Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 2144, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140269

ABSTRACT

While the signaling pathways and transcription factors involved in the differentiation of thyroid follicular cells, both in embryonic and adult life, are increasingly well understood, the underlying mechanisms and potential crosstalk between the thyroid transcription factors Nkx2.1, Foxe1 and Pax8 and inductive signals remain unclear. Here, we focused on the transcription factor Sox9, which is expressed in Nkx2.1-positive embryonic thyroid precursor cells and is maintained from embryonic development to adulthood, but its function and control are unknown. We show that two of the main signals regulating thyroid differentiation, TSH and TGFß, modulate Sox9 expression. Specifically, TSH stimulates the cAMP/PKA pathway to transcriptionally upregulate Sox9 mRNA and protein expression, a mechanism that is mediated by the binding of CREB to a CRE site within the Sox9 promoter. Contrastingly, TGFß signals through Smad proteins to inhibit TSH-induced Sox9 transcription. Our data also reveal that Sox9 transcription is regulated by the thyroid transcription factors, particularly Pax8. Interestingly, Sox9 significantly increased the transcriptional activation of Pax8 and Foxe1 promoters and, consequently, their expression, but had no effect on Nkx2.1. Our study establishes the involvement of Sox9 in thyroid follicular cell differentiation and broadens our understanding of transcription factor regulation of thyroid function.


Subject(s)
SOX9 Transcription Factor/metabolism , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/metabolism , Thyroid Gland/metabolism , Thyrotropin/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Cell Line , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Mice , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , Promoter Regions, Genetic , SOX9 Transcription Factor/genetics , Signal Transduction , Thyroid Gland/cytology , Thyroid Gland/embryology , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism , Thyrotropin/pharmacology , Transcription, Genetic , Transforming Growth Factor beta/pharmacology
2.
Toxicology ; 461: 152918, 2021 09.
Article in English | MEDLINE | ID: mdl-34464681

ABSTRACT

The aim of this work was to determine whether chronic exposure to nonylphenol (NP), a representative substance of environmental endocrine disruptors (EEDs), at environmental concentration would have toxic effects on thyroid function and thyroid hyperplasia disease. Two hundred SPF Sprague-Dawley rats were divided into five groups (n = 40 per group): blank control group (corn oil), low-dose NP exposure group (0.4 mg/kg/d), medium-dose NP exposure group (4 mg/kg/d), high-dose NP exposure group (40 mg/kg/d), and estradiol control group (E2: 30 µg/kg/d). The rats were treated by gavage for 34 weeks, which were sampled twice (17 weeks and 34 weeks respectively). NP accumulation in the thyroid tissue (F = 52.93, P < 0.001) and serum (F = 5.54, P = 0.00) continuously increased in a significant dose-effect relationship. After NP exposure, the serum FT3 levels exhibited a dose-dependent increasing trend (F = 4.68, P = 0.01), while the serum FT4 level showed an opposite trend (F = 3.93, P= 0.01). Compared with the control group, hyperechoic areas (i.e., calcification points) were observed in the high-dose group. Follicular epithelial stratification was extremely severe, the monolayer cubic epithelial cells became flat, and the area of single follicles was even smaller in the high-dose group. In the high-dose NP group, there were numerous mitochondria that were severely swollen. The rough endoplasmic reticulum was abundant, with obvious expansion and vesiculation. The relative expression of ERα (F = 5.29, P = 0.00), ERß (F = 10.17, P = 0.00), TRα (F = 7.71, P = 0.00), TRß (F = 3.52.17, P = 0.02) and HMGB1 (F = 10.16, P = 0.01) proteins in the thyroid tissue in each NP exposure group was increased compared with the control group, and the relative expression of proteins increased if the exposure time was prolonged under the same exposure dose. Chronic exposure to NP at environmental concentration could have toxic effects on thyroid function, and induce thyroid hyperplasia disease in male rats.


