in evolution is essentially synchronous with the appearance of the follicular architecture of the vertebrate thyroid gland. Thyroglobulin is the most highly expressed thyroid gene and represents the most abundantly expressed thyroid protein. Wildtype thyroglobulin protein is a large and complex glycoprotein that folds in the endoplasmic reticulum, leading to homodimerization and export via the classical secretory pathway to the follicle lumen. However, of the hundreds of human thyroglobulin genetic variants, most exhibit increased susceptibility to misfolding with defective export from the endoplasmic reticulum, triggering hypothyroidism as well as thyroidal endoplasmic reticulum stress. The human disease of hypothyroidism with defective thyroglobulin (either homozygous, or compound heterozygous) can be experimentally modeled in thyrocyte cell culture, or in whole animals, such as mice that are readily amenable to genetic manipulation. From a combination of approaches, it can be demonstrated that in the setting of thyroglobulin misfolding, thyrocytes under chronic continuous ER stress exhibit increased susceptibility to cell death, with interesting cell biological and pathophysiological consequences.


Subject(s)
Hypothyroidism , Thyroid Epithelial Cells , Mice , Humans , Animals , Thyroglobulin/metabolism , Hypothyroidism/metabolism , Thyroid Epithelial Cells/metabolism , Endoplasmic Reticulum/metabolism , Proteins/metabolism
11.
Front Endocrinol (Lausanne) ; 13: 992883, 2022.
Article in English | MEDLINE | ID: mdl-36187113

ABSTRACT

Thyroid hormones (THs), including T4 and T3, are produced and released by the thyroid gland under the stimulation of thyroid-stimulating hormone (TSH). The homeostasis of THs is regulated via the coordination of the hypothalamic-pituitary-thyroid axis, plasma binding proteins, and local metabolism in tissues. TH synthesis and secretion in the thyrocytes-containing thyroid follicles are exquisitely regulated by an elaborate molecular network comprising enzymes, transporters, signal transduction machineries, and transcription factors. In this article, we synthesized the relevant literature, organized and dissected the complex intrathyroidal regulatory network into structures amenable to functional interpretation and systems-level modeling. Multiple intertwined feedforward and feedback motifs were identified and described, centering around the transcriptional and posttranslational regulations involved in TH synthesis and secretion, including those underpinning the Wolff-Chaikoff and Plummer effects and thyroglobulin-mediated feedback regulation. A more thorough characterization of the intrathyroidal network from a systems biology perspective, including its topology, constituent network motifs, and nonlinear quantitative properties, can help us to better understand and predict the thyroidal dynamics in response to physiological signals, therapeutic interventions, and environmental disruptions.


Subject(s)
Thyroglobulin , Thyroid Gland , Feedback , Thyroglobulin/metabolism , Thyroid Gland/metabolism , Thyroid Hormones/metabolism , Thyrotropin/metabolism , Transcription Factors/metabolism
12.
J Proteomics ; 267: 104700, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35995381

ABSTRACT

Human thyroglobulin (Tg), which has many glycosylation sites, is an essential protein produced by the human thyroid glands. Although human Tg N-glycans play critical roles in the cellular events of the Thyroid gland, the site-specific distribution of glycan structures has not been studied in detail. This study aimed to profile human Tg N-glycosylation sites and their glycan contents by using high-throughput glyco-analytical strategies, including glycopeptide and glycan levels. The sulfated complex and hybrid type N-glycan species were determined by the analysis of the human Tg samples with HPLC-HILIC-FLD-MS/MS. It was found that all fucosylated N-glycans carried fucose residue on their N-glycan core structure. The human Tg was digested with multiple enzymes by applying both in-gel and in-solution protocols to enhance site-specific glycosylation analysis. In total, 17 out of 20 N-glycosylation sites were characterized. It was noticed that 6 N-glycosylation sites contain only high-mannose type glycans, while other regions include complex and hybrid type glycans. In addition, sulfated glycoform structures were detected at the glycopeptide level in glycosylation sites containing complex and hybrid type glycans. It is expected that the results obtained from this study will contribute to functional studies to be conducted on human Tg protein. BIOLOGICAL SIGNIFICANCE: N-glycans of human thyroglobulin modulate thyroid hormone synthesis both in vivo and in vitro. Therefore, a comprehensive analysis of the N-glycosylation sites of human thyroglobulin is essential to improve our understanding of the function of its N-glycans. The present research significantly expanded the knowledge regarding N-glycosylation profiles of human thyroid thyroglobulin protein. For instance, as highlighted here, sulfated N-glycan structures were characterized using comprehensive glyco-analytical strategies. N-glycan patterns for the sites Asn110, Asn1869, and Asn2122 were described for the first time in this current work. In addition, N-glycan structures containing core-fucosylation and bisecting types were confirmed for all determined glycosylation sites.


