Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article En | MEDLINE | ID: mdl-34916298

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.


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
2.
Thyroid ; 31(8): 1151-1159, 2021 08.
Article En | MEDLINE | ID: mdl-33678005

Background: The development of the thyroid follicular cell has been well characterized as it progresses from the original stem cell, either embryonic or adult, through a series of transitions to form a differentiated and functional thyroid cell. Summary: In this review, we briefly outline what is known about this transitional process with emphasis on characterizing the thyroid progenitor stem cell by using data obtained from both in vitro and in vivo studies and both mouse and human cells. It is of particular importance to note the influence of independent factors that guide the transcriptional control of the developing thyroid cell as it is subjected to extracellular signals, often working via epigenetic changes, and initiating intrinsic transcriptional changes leading to a functional cell. Conclusion: Thyroid stem cells fall into the category of dispositional stem cells and are greatly influenced by their environment.


Stem Cells/physiology , Thyroid Epithelial Cells/physiology , Thyroid Gland/cytology , Thyroid Gland/diagnostic imaging , Animals , Humans , Mice
3.
Front Endocrinol (Lausanne) ; 12: 760541, 2021.
Article En | MEDLINE | ID: mdl-34975747

Follicular thyroid tissue originates from progenitors derived from a midline endodermal primordium. Current understanding infers that folliculogenesis in the embryonic thyroid designates the latest morphogenetic event taking place after the final anatomical shape and position of the gland is established. However, this concept does not consider the fact that the thyroid isthmus develops chronologically before the lobes and also contains all progenitors required for lobulation. To elucidate whether cells committed to a thyroid fate might be triggered to differentiate asynchronously related to maturation and developmental stage, mouse embryonic thyroid tissues from E12.5-17.5 were subjected to immunofluorescent labeling of biomarkers (progenitors: NKX2-1; differentiation: thyroglobulin/TG); folliculogenesis: E-cadherin/CDH1; luminogenesis: mucin 1/MUC1; apical polarity: pericentrin/PCNT; basement membrane: laminin; growth: Ki67), quantitative RT-PCR analysis (Nkx2.1, Tg, Muc1) and transmission electron microscopy. Tg expression was detectable as early as E12.5 and gradually increased >1000-fold until E17.5. Muc1 and Nkx2.1 transcript levels increased in the same time interval. Prior to lobulation (E12.5-13.5), MUC1 and TG distinguished pre-follicular from progenitor cells in the developing isthmus characterized by intense cell proliferation. Luminogenesis comprised redistribution of MUC1+ vesicles or vacuoles, transiently associated with PCNT, to the apical cytoplasm and the subsequent formation of MUC1+ nascent lumens. Apical polarization of pre-follicular cells and lumen initiation involved submembraneous vesicular traffic, reorganization of adherens junctions and ciliogenesis. MUC1 did not co-localize with TG until a lumen with a MUC1+ apical membrane was established. MUC1 delineated the lumen of all newly formed follicles encountered in the developing lobes at E15.5-17.5. Folliculogenesis started before establishment of a complete follicular basal lamina. These observations indicate that embryonic thyroid differentiation is an asynchronous process consistent with the idea that progenitors attaining a stationary position in the connecting isthmus portion undergo apical polarization and generate follicles already at a primordial stage of thyroid development, i.e. foregoing growth of the lobes. Although the thyroid isthmus eventually comprises minute amounts of the total thyroid volume and contributes little to the overall hormone production, it is of principal interest that local cues related to the residence status of cells - independently of a prevailing high multiplication rate - govern the thyroid differentiation program.