Subject(s)
Endocrine Disruptors/toxicity , Phenols/toxicity , Thyroid Epithelial Cells/drug effects , Thyroid Gland/drug effects , Animals , Dose-Response Relationship, Drug , Endocrine Disruptors/administration & dosage , Endoplasmic Reticulum/drug effects , Hyperplasia/chemically induced , Male , Phenols/administration & dosage , Rats , Rats, Sprague-Dawley , Thyroid Epithelial Cells/cytology , Thyroid Gland/pathology , Time Factors
3.
Front Endocrinol (Lausanne) ; 12: 666565, 2021.
Article in English | MEDLINE | ID: mdl-33959101

ABSTRACT

Stem cell-based therapies to reconstitute in vivo organ function hold great promise for future clinical applications to a variety of diseases. Hypothyroidism resulting from congenital lack of functional thyrocytes, surgical tissue removal, or gland ablation, represents a particularly attractive endocrine disease target that may be conceivably cured by transplantation of long-lived functional thyroid progenitors or mature follicular epithelial cells, provided a source of autologous cells can be generated and a variety of technical and biological challenges can be surmounted. Here we review the emerging literature indicating that thyroid follicular epithelial cells can now be engineered in vitro from the pluripotent stem cells (PSCs) of mice, normal humans, or patients with congenital hypothyroidism. We review the in vivo embryonic development of the thyroid gland and explain how emerging discoveries in developmental biology have been utilized as a roadmap for driving PSCs, which resemble cells of the early embryo, into mature functional thyroid follicles in vitro. Finally, we discuss the bioengineering, biological, and clinical hurdles that now need to be addressed if the goals of life-long cure of hypothyroidism through cell- and/or gene-based therapies are to be attained.


Subject(s)
Cell Differentiation , Pluripotent Stem Cells/cytology , Regenerative Medicine , Stem Cell Transplantation , Thyroid Diseases/therapy , Thyroid Epithelial Cells/cytology , Animals , Humans
4.
Front Endocrinol (Lausanne) ; 12: 652675, 2021.
Article in English | MEDLINE | ID: mdl-33953698

ABSTRACT

Thyroid cancer incidence is markedly increased in volcanic areas where residents are biocontaminated by chronic lifelong exposure to slightly increased metals in the environment. Metals can influence the biology of living cells by a variety of mechanisms, depending not only on the dose and length of exposure but also on the type and stage of differentiation of target cells. We explored the effect of five heavy metals (Cu, Hg, Pd, W and Zn) at nanomolar concentrations (the biocontamination level in residents of the volcanic area in Sicily where thyroid cancer is increased) on stimulating the proliferation of undifferentiated (thyrospheres) and differentiated human thyroid cells. Thyrosphere proliferation was significantly increased after exposure to each individual metal and a greater stimulating effect was observed when a mixture of the examined metals was used. No effect was seen in differentiated thyrocytes. For all metals, the dose-response curve followed a biphasic pattern that is typical of hormesis. Thyrosphere growth concerned the size rather than number, except with the metal mixture. An altered morphology was also observed in metal-treated thyrospheres. Metal-induced proliferation was due to activation of the ERK1/2 pathway, as confirmed by growth inhibition when ERK1/2 signaling was blocked. These studies show that stem/precursor thyroid cells are sensitive to small increases in environmental metal concentrations that are harmless for differentiated thyrocytes.


Subject(s)
Metals, Heavy/adverse effects , Neoplastic Stem Cells/cytology , Thyroid Epithelial Cells/cytology , Thyroid Gland/cytology , Thyroid Neoplasms/metabolism , Adult , Aged , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chlorides/adverse effects , Copper Sulfate/adverse effects , Culture Media , Dose-Response Relationship, Drug , Environmental Exposure , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Incidence , Mercuric Chloride/adverse effects , Microscopy, Phase-Contrast , Middle Aged , Neoplastic Stem Cells/metabolism , Palladium/adverse effects , Phosphorylation , Sicily/epidemiology , Thyroid Gland/metabolism , Thyroid Neoplasms/epidemiology , Tungsten Compounds/adverse effects , Volcanic Eruptions , Zinc Compounds/adverse effects
5.
Int J Mol Sci ; 22(9)2021 Apr 22.
Article in English | MEDLINE | ID: mdl-33922129