Subject(s)
Thyroglobulin , Thyroid Gland , Glycopeptides/chemistry , Glycosylation , Humans , Polysaccharides/chemistry , Tandem Mass Spectrometry , Thyroglobulin/chemistry , Thyroglobulin/metabolism , Thyroid Gland/metabolism
13.
Endocr J ; 69(10): 1217-1225, 2022 Oct 28.
Article in English | MEDLINE | ID: mdl-35644541

ABSTRACT

Solute carrier family 26 member 7 (SLC26A7), identified as a causative gene for congenital hypothyroidism, was found to be a novel iodide transporter expressed on the apical side of the follicular epithelium of the thyroid. We recently showed that TSH suppressed the expression of SLC26A7 and induces its localization to the plasma membrane, where it functions. We also showed that the ability of TSH to induce thyroid hormone synthesis is completely reversed by an autocrine negative-feedback action of thyroglobulin (Tg) stored in the follicular lumen. In the present study, we investigated the potential effect of follicular Tg on SLC26A7 expression and found that follicular Tg significantly suppressed the promoter activity, mRNA level, and protein level of SLC26A7 in rat thyroid FRTL-5 cells. In addition, follicular Tg inhibited the ability of TSH to induce the membrane localization of SLC26A7. In rat thyroid sections, the expression of SLC26A7 was weaker in follicles with a higher concentration of Tg, as evidenced by immunofluorescence staining. These results indicate that Tg stored in the follicular lumen is a feedback suppressor of the expression and membrane localization of SLC26A7, thereby downregulating the transport of iodide into the follicular lumen.


Subject(s)
Thyroglobulin , Thyroid Epithelial Cells , Animals , Rats , Antiporters/genetics , Antiporters/metabolism , Iodides/metabolism , Sulfate Transporters/genetics , Sulfate Transporters/metabolism , Thyroglobulin/genetics , Thyroglobulin/metabolism , Thyroid Epithelial Cells/metabolism , Thyroid Gland/metabolism , Thyrotropin/metabolism
14.
Neurochem Int ; 155: 105305, 2022 05.
Article in English | MEDLINE | ID: mdl-35181395

ABSTRACT

The widespread extra-thyroidal localisation of thyroid-specific proteins, thyroid-stimulating hormone receptor (TSH-R) and thyroglobulin (TG), has been well documented. However, more recent years has seen the focus of this research area shift to the distribution of these thyroid-specific proteins, in the central nervous system (CNS). This is largely attributed to the well-known associations between thyroid auto-immunity and neuro-psychiatric disorders. Although these associations have not yet been well defined, there are several studies that demonstrate the presence of TSH-R and TG proteins in CNS regions and its cellular structures. In addition, there is an emerging body of evidence to describe the potential functional roles of these thyroid proteins in various regions of the CNS. In this review, the neural distribution of TSH-R and TG as well as their possible physiological implications in various regions of human and non-human brain is discussed.


Subject(s)
Receptors, Thyrotropin , Thyroglobulin , Brain/metabolism , Humans , Receptors, Thyrotropin/metabolism , Thyroglobulin/metabolism , Thyroid Gland , Thyrotropin/metabolism
15.
Surg Oncol ; 40: 101705, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35066380

ABSTRACT

BACKGROUND: The extent of neck dissection for patients with papillary thyroid carcinoma (PTC) metastasis in lateral cervical lymph nodes is still debated. Studies aiming to omit level IIb were generally based on postoperative histopathologic information. The purpose of this study was to evaluate the predictive value of fine-needle aspiration (FNA) for level II lymph nodes in identifying candidates for neck dissection sparing level IIb before surgery. METHODS: We prospectively enrolled 156 consecutive previously untreated PTC patients with lateral neck metastases who were subjected to 178 therapeutic lateral neck dissections (including level IIa, IIb, III, IV, and Vb) between June 2018 and August 2021. Ultrasound-guided FNA of suspicious lymph nodes at level II was preoperatively performed. The cytology of FNA and thyroglobulin (Tg) washout concentration with other clinical predictors was analyzed for lymph node metastases at level IIb. RESULTS: Preoperative ultrasonography revealed suspicious lymph nodes at level II in 118 cases, and fifty were positive on FNA results. Metastasis at level IIb was seen in 17 (9.6%) of the postoperative specimens. By univariate analysis, the rate of level IIb metastasis was significantly higher in patients with FNA-positive lymph nodes at level II (P<0.001, odds ratio = 16.899). The tumor sizes of the two FNA-negative level IIb metastatic lymph nodes were 0.4 mm and 3 mm. CONCLUSIONS: Level IIb lymph node dissection may be omitted in the treatment of N1b PTC patients if FNA to level II lymph nodes is negative.