Basement Membrane/physiology , Cell Differentiation/physiology , Embryonic Development/physiology , Thyroid Epithelial Cells/physiology , Thyroid Gland/physiology , Animals , Basement Membrane/metabolism , Biomarkers/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Stem Cells/metabolism , Stem Cells/physiology , Thyroid Epithelial Cells/metabolism , Thyroid Gland/metabolism
4.
Mol Cell Endocrinol ; 518: 111032, 2020 12 01.
Article En | MEDLINE | ID: mdl-32941925

Thyroid transcription factors (TTFs) - NKX2-1, FOXE1, PAX8 and HHEX - regulate multiple genes involved in thyroid development in mice but little is known about TTF regulation of thyroid-specific genes - thyroglobulin (TG), thyroid peroxidase (TPO), deiodinase type 2 (DIO2), sodium/iodide symporter (NIS) and TSH receptor (TSHR) - in adult, human thyrocytes. Thyrotropin (thyroid-stimulating hormone, TSH) regulation of thyroid-specific gene expression in primary cultures of human thyrocytes is biphasic yielding an inverted U-shaped dose-response curve (IUDRC) with upregulation at low doses and decreases at high doses. Herein we show that NKX2-1, FOXE1 and PAX8 are required for TSH-induced upregulation of the mRNA levels of TG, TPO, DIO2, NIS, and TSHR whereas HHEX has little effect on the levels of these thyroid-specific gene mRNAs. We show that TSH-induced upregulation is mediated by changes in their transcription and not by changes in the degradation of their mRNAs. In contrast to the IUDRC of thyroid-specific genes, TSH effects on the levels of the mRNAs for NKX2-1, FOXE1 and PAX8 exhibit monophasic decreases at high doses of TSH whereas TSH regulation of HHEX mRNA levels exhibits an IUDRC that overlaps the IUDRC of thyroid-specific genes. In contrast to findings during mouse development, TTFs do not have major effects on the levels of other TTF mRNAs in adult, human thyrocytes. Thus, we found similarities and important differences in the regulation of thyroid-specific genes in mouse development and TSH regulation of these genes in adult, human thyrocytes.


Cell Differentiation , Thyroid Epithelial Cells/drug effects , Thyrotropin/pharmacology , Transcription, Genetic/drug effects , Adult , Autoantigens/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Humans , Iodide Peroxidase/genetics , Iron-Binding Proteins/genetics , Organ Specificity/drug effects , Organ Specificity/genetics , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/physiology , Primary Cell Culture , RNA Stability/drug effects , RNA Stability/genetics , Receptors, Thyrotropin/genetics , Thyroglobulin/genetics , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/physiology , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/physiology , Iodothyronine Deiodinase Type II
5.
Biochem Biophys Res Commun ; 531(4): 552-558, 2020 10 22.
Article En | MEDLINE | ID: mdl-32811644

Thyroid stimulating hormone deficiency is the cornerstone of treatment for metastatic thyroid cancer. Due to the loss of follicular epithelial cells in thyroid cancer, the thyroid gland degenerates to 85% of its original size. When thyroid stimulating hormone is restored, follicular epithelial cells in thyroid cancer regenerate, which is postulated to be related to stem-like cells. By single cell RNA seq, we found a group of rare thyroid follicular epithelial cells in mouse metastatic thyroid cancer, which expressed stem-like genes (CD44V6+ and CD133+) and a large number of differentiated cells (CD44V6+ and CD24+). In mouse and in organoids, the two subsets contribute equally to metastatic thyroid cancer regeneration. The analysis of human metastatic thyroid cancer revealed that the differentiated thyroid follicular epithelial cell subpopulation was similar to that of the stem like epithelial cell subpopulation, and the regeneration potential was also enhanced after thyroid stimulating hormone ablation. Accordingly, we propose that the regeneration of metastatic thyroid cancer is driven by almost all persistent thyroid follicular epithelial cells, not only by few stem-like cells.