ABSTRACT

Recently, ER stress induced by tunicamycin (TM) was reported to inhibit the expression of key genes involved in thyroid hormone synthesis, such as sodium/iodide symporter (NIS), thyroid peroxidase (TPO) and thyroglobulin (TG), and their regulators such as thyrotropin receptor (TSHR), thyroid transcription factor-1 (TTF-1), thyroid transcription factor-2 (TTF-2) and paired box gene 8 (PAX-8), in FRTL-5 thyrocytes. The present study tested the hypothesis that resveratrol (RSV) alleviates this effect of TM in FRTL-5 cells. While treatment of FRTL-5 cells with TM alone (0.1 µg/mL) for 48 h strongly induced the ER stress-sensitive genes heat shock protein family A member 5 (HSPA5) and DNA damage inducible transcript 3 (DDIT3) and repressed NIS, TPO, TG, TSHR, TTF-1, TTF-2 and PAX-8, combined treatment with TM (0.1 µg/mL) and RSV (10 µM) for 48 h attenuated this effect of TM. In conclusion, RSV alleviates TM-induced ER stress and attenuates the strong impairment of expression of genes involved in thyroid hormone synthesis and their regulators in FRTL-5 thyrocytes exposed to TM-induced ER stress. Thus, RSV may be useful for the treatment of specific thyroid disorders, provided that strategies with improved oral bioavailability of RSV are applied.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Resveratrol/pharmacology , Thyroid Epithelial Cells/drug effects , Thyroid Gland/drug effects , Thyroid Hormones/genetics , Tunicamycin/toxicity , Animals , Anti-Bacterial Agents/toxicity , Antioxidants/pharmacology , Rats , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/metabolism , Thyroid Gland/cytology , Thyroid Gland/metabolism , Thyroid Hormones/biosynthesis
6.
Front Endocrinol (Lausanne) ; 12: 608697, 2021.
Article in English | MEDLINE | ID: mdl-33716965

ABSTRACT

Previous studies have demonstrated that, in addition to inducing structural changes in thyroid follicles, cadmium (Cd) increased the number of C cells. We examined the effects of myo-inositol (MI), seleno-L-methionine (Se), MI + Se, and resveratrol on C cells of mice exposed to cadmium chloride (Cd Cl2), as no data are currently available on the possible protective effects of these molecules. In contrast, we have previously shown this protective effect against CdCl2 on the thyroid follicles of mice. Ninety-eight C57 BL/6J adult male mice were divided into 14 groups of seven mice each: (i) 0.9% NaCl (vehicle; 1 ml/kg/day i.p.); (ii) Se (0.2 mg/kg/day per os); (iii) Se (0.4 mg/kg/day per os); (iv) MI (360 mg/kg/day per os); (v) Se (0.2 mg/kg/day) + MI; (vi) Se (0.4 mg/kg/day) + MI; (vii) resveratrol (20 mg/kg); (viii) CdCl2 (2 mg/kg/day i.p.) + vehicle; (ix) CdCl2 + Se (0.2 mg/kg/day); (x) CdCl2 + Se (0.4 mg/kg/day); (xi) CdCl2 + MI; (xii) CdCl2 + Se (0.2 mg/kg/day) + MI; (xiii) CdCl2 + Se (0.4 mg/kg/day) + MI; (xiv) CdCl2 + resveratrol (20 mg/kg). After 14 days, thyroids were processed for histological, immunohistochemical, and morphometric evaluation. Compared to vehicle, Cd significantly decreased follicle mean diameter, increased CT-positive cells number, area and cytoplasmic density, and caused the disappearance of TUNEL-positive C cells, namely, the disappearance of C cells undergoing apoptosis. Se at either 0.2 or 0.4 mg/kg/day failed to significantly increase follicular mean diameter, mildly decreased CT-positive cells number, area and cytoplasmic density, and was ineffective on TUNEL-positive C cells. Instead, MI alone increased significantly follicular mean diameter and TUNEL-positive cells number, and decreased significantly CT-positive cells number, area and cytoplasmic density. MI + Se 0.2 mg/kg/day or MI + Se 0.4 mg/kg/day administration improved all five indices more markedly. Indeed, follicular mean diameter and TUNEL-positive cells number increased significantly, while CT-positive cells number, area and cytoplasmic density decreased significantly. Thus, all five indices overlapped those observed in vehicle-treated mice. Resveratrol improved significantly all the considered parameters, with a magnitude comparable to that of MI alone. In conclusion, the association Myo + Se is effective in protecting the mouse thyroid from the Cd-induced hyperplasia and hypertrophy of C cells. This benefit adds to that exerted by Myo + Se on thyrocytes and testis.


Subject(s)
Cadmium/pharmacology , Inositol/pharmacology , Selenium/pharmacology , Thyroid Gland/drug effects , Animals , Cell Size/drug effects , Goiter/chemically induced , Goiter/pathology , Hyperplasia/chemically induced , Hypertrophy/chemically induced , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/drug effects , Thyroid Gland/cytology , Thyroid Gland/pathology
7.
Front Endocrinol (Lausanne) ; 11: 588685, 2020.
Article in English | MEDLINE | ID: mdl-33240221