Subject(s)
Biopsy, Fine-Needle , Neck Dissection , Thyroid Cancer, Papillary/secondary , Thyroid Cancer, Papillary/surgery , Thyroid Neoplasms/pathology , Thyroid Neoplasms/surgery , Adult , Cohort Studies , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Thyroglobulin/metabolism
16.
Nat Commun ; 13(1): 61, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013249

ABSTRACT

The thyroglobulin (TG) protein is essential to thyroid hormone synthesis, plays a vital role in the regulation of metabolism, development and growth and serves as intraglandular iodine storage. Its architecture is conserved among vertebrates. Synthesis of triiodothyronine (T3) and thyroxine (T4) hormones depends on the conformation, iodination and post-translational modification of TG. Although structural information is available on recombinant and deglycosylated endogenous human thyroglobulin (hTG) from patients with goiters, the structure of native, fully glycosylated hTG remained unknown. Here, we present the cryo-electron microscopy structure of native and fully glycosylated hTG from healthy thyroid glands to 3.2 Å resolution. The structure provides detailed information on hormonogenic and glycosylation sites. We employ liquid chromatography-mass spectrometry (LC-MS) to validate these findings as well as other post-translational modifications and proteolytic cleavage sites. Our results offer insights into thyroid hormonogenesis of native hTG and provide a fundamental understanding of clinically relevant mutations.


Subject(s)
Cryoelectron Microscopy , Thyroglobulin/chemistry , Thyroglobulin/metabolism , Goiter , Humans , Iodides , Iodine , Models, Molecular , Protein Conformation , Proteolysis , Thyroglobulin/genetics , Thyroid Gland/metabolism , Thyroid Hormones/chemistry , Thyroid Hormones/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism
17.
Cell Signal ; 90: 110212, 2022 02.
Article in English | MEDLINE | ID: mdl-34896620

ABSTRACT

The TSH receptor (TSHR) is the major regulator of thyroid hormone biosynthesis in human thyrocytes by regulating the transcription of a number of genes including thyroglobulin (TG) and thyroperoxidase (TPO). Until recently, it was thought that TSHR initiated signal transduction pathways only at the cell-surface and that internalization was primarily involved in TSHR desensitization and downregulation. Studies primarily in mouse cells showed that TSHR internalization regulates gene transcription at an intracellular site also. However, this has not been shown for genes involved in thyroid hormone biosynthesis in human thyrocytes. We used human thyrocytes in primary culture. In these cells, the dose-response to TSH for gene expression is biphasic with low doses upregulating gene expression and higher doses decreasing gene expression. We used two approaches to inhibit internalization. In the first, we used inhibitors of dynamins, dynasore and dyngo-4a. Pretreatment with dynasore or dyngo-4a markedly inhibited TSH upregulation of TG and TPO mRNAs, as well as TG secretion. In the second, we used knockdown of dynamin 2, which is the most abundant dynamin in human thyrocytes. We showed that dynamin 2 knockdown inhibited TSHR internalization and decreased the TSH-stimulated levels of TG and TPO mRNAs and proteins. Lastly, we showed that the level of the activatory transcription factor phosphorylated cAMP response element binding protein (pCREB) in the cell nuclei was reduced by 68% when internalization was inhibited. We conclude that upregulation of genes involved in thyroid hormone synthesis in human thyrocytes is, in part, dependent on internalization leading to nuclear localization of an activated transcription factor(s).


Subject(s)
Iodide Peroxidase , Thyroglobulin , Animals , Humans , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Mice , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Thyroglobulin/genetics , Thyroglobulin/metabolism , Thyrotropin/genetics , Thyrotropin/pharmacology , Transcription, Genetic
18.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34916298

ABSTRACT

The thyroid maintains systemic homeostasis by regulating serum thyroid hormone concentrations. Here we report the establishment of three-dimensional (3D) organoids from adult thyroid tissue representing murine and human thyroid follicular cells (TFCs). The TFC organoids (TFCOs) harbor the complete machinery of hormone production as visualized by the presence of colloid in the lumen and by the presence of essential transporters and enzymes in the polarized epithelial cells that surround a central lumen. Both the established murine as human thyroid organoids express canonical thyroid markers PAX8 and NKX2.1, while the thyroid hormone precursor thyroglobulin is expressed at comparable levels to tissue. Single-cell RNA sequencing and transmission electron microscopy confirm that TFCOs phenocopy primary thyroid tissue. Thyroid hormones are readily detectable in conditioned medium of human TFCOs. We show clinically relevant responses (increased proliferation and hormone secretion) of human TFCOs toward a panel of Graves' disease patient sera, demonstrating that organoids can model human autoimmune disease.