Thyroid Epithelial Cells/physiology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , AC133 Antigen/genetics , Animals , Humans , Hyaluronan Receptors/genetics , Keratin-19/genetics , Mice, Mutant Strains , Sequence Analysis, RNA , Single-Cell Analysis , Thyroid Neoplasms/therapy , Thyrotropin/antagonists & inhibitors , Thyroxine/pharmacology , Tissue Culture Techniques , Transcriptome , Xenograft Model Antitumor Assays
7.
Int J Mol Sci ; 21(5)2020 Feb 25.
Article En | MEDLINE | ID: mdl-32106432

Exposure to environmental endocrine disruptors has been associated with an increased frequency of thyroid pathology. In this study, we evaluated the effects of various concentrations of methylmercury (MeHg) on immortalized, non-tumorigenic thyroid cells (Nthy-ori-3-1). Exposure to MeHg at 2.5 and 5 µM for 24 h caused a reduction in cell viability with a decrease of the cell population in sub-G0 phase, as detected by MTT and flow cytometry. Conversely, MeHg at the lower concentration of 0.1 µM increased the cell viability with a rise of G2/M phase. An immunoblot analysis showed higher expression levels of phospho-ERK and not of phospho-Akt. Further enhancement of the cell growth rate was observed after a prolonged exposure of the cells up to 18 days to MeHg 0.1 µM. The present findings demonstrate the toxicity of high concentrations of MeHg on thyroid cells, while showing that treatment with lower doses of Hg, as may occur after prolonged exposure to this environmental contaminant, exerts a promoting effect on thyroid cell proliferation, by acting on the ERK-mediated pro-oncogenic signal transduction pathway.


Cell Proliferation , Endocrine Disruptors/pharmacology , MAP Kinase Signaling System , Methylmercury Compounds/pharmacology , Thyroid Epithelial Cells/drug effects , Cell Line , Humans , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/physiology
8.
Int J Mol Sci ; 20(10)2019 May 24.
Article En | MEDLINE | ID: mdl-31137658

Microgravity in space or simulated by special ground-based devices provides an unusual but unique environment to study and influence tumour cell processes. By investigating thyroid cancer cells in microgravity for nearly 20 years, researchers got insights into tumour biology that had not been possible under normal laboratory conditions: adherently growing cancer cells detach from their surface and form three-dimensional structures. The cells included in these multicellular spheroids (MCS) were not only altered but behave also differently to those grown in flat sheets in normal gravity, more closely mimicking the conditions in the human body. Therefore, MCS became an invaluable model for studying metastasis and developing new cancer treatment strategies via drug targeting. Microgravity intervenes deeply in processes such as apoptosis and in structural changes involving the cytoskeleton and the extracellular matrix, which influence cell growth. Most interestingly, follicular thyroid cancer cells grown under microgravity conditions were shifted towards a less-malignant phenotype. Results from microgravity research can be used to rethink conventional cancer research and may help to pinpoint the cellular changes that cause cancer. This in turn could lead to novel therapies that will enhance the quality of life for patients or potentially develop new preventive countermeasures.


Cell Proliferation , Thyroid Epithelial Cells/physiology , Thyroid Neoplasms/pathology , Weightlessness Simulation , Animals , Humans , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , Thyroid Neoplasms/metabolism
9.
Anaerobe ; 53: 43-49, 2018 Oct.
Article En | MEDLINE | ID: mdl-29895394

Epsilon toxin (Etx) is produced by Clostridium perfringens and induces enterotoxemia in ruminants. Etx crosses the blood-brain barrier, binds to myelin structures, and kills oligodendrocytes, inducing central nervous system demyelination. In addition, Etx has a cytotoxic effect on distal and collecting kidney tubules. There are few cell lines sensitive to Etx. At present, the most sensitive in vitro model for Etx is the Madin-Darby canine kidney (MDCK) cell line, where Etx oligomerizes and forms a pore with consequent ion efflux and cell death. Although the Etx receptor has not yet been fully clarified, it is known that caveolin 1 and 2 potentiate Etx cytotoxicity and oligomerization, and more recently, the myelin and lymphocyte (MAL) protein has been implicated in Etx binding and activity. Here, we studied the effect of Etx on Fischer rat thyroid cells (FRT) and observed similar effects as those seen in MDCK cells. Etx incubated with FRT cells showed binding to the plasma membrane, and western blotting assays revealed oligomeric complex formation. Moreover, cytotoxic assays on FRT cells after Etx incubation indicated cell death at a similar level as in MDCK cells. In addition, a luminescent ATP detection assay revealed ATP depletion in FRT cells after Etx exposure. Previous studies have reported that FRT cells do not express caveolins and do not form caveolae but express MAL protein in glycolipid-enriched membrane microdomains. Our results indicate that caveolins are not directly implicated in Etx cytotoxicity, supporting the notion that the MAL protein is involved in Etx action. In addition, a cell line of thyroid origin is described for the first time as a good model to study Etx action.