ABSTRACT

The endoplasmic reticulum stress and the unfolded protein response are triggered following an imbalance between protein load and protein folding. Until recently, two possible outcomes of the unfolded protein response have been considered: life or death. We sought to substantiate a third alternative, dedifferentiation, mesenchymal shift, and activation of the antioxidant response by using typical endocrine cells, i.e. thyroid cells. The thyroid is a unique system both of endoplasmic reticulum stress (a single protein, thyroglobulin represents the majority of proteins synthesized in the endoplasmic reticulum by the thyrocyte) and of polarized epithelium (the single layer of thyrocytes delimiting the follicle). Following endoplasmic reticulum stress, in thyroid cells the folding of thyroglobulin was disrupted. The mRNAs of unfolded protein response were induced or spliced (X-box binding protein-1). Differentiation was inhibited: mRNA levels of thyroid specific genes, and of thyroid transcription factors were dramatically downregulated, at least in part, transcriptionally. The dedifferentiating response was accompanied by an upregulation of mRNAs of antioxidant genes. Moreover, cadherin-1, and the thyroid (and kidney)-specific cadherin-16 mRNAs were downregulated, vimentin, and SNAI1 mRNAs were upregulated. In addition, loss of cortical actin and stress fibers formation were observed. Together, these data indicate that ER stress in thyroid cells induces dedifferentiation, loss of epithelial organization, shift towards a mesenchymal phenotype, and activation of the antioxidant response, highlighting, at the same time, a new and wide strategy to achieve survival following ER stress, and, as a sort of the other side of the coin, a possible new molecular mechanism of decline/loss of function leading to a deficit of thyroid hormones formation.


Subject(s)
Antioxidants/metabolism , Cell Differentiation , Endoplasmic Reticulum Stress , Mesoderm/cytology , Thyroglobulin/metabolism , Thyroid Epithelial Cells/cytology , Unfolded Protein Response , Animals , Cells, Cultured , Gene Expression Regulation , Mesoderm/metabolism , Rats , Thyroid Epithelial Cells/metabolism
8.
Bull Exp Biol Med ; 169(4): 564-570, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32910397

ABSTRACT

The results of 3D culturing of human thyroid follicle-like structures in a gel based on platelet lysate at the gel-air interface are presented. During culturing up to 4 months, no new follicle-like structures were formed and none were destroyed. During the first 2 months, most follicle-like structures increased in size; then, their grown decelerated, but they retained viability. Ki-67+ cells were observed in the majority of follicle-like structures. Most of them produced thyroglobulin. Follicle-like structures get closer, the number of contacts between them increased, and cluster appeared. Thus, the developed 3D culturing system in a gel based on platelet lysate is an adequate approach for maintaining structure and functional activity of human follicle-like structures in vitro for at least 2 months.


Subject(s)
Blood Platelets/chemistry , Complex Mixtures/pharmacology , Culture Media/pharmacology , Thyroid Epithelial Cells/drug effects , Biomarkers/metabolism , Cell Size , Culture Media/chemistry , Gels , Gene Expression , Humans , Ki-67 Antigen/biosynthesis , Ki-67 Antigen/genetics , Primary Cell Culture , Thyroglobulin/biosynthesis , Thyroglobulin/genetics , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/metabolism , Thyroid Gland/cytology , Thyroid Gland/metabolism , Thyroid Gland/surgery , Thyroidectomy
9.
Mol Cell Endocrinol ; 518: 111032, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32941925

ABSTRACT

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.


Subject(s)
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
10.
Article in English | MEDLINE | ID: mdl-32760346

ABSTRACT

Background: The success in rescuing thyroid deficiency in mice using thyroid cells derived from embryonic stem (ES) cells, together with the discovery of human induced pluripotent stem cells (iPSCs) from somatic cells, has raised the possibility of patient-specific thyroid cell replacement. In this study we demonstrate that human thyroid follicular cells can be derived from human iPSCs and show the ability of highly purified and differentiated cells to secrete thyroid hormone. Research Design and Methods: Human iPSCs were derived from adult skin fibroblasts using RNA reprogramming and differentiated in vitro into thyroid follicular cells by exposure to activin A, ethacridine and TSH as we have previously described for human ES cells. The resulting thyroid cells were then highly purified using double antibody cell sorting. Results: The iPSCs derived from human dermal fibroblasts showed stem cell-like morphologic changes and expressed pluripotent stem cell markers as assessed using qPCR, immunofluorescence staining, and FACS analysis. These cells retained their pluripotential characteristics as shown by teratoma formation after murine transplantation. Definitive endoderm cells were induced with activin A and the transcription factor TAZ was significantly induced on ethacridine treatment and translocated to the nucleus. Thyroid transcription factors NKX2-1 and PAX8 were also highly expressed in activin A derived endoderm cells and further induced by ethacridine. Following terminal differentiation with TSH, there was enhanced thyroid follicle formation, high expression of the thyroid specific genes-TG, TPO, TSHR and NIS, and secretion of thyroid hormone (T4) in vitro. Furthermore, we were able to achieve a 97% purification of TSHR+/NIS+ expressing cells after differentiation using a single purification procedure. Conclusions: These findings demonstrate that mature adult dermal fibroblasts can be matured into human iPSCs which have the potential to form functional thyroid follicular cells. This lays the groundwork for future person-specific thyroid regenerative therapy.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Fibroblasts/cytology , Human Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Teratoma/pathology , Thyroid Epithelial Cells/cytology , Animals , Fibroblasts/metabolism , Gene Expression Regulation , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Nude , Middle Aged , Teratoma/metabolism , Thyroid Epithelial Cells/metabolism
12.
Article in English | MEDLINE | ID: mdl-32425890