Subject(s)
Gene Expression Regulation/physiology , Graves Disease/metabolism , Organoids/metabolism , Thyroid Epithelial Cells/physiology , Animals , Culture Media , Humans , Mice , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , Thyroglobulin/genetics , Thyroglobulin/metabolism , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism
19.
Acta Crystallogr D Struct Biol ; 77(Pt 11): 1451-1459, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34726172

ABSTRACT

Thyroglobulin is a homodimeric glycoprotein that is essential for the generation of thyroid hormones in vertebrates. Upon secretion into the lumen of follicles in the thyroid gland, tyrosine residues within the protein become iodinated to produce monoiodotyrosine (MIT) and diiodotyrosine (DIT). A subset of evolutionarily conserved pairs of DIT (and MIT) residues can then engage in oxidative coupling reactions that yield either thyroxine (T4; produced from coupling of a DIT `acceptor' with a DIT `donor') or triiodothyronine (T3; produced from coupling of a DIT acceptor with an MIT donor). Although multiple iodotyrosine residues have been identified as potential donors and acceptors, the specificity and structural context of the pairings (i.e. which donor is paired with which acceptor) have remained unclear. Here, single-particle cryogenic electron microscopy (cryoEM) was used to generate a high-resolution reconstruction of bovine thyroglobulin (2.3 Šresolution in the core region and 2.6 Šoverall), allowing the structural characterization of two post-reaction acceptor-donor pairs as well as tyrosine residues modified as MIT and DIT. A substantial spatial separation between donor Tyr149 and acceptor Tyr24 was observed, suggesting that for thyroxine synthesis significant peptide motion is required for coupling at the evolutionarily conserved thyroglobulin amino-terminus.


Subject(s)
Cattle , Thyroglobulin/chemistry , Animals , Cattle/metabolism , Cryoelectron Microscopy , Halogenation , Protein Conformation , Protein Domains , Protein Multimerization , Thyroglobulin/metabolism , Thyroglobulin/ultrastructure
20.
Front Endocrinol (Lausanne) ; 12: 700083, 2021.
Article in English | MEDLINE | ID: mdl-34552555

ABSTRACT

Primary cilia are sensory organelles with a variety of receptors and channels on their membranes. Recently, primary cilia were proposed to be crucial sites for exocytosis and endocytosis of vesicles associated with endocytic control of various ciliary signaling pathways. Thyroglobulin (Tg) synthesis and Tg exocytosis/endocytosis are critical for the functions of thyroid follicular cells, where primary cilia are relatively well preserved. LRP2/megalin has been detected on the apical surface of absorptive epithelial cells, including thyrocytes. LRP2/megalin on thyrocytes serves as a Tg receptor and can mediate Tg endocytosis. In this study, we investigated the role of primary cilia in LRP2/megalin expression in thyroid gland stimulated with endogenous TSH using MMI-treated and Tg-Cre;Ift88flox/flox mice. LRP2/megalin expression in thyroid follicles was higher in MMI-treated mice than in untreated control mice. MMI-treated mice exhibited a significant increase in ciliogenesis in thyroid follicular cells relative to untreated controls. Furthermore, MMI-induced ciliogenesis accompanied increases in LRP2/megalin expression in thyroid follicular cells, in which LRP2/megalin was localized to the primary cilium. By contrast, in Tg-Cre;Ift88flox/flox mice, thyroid with defective primary cilia expressed markedly lower levels of LRP2/megalin. Serum Tg levels were elevated in MMI-treated mice and reduced in Tg-Cre;Ift88flox/flox mice. Taken together, these results indicate that defective ciliogenesis in murine thyroid follicular cells is associated with impaired LRP2/megalin expression and reduced serum Tg levels. Our results strongly suggest that primary cilia harbors LRP2/megalin, and are involved in TSH-mediated endocytosis of Tg in murine thyroid follicles.


Subject(s)
Cilia/physiology , Endocytosis , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Thyroglobulin/metabolism , Thyroid Gland/metabolism , Thyrotropin/pharmacology , Tumor Suppressor Proteins/physiology , Animals , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Thyroid Gland/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...