Bacterial Toxins/toxicity , Clostridium perfringens/metabolism , Thyroid Epithelial Cells/drug effects , Thyroid Epithelial Cells/physiology , Adenosine Triphosphate/analysis , Animals , Blotting, Western , Cell Membrane/metabolism , Cell Survival/drug effects , Cells, Cultured , Cytosol/chemistry , Glycolipids/analysis , Myelin and Lymphocyte-Associated Proteolipid Proteins/metabolism , Protein Binding , Protein Multimerization , Rats, Inbred F344
10.
Endocr J ; 65(9): 893-901, 2018 Sep 27.
Article En | MEDLINE | ID: mdl-29877208

Hashimoto's thyroiditis (HT) is considered a T helper-type 1 (Th1) cytokine-dominant autoimmune thyroid disease. Caveolin-1 (Cav-1), a part of the thyroxisome multiprotein complex, is localized at the apical pole of thyrocytes and is indispensable for synthesis of thyroid hormones and modulation of oxidative stress in order to avoid cell damage and apoptosis. Reduced autophagy induces thyroid follicular cells (TFC) apoptosis by activating reactive oxygen species (ROS) in HT patients. Nevertheless, whether Cav-1 has roles in the regulation of autophagy remains largely unclear. In this study, we examined Th1 cytokines and Cav-1 expression in HT thyroid tissues, determined the effects of interleukin-1beta (IL-1ß) and interferon-gamma (IFN-γ) on Cav-1 and autophagy activity in TFC, and investigated the association between Cav-1 and autophagy activity in vitro. Our results indicate that higher levels of IL-1ß and IFN-γ and lower levels of Cav-1 were expressed in thyroid tissues of HT patients than in those of normal controls. Cav-1 mRNA and protein levels were significantly decreased in TFC exposed to IL-1ß and IFN-γ, accompanied by decreased expression of autophagy-related protein LC3B-II. Interestingly, small interfering RNA (siRNA)-mediated Cav-1 knockdown in TFC reduced LC3B-II protein expression. Taken together, these results suggest that lack of Cav-1 expression inhibited autophagy activity in TFC exposed to Th1 cytokines (IL-1ß and IFN-γ), which might be a novel pathogenetic mechanism of HT.


Autophagy/genetics , Caveolin 1/physiology , Hashimoto Disease/genetics , Thyroid Epithelial Cells/physiology , Autophagy/drug effects , Case-Control Studies , Caveolin 1/genetics , Cells, Cultured , Gene Knockdown Techniques , Hashimoto Disease/metabolism , Humans , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Th1 Cells/metabolism , Th1 Cells/physiology , Thyroid Epithelial Cells/drug effects , Thyroid Epithelial Cells/metabolism
11.
Front Immunol ; 9: 1197, 2018.
Article En | MEDLINE | ID: mdl-29915579