ABSTRACT

Thyrotropin hormone (TSH) was reported to exhibit biphasic regulation of cAMP production in human thyroid slices; specifically, upregulation at low TSH doses transitioning to inhibition at high doses. We observed this phenomenon in HEK293 cells overexpressing TSH receptors (TSHRs) but in only 25% of human thyrocytes (hThyros) in vitro. Because TSHR expression in hThyros in vitro was low, we tested the hypothesis that high, in situ levels of TSHRs were needed for biphasic cAMP regulation. We increased expression of TSHRs by infecting hThyros with adenoviruses expressing human TSHR (AdhTSHR), measured TSH-stimulated cAMP production and TSHR homodimerization. TSHR mRNA levels in hThyros in vitro were 100-fold lower than in human thyroid tissue. AdhTSHR infection increased TSHR mRNA expression to levels found in thyroid tissue and flow cytometry showed that cell-surface TSHRs increased more than 15-fold. Most uninfected hThyro preparations exhibited monotonic cAMP production. In contrast, most hThyro preparations infected with AdhTSHR expressing TSHR at in vivo levels exhibited biphasic TSH dose responses. Treatment of AdhTSHR-infected hThyros with pertussis toxin resulted in monotonic dose response curves demonstrating that lower levels of cAMP production at high TSH doses were mediated by Gi/Go proteins. Proximity ligation assays confirmed that AdhTSHR infection markedly increased the number of TSHR homodimers. We conclude that in situ levels of TSHRs as homodimers are needed for hThyros to exhibit biphasic TSH regulation of cAMP production.


Subject(s)
Cyclic AMP/metabolism , Dimerization , Receptors, Thyrotropin/chemistry , Receptors, Thyrotropin/metabolism , Thyroid Epithelial Cells/metabolism , Thyroid Gland/metabolism , Cells, Cultured , Humans , In Vitro Techniques , Receptors, Thyrotropin/genetics , Signal Transduction , Thyroid Epithelial Cells/cytology , Thyroid Gland/cytology
13.
Thyroid ; 30(11): 1666-1675, 2020 11.
Article in English | MEDLINE | ID: mdl-32345138

ABSTRACT

Objective: It has been demonstrated that the transcription factors TAZ (transcriptional coactivator with PDZ-binding motif), paired box gene 8 (PAX8), and NK2 homeobox 1 (NKX2-1) are coexpressed in the nucleus of thyroid cells. Furthermore, TAZ is known to enhance the transcriptional activity of PAX8 and NKX2-1 as well as the key thyroid-specific gene, thyroglobulin (TG), suggesting a critical role for TAZ in the control of thyroid cell speciation. We previously reported that the small molecule ethacridine, identified as a TAZ activator, was able to induce thyroid-specific transcription in endodermal cells differentiated from human embryonic stem (hES) cells using activin A. Since transcription factors are epigenetically regulated in cell differentiation, we investigated the epigenetic changes in the promoter regions of these key transcription factors during in vitro differentiation of hES cells into thyrocytes. Methods: We initially profiled chromatin accessibility using the technique of Assay for Transposase Accessible Chromatin sequencing (ATAC-seq), and then examined DNA methylation and histone acetylation in the promoter regions of the three selected thyroid transcription factors and the thyroid-specific genes during hES cell differentiation. Results: ATAC-seq analysis showed enriched chromatin accessibility of TAZ, NKX2-1, and PAX8 after exposure to activin A and ethacridine. There were no methylation changes found in the NKX2-1, PAX8, and TAZ promoters by bisulfite sequencing. In contrast, acetylation of histone H4, specifically acetylation of lysine 16, was observed in each of the promoters when measured by chromatin immunoprecipitation polymerase chain reaction assays, which correlated with the activity and expression of NKX2-1 and PAX8 as well as sodium/iodide symporter, thyroid stimulating hormone receptor, and TG genes. Conclusions: These results indicate that ethacridine treatment of activin A-derived endodermal hES cells leads to enhanced chromatin accessibility, which, in turn, allows histone H4 acetylation in the regulation of active genes for speciation of thyroid follicular cells from hES cells.