Background: Inflammasomes, which mediate maturation of interleukin-1ß (IL-ß) and interleukin-18 (IL-18) and lead to pyroptosis, have been linked to various autoimmune disorders. This study investigated whether they are involved in the pathogenesis of autoimmune thyroiditis (AIT). Methods: We collected thyroid tissues from 50 patients with AIT and 50 sex- and age-matched controls. Serum levels of free T3, free T4, thyrotropin, thyroid peroxidase antibody (TPOAb), and thyroglobulin antibody (TgAb) were measured by electrochemiluminescent immunoassays. Expression of several inflammasome components, the NOD-like receptor (NLR) family pyrin domain containing 1 (NLRP1), NLRP3, CARD-domain containing 4 (NLRC4), absent in melanoma 2 (AIM2), the apoptosis-associated speck-like protein that contains a caspase recruitment domain (ASC), caspase-1, IL-1ß, and IL-18 was determined by real-time PCR and western blot. Immunohistochemistry was used to localize the expression of NLRP1, NLRP3, NLRC4, and AIM2. The Nthy-ori 3-1 thyroid cell line was stimulated with tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), interleukin-17A, interleukin-6, and poly(dA:dT). The levels of IL-18 and IL-1ß in the cell supernatant were measured by enzyme-linked immunosorbent assay, and lactate dehydrogenase was quantified by absorptiometry. ASC specks were examined by confocal immunofluorescence microscopic analysis. Cell death was examined by flow cytometry, and the N-terminal domain of gasdermin D was detected by western blot analysis. Results: Expression of NLRP1, NLRP3, NLRC4, AIM2, ASC, caspase-1, pro IL-1ß, pro IL-18, mRNA, and protein was significantly increased in thyroid tissues from patients with AIT, and enhanced posttranslational maturation of caspase-1, IL-18 and IL-1ß was also observed. Expression of NLRP1, NLRP3, NLRC4, and AIM2 was localized mainly in thyroid follicular cells adjacent to areas of lymphatic infiltration. The thyroid mRNA level of NLRP1 and ASC was correlated to the serum TPOAb and TgAb levels in the AIT group. TNF-α and IFN-γ had a priming effect on the expression of multiple inflammasome components in thyroid cells. IFN-γ was found to strengthen poly(dA:dT)-induced cell pyroptosis and bioactive IL-18 release. Conclusion: Our work has demonstrated for the first time that multiple inflammasomes are associated with AIT pathogenesis. The identified NLRP3, NLRP1, NLRC4, AIM2 inflammasomes and their downstream cytokines may represent potential therapeutic targets and biomarkers of AIT.


Hashimoto Disease/metabolism , Inflammasomes/metabolism , Thyroid Epithelial Cells/physiology , Thyroiditis, Autoimmune/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autoantibodies/blood , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Caspase 1/metabolism , Cells, Cultured , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Hashimoto Disease/immunology , Humans , Iodide Peroxidase/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins , Pyroptosis , Thyroglobulin/immunology , Thyroiditis, Autoimmune/immunology
12.
Mol Cell Endocrinol ; 459: 21-27, 2017 Dec 25.
Article En | MEDLINE | ID: mdl-28412521

This mini review summarizes the current knowledge of the hypothalamic-pituitary-thyroid (HPT) endocrine system in lampreys, jawless vertebrates. Lampreys and hagfish are the only two extant members of the class of agnathans, the oldest lineage of vertebrates. The high conservation of the hypothalamic-pituitary-gonadal (HPG) axis in lampreys makes the lamprey model highly appropriate for comparative and evolutionary analyses. However, there are still many unknown questions concerning the hypothalamic-pituitary (HP) axis in its regulation of thyroid activities in lampreys. As an example, the hypothalamic and pituitary hormone(s) that regulate the HPT axis have not been confirmed and/or characterized. Similar to gnathostomes (jawed vertebrates), lampreys produce thyroxine (T4) and triiodothyronine (T3) from thyroid follicles that are suggested to be involved in larval development, metamorphosis, and reproduction. The existing data provide evidence of a primitive, overlapping yet functional HPG and HPT endocrine system in lamprey. We hypothesize that lampreys are in an evolutionary intermediate stage of hypothalamic-pituitary development, leading to the emergence of the highly specialized HPG and HPT endocrine axes in jawed vertebrates. Study of the ancient lineage of jawless vertebrates, the agnathans, is key to understanding the origins of the neuroendocrine system in vertebrates.


Lampreys/physiology , Neurosecretory Systems/physiology , Pituitary Gland/physiology , Reproduction/physiology , Signal Transduction , Thyroid Epithelial Cells/physiology , Animals , Biological Evolution , Gene Expression Regulation , Hypothalamus/physiology , Lampreys/classification , Metamorphosis, Biological/physiology , Phylogeny , Pituitary Hormones/genetics , Pituitary Hormones/metabolism , Thyroxine/genetics , Thyroxine/metabolism , Triiodothyronine/genetics , Triiodothyronine/metabolism
13.
Mol Cell Endocrinol ; 459: 59-63, 2017 Dec 25.
Article En | MEDLINE | ID: mdl-28267601

Thyroid hormones, or THs, are well-known regulators of a wide range of biological processes that occur throughout the lifespan of all vertebrates. THs act through genomic mechanisms mediated by thyroid hormone receptors (TRs). The main product of the thyroid gland is thyroxine or T4, which can be further transformed by different biochemical pathways to produce at least 15 active or inactive molecules. T3, a product of T4 outer-ring deiodination, has been recognized as the main bioactive TH. However, growing evidence has shown that other TH derivatives are able to bind to, and/or activate TRs, to induce thyromimetic effects. The compiled data in this review points to at least two of these TR alternative ligands: TRIAC and T2. Taking this into account, non-mammalian models have proven to be advantageous to explore new TH derivatives with potential novel actions, prompting a re-evaluation of the role and mechanism of action of TR alternative ligands that were previously believed to be inactive. The functional implications of these ligands across different vertebrates may require us to reconsider current established notions of thyroid physiology.


Diiodothyronines/metabolism , Invertebrates/metabolism , Receptors, Thyroid Hormone/metabolism , Thyroid Epithelial Cells/physiology , Thyroxine/metabolism , Triiodothyronine/analogs & derivatives , Triiodothyronine/metabolism , Animals , Biological Evolution , Fishes/classification , Fishes/genetics , Fishes/metabolism , Gene Expression Regulation , Invertebrates/classification , Invertebrates/genetics , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Ligands , Phylogeny , Receptors, Thyroid Hormone/genetics , Signal Transduction , Species Specificity , Thyroxine/genetics , Triiodothyronine/genetics
14.
Pathol Int ; 66(3): 148-157, 2016 Mar.
Article En | MEDLINE | ID: mdl-26811269

Adipose tissue (AT)-thyrocyte interaction is largely unknown. Here we described the interaction in a co-culture system, in which thyrocytes were cultured on AT fragment (ATF)-embedded collagen gel, using electron microscopy, immunocytochemistry, real-time reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). ATFs promoted the hypertrophy, polarization and lipid accumulation of thyrocytes. ATFs did not affect the growth of thyroyctes, and inhibited their apoptosis. ATFs increased the protein expression of thyroglobulin (Tg) and paired box gene 8 (PAX8) in thyrocytes. In turn, thyrocytes decreased the concentration of leptin and adiponectin, and increased the expression of these mRNAs in ATFs. Thyrotropin (TSH) enhanced the ATF-induced nuclear hypertrophy and Tg protein expression in thyrocytes, while TSH enhanced the thyrocyte-induced expression of leptin and adiponectin mRNAs in ATFs. Finally, leptin promoted the hypertrophy and Tg protein expression in thyrocytes. TSH enhanced these leptin-induced effects. The data indicate an active interaction between thyrocytes and AT, suggesting that (i) ATFs may serve to regulate the morphology, survival and differentiation of thyrocytes probably through lipid accumulation partly in a TSH-synergistic way; (ii) thyrocytes may affect adipokine production from ATFs in a TSH-independent manner; and (3) leptin may be related to the hypertrophy and differentiation of thyrocytes in a TSH-synergistic way.


Adipose Tissue/physiology , Thyroglobulin/metabolism , Thyroid Epithelial Cells/physiology , Thyrotropin/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue/cytology , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Collagen , Humans , Hypertrophy , Leptin/genetics , Leptin/metabolism , Lipid Metabolism , Male , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , RNA, Messenger/genetics , Rats, Wistar , Thyroid Epithelial Cells/cytology
...