Subject(s)
Cell Differentiation , DNA Methylation , Epigenesis, Genetic , Thyroid Gland/cytology , Thyroid Gland/immunology , Activins/metabolism , Chromatin/chemistry , Ethacridine/pharmacology , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Lysine , PAX8 Transcription Factor/biosynthesis , PAX8 Transcription Factor/genetics , Promoter Regions, Genetic , Sequence Analysis, DNA , Thyroid Epithelial Cells/cytology , Thyroid Nuclear Factor 1/biosynthesis , Thyroid Nuclear Factor 1/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins
14.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L908-L920, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32159371

ABSTRACT

Over 2,000 mutations have been reported in the cystic fibrosis transmembrane conductance regulator (cftr) gene, many of which cause disease but are rare and have no effective treatment. Thus, there is an unmet need for new, mutation-agnostic therapies for cystic fibrosis (CF). Phosphodiesterase (PDE) inhibitors are one such class of therapeutics that have been shown to elevate intracellular cAMP levels and stimulate CFTR-dependent anion secretion in human airway epithelia; however, the number of people with CF that could be helped by PDE inhibitors remains to be determined. Here we used Fisher rat thyroid (FRT) cells stably transduced with rare human CFTR mutants and studied their responsiveness to the dual phosphodiesterase 3/4 inhibitor RPL554 (Verona Pharma). Through its inhibitory effect on PDE4D, we find that RPL554 can elevate intracellular cAMP leading to a potentiation of forskolin-stimulated current mediated by R334W, T338I, G551D, and S549R mutants of CFTR when used alone or in combination with CFTR modulators. We also were able to reproduce these effects of RPL554 on G551D-CFTR when it was expressed in primary human bronchial epithelial cells, indicating that RPL554 would have stimulatory effects on rare CFTR mutants in human airways and validating FRT cells as a model for PDE inhibitor studies. Furthermore, we provide biochemical evidence that VX-809 causes surprisingly robust correction of several class III and IV CFTR mutants. Together, our findings further support the therapeutic potential of RPL554 for patients with CF with class III/IV mutations and emphasize the potential of PDEs as potential drug targets that could benefit patients with CF.


Subject(s)
Cyclic AMP/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Isoquinolines/pharmacology , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Pyrimidinones/pharmacology , Thyroid Epithelial Cells/drug effects , Aminopyridines/pharmacology , Animals , Benzodioxoles/pharmacology , Bronchi/cytology , Bronchi/drug effects , Bronchi/metabolism , Cell Line , Colforsin/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/agonists , Cystic Fibrosis Transmembrane Conductance Regulator/classification , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Mutation , Primary Cell Culture , Rats , Rats, Inbred F344 , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/metabolism , Transgenes
15.
Biochemistry (Mosc) ; 85(1): 108-118, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32079522

ABSTRACT

The MAPK (RAS/BRAF/MEK/ERK) signaling pathway is a kinase cascade involved in the regulation of cell proliferation, differentiation, and survival in response to external stimuli. The V600E mutation in the BRAF gene has been detected in various tumors, resulting in a 500-fold increase in BRAF kinase activity. However, monotherapy with selective BRAF V600E inhibitors often leads to reactivation of MAPK signaling cascade and emergence of drug resistance. Therefore, new targets are being developed for the inhibition of components of the aberrantly activated cascade. It was recently discovered that resistance to BRAF V600E inhibitors may be associated with the activity of the tyrosine phosphatase SHP-2 encoded by the PTPN11 gene. In this paper, we analyzed transcriptional effects of PTPN11 gene knockdown and selective suppression of BRAF V600E in a model of thyroid follicular epithelium. We found that the siRNA-mediated knockdown of PTPN11 after vemurafenib treatment prevented an increase in the expression CCNA1 and NOTCH4 genes involved in the formation of drug resistance of tumors. On the other hand, downregulation of PTPN11 expression blocked the transcriptional activation of genes (p21, p15, p16, RB1, and IGFBP7) involved in cell cycle regulation and oncogene-induced senescence in response to BRAF V600E expression. Therefore, it can be assumed that SHP-2 participates not only in emergence of drug resistance in cancer cells, but also in oncogene-induced cell senescence.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Proto-Oncogene Proteins B-raf/metabolism , Thyroid Epithelial Cells , Thyroid Neoplasms/metabolism , Cell Cycle , Cell Line , Cellular Senescence , Drug Resistance, Neoplasm/physiology , Gene Knockdown Techniques , Humans , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/metabolism , Thyroid Gland/cytology , Thyroid Gland/metabolism , Thyroid Neoplasms/pathology , Vemurafenib/therapeutic use
16.
Thyroid ; 30(1): 8-12, 2020 01.
Article in English | MEDLINE | ID: mdl-31822204

ABSTRACT

Basic research in 2019 yielded exciting discoveries and advancements in thyroidology. Specifically, there have been breakthroughs in our understanding of the molecular actions of thyroid hormone and thyroid hormone receptors, thyroid hormone metabolism and transport, autoimmunity, and thyroid cancer. Next, I summarize important studies published over the past year and whose major data I presented during the 89th American Thyroid Association annual meeting at the opening plenary session The Year in Thyroidology.


Subject(s)
Endocrinology/methods , Endocrinology/trends , Thyroid Gland/physiology , Thyroid Hormones/metabolism , Thyroid Neoplasms/metabolism , Autoimmunity , Gene Expression Regulation , Heart/physiology , Humans , Myelin Sheath/physiology , Receptors, Thyroid Hormone/physiology , Regeneration , Signal Transduction , Societies, Medical , Symporters/physiology , Thyroid Epithelial Cells/cytology
17.
Biochimie ; 166: 270-285, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31302164

ABSTRACT

Taar1 is a G protein-coupled receptor (GPCR) confined to primary cilia of rodent thyroid epithelial cells. Taar1-deficient mouse thyroid follicles feature luminal accumulation of thyroglobulin suggesting that Taar1 acts as a regulator of extra- and pericellular thyroglobulin processing, which is mediated by cysteine cathepsin proteases present at the apical plasma membrane of rodent thyrocytes. Here, by immunostaining and confocal laser scanning microscopy, we demonstrated co-localization of cathepsin L, but only little cathepsin B, with Taar1 at primary cilia of rat thyrocytes, the FRT cells. Because proteases were shown to affect half-lives of certain receptors, we determined the effect of cathepsin activity inhibition on sub-cellular localization of Taar1 in FRT cells, whereupon Taar1 localization altered such that it was retained in compartments of the secretory pathway. Since the same effect on Taar1 localization was observed in both cathepsin B and L inhibitor-treated cells, the interaction of cathepsin activities and sub-cellular localization of Taar1 was thought to be indirect. Indeed, we observed that cathepsin inhibition resulted in a lack of primary cilia from FRT cells. Next, we proved that primary cilia are a necessity for Taar1 trafficking to reach the plasma membrane of FRT cells, since the disruption of primary cilia by treatment with ß-cyclodextrin resulted in Taar1 retention in compartments of the secretory pathway. Furthermore, in less well-polarized rat thyrocytes, namely in FRTL-5 cells lacking primary cilia, Taar1 was mainly confined to the compartments of the secretory pathway. We conclude that Taar1 localization in polarized thyroid epithelial cells requires the presence of primary cilia, which is dependent on the proteolytic activity of cysteine cathepsins B and L.


Subject(s)
Cathepsin B/metabolism , Cathepsin L/metabolism , Cilia/drug effects , Endoplasmic Reticulum/metabolism , Receptors, G-Protein-Coupled/metabolism , Thyroid Epithelial Cells/metabolism , Animals , Cathepsin B/antagonists & inhibitors , Cathepsin L/antagonists & inhibitors , Cell Line , Protein Transport/drug effects , Thyroid Epithelial Cells/cytology
18.
Sci Rep ; 9(1): 10090, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31300682

ABSTRACT

Circulating T follicular helper (cTfh) cells have been identified as counterparts of germinal center Tfh (GC Tfh) cells in humans and can support T-dependent B cell maturation and antibody production in vitro. However, the role of cTfh cells in neutralizing antibody (nAb) responses in HCV infection remains unclear. Here, we characterized the phenotype and function of cTfh cells and demonstrated the associations of cTfh cells and their subsets with nAb responses in HCV infection. A total of 38 HCV-infected individuals and 28 healthy controls were enrolled from a pool of injection drug users. The frequency and function of blood Tfh cells were analyzed by flow cytometry. The titers and breadths of serum nAbs were measured using HCV pseudo-particle neutralization assays. Herein, we report several key observations. First, HCV infection skewed cTfh toward CXCR3+ cTfh cell differentiation. Second, the frequency of CXCR3+ cTfh cells positively correlated with HCV nAb titers and breadths. Third, CXCR3+ cTfh cells showed higher expression of Tfh-associated molecules (PD-1, ICOS, IL-21, Bcl-6) compared with CXCR3- cTfh cells from individuals with HCV infection. Coculture of cTfh cells and autologous memory B cells in vitro indicated that CXCR3+ cTfh cells show a superior ability to support HCV E2-specific B cell expansion compared with CXCR3- cTfh cells from individuals with HCV infection. HCV infection skews cTfh cells toward CXCR3-biased Tfh cell differentiation, which positively correlates with the magnitude and breadth of the HCV nAb response. It is our hope that these findings will provide insights for the rational design of a nAb-based HCV vaccine.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Hepacivirus/immunology , Receptors, CXCR3/blood , T-Lymphocytes, Helper-Inducer/immunology , Thyroid Epithelial Cells/immunology , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Differentiation/immunology , Cell Line , Female , Germinal Center/cytology , Germinal Center/immunology , HEK293 Cells , Hepatitis C/immunology , Humans , Lymphocyte Activation/immunology , Male , Middle Aged , T-Lymphocytes, Helper-Inducer/cytology , Thyroid Epithelial Cells/cytology
19.
Biochemistry (Mosc) ; 84(4): 416-425, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31228933

ABSTRACT

The Q61R mutation of the NRAS gene is one of the most frequent driver mutations of thyroid cancer. Tumors with this mutation are characterized by invasion into blood vessels and formation of distant metastases. To study the role of this mutation in the growth of thyroid cancer, we developed a model system on the basis of thyroid epithelial cell line Nthy-ori 3-1 transduced by a lentiviral vector containing the NRAS gene with the Q61R mutation. It was found that the expression of NRAS(Q61R) in thyroid epithelial cells has a profound influence on groups of genes involved in the formation of intercellular contacts, as well as in processes of epithelial-mesenchymal transition and cell invasion. The alteration in the expression of these genes affects the phenotype of the model cells, which acquire traits of mesenchymal cells and demonstrate increased ability for survival and growth without attachment to the substrate. The key regulators of these processes are transcription factors belonging to families SNAIL, ZEB, and TWIST, and in different types of tumors the contribution of each individual factor can vary greatly. In our model system, phenotype change correlates with an increase in the expression of SNAIL2 and TWIST2 factors, which indicates their possible role in regulating invasive growth of thyroid cancer with the mutation of NRAS(Q61R).


Subject(s)
Epithelial-Mesenchymal Transition , GTP Phosphohydrolases/metabolism , Membrane Proteins/metabolism , Thyroid Neoplasms/genetics , Transcriptome , Cell Line, Tumor , Cell Movement , Cell Proliferation , GTP Phosphohydrolases/genetics , Humans , Membrane Proteins/genetics , Mutagenesis, Site-Directed , Phenotype , Signal Transduction , Snail Family Transcription Factors/metabolism , Thyroid Epithelial Cells/cytology , Thyroid Epithelial Cells/metabolism , Twist Transcription Factors/metabolism
20.
Biochimie ; 162: 208-215, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31071356

ABSTRACT

Our group showed that repetitive dose of potassium iodide (KI) for eight days offers an efficient protection for exposure to repeated radioactive emissions without adverse effects on adult rats. However, differential expression of genes implicated in Wolff-Chaikoff effect was observed. To understand the Wolff-Chaikoff regulation and its molecular constituents during repetitive administration of KI, a biochemical reaction network was constructed as a "geographical" map of the thyrocyte depicting iodide and thyroid hormone synthesis. Path analysis of the network has been performed to investigate the presence of a regulatory circuit of the node iodide to the node "nis transcription". NIS is responsible for the uptake of KI and plays an important role in the Wolff-Chaikoff effect. The map is a source for the most updated information about iodide and thyroid hormone metabolism. Based on this map, we propose a hypothesis that shows a putative mechanism behind NIS regulation and KI uptake.


Subject(s)
Potassium Iodide/administration & dosage , Radiation Injuries/prevention & control , Symporters/metabolism , Systems Biology/methods , Thyroid Epithelial Cells/metabolism , Animals , Biological Transport , Potassium Iodide/pharmacokinetics , Potassium Iodide/pharmacology , Rats , Thyroid Epithelial Cells/cytology , Thyroid Gland/metabolism , Thyroid Hormones/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...