ABSTRACT
Background: Thyroid hormone (TH) synthesis is essential for the control of development, growth, and metabolism in vertebrates and depends on a sufficient dietary iodine intake. Importantly, both iodine deficiency and iodine excess (IE) impair TH synthesis, causing serious health problems especially during fetal/neonatal development. While it is known that IE disrupts thyroid function by inhibiting thyroid gene expression, its effects on thyroid development are less clear. Accordingly, this study sought to investigate the effects of IE during the embryonic development/differentiation of endoderm and the thyroid gland. Methods: We used the murine embryonic stem (ES) cell model of in vitro directed differentiation to assess the impact of IE on the generation of endoderm and thyroid cells. Additionally, we subjected endoderm and thyroid explants obtained during early gestation to IE and evaluated gene and protein expression of endodermal markers in both models. Results: ES cells were successfully differentiated into endoderm cells and, subsequently, into thyrocytes expressing the specific thyroid markers Tshr, Slc5a5, Tpo, and Tg. IE exposure decreased the messenger RNA (mRNA) levels of the main endoderm markers Afp, Crcx4, Foxa1, Foxa2, and Sox17 in both ES cell-derived endoderm cells and embryonic explants. Interestingly, IE also decreased the expression of the main thyroid markers in ES cell-derived thyrocytes and thyroid explants. Finally, we demonstrate that DNA methyltransferase expression was increased by exposure to IE, and this was accompanied by hypermethylation and hypoacetylation of histone H3, pointing to an association between the gene repression triggered by IE and the observed epigenetic changes. Conclusions: These data establish that IE treatment is deleterious for embryonic endoderm and thyroid gene expression.
Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Endoderm/drug effects , Epigenesis, Genetic/drug effects , Gene Expression/drug effects , Sodium Iodide/pharmacology , Thyroid Gland/drug effects , Animals , Embryonic Stem Cells/cytology , Endoderm/cytology , Gene Expression Regulation, Developmental/drug effects , Mice , Thyroid Gland/cytologyABSTRACT
Several hormones regulate anuran larval development, most notably thyroid hormones (THs). In anurans, metamorphosis fails when the thyroid gland is absent or inactivated, resulting in giant tadpoles. Larval gigantism occurs naturally in neotropical frogs of the genus Pseudis as a result of a prolonged larval period. Its thyroid function is poorly investigated and the focus of this study. We describe qualitative and quantitative variations in larval development for field-captured specimens of Pseudis platensis and compare those to the development of two sympatric species, Phyllomedusa sauvagii and Pithecopus azureus, which have small tadpoles and a shorter larval period. We describe morphological changes in the thyroid glands of larval and adult specimens. In contrast to other species with similar ecological requirements, P. platensis exhibits distinct glandular activity. During premetamorphosis, there was little or no thyroid activity, a period in which the tadpole reached 70% of its maximum size. Development and degree of activity of the thyroid gland determine the duration of the early stages of the larval period. Thyroid gland histology in tadpoles appears to correlate with the TH activity, and in turn with the diversity in anuran life history transitions.
Subject(s)
Anura/anatomy & histology , Anura/growth & development , Thyroid Gland/anatomy & histology , Thyroid Gland/cytology , Animals , Body Size , Larva/anatomy & histology , Larva/growth & development , Thyroid Gland/growth & developmentABSTRACT
Thyroid cancer is the most common endocrine malignancy. Anaplastic thyroid cancer is one of the most aggressive thyroid tumors. It is known that activation of oncogenes and/or inactivation of tumor suppressor genes in tumor cells promotes tumorigenesis. The microenvironment of the tumor also plays a key role on cancer development and progression in a variety of tumors. However, the mechanisms by which tumor-stroma crosstalk in thyroid cancer remains poorly characterized. In this study we aimed to understand how interactions between fibroblasts and anaplastic thyroid cancer cells contribute to thyroid carcinogenesis. We first characterized the phenotypic changes of human fibroblasts in vitro through co-cultures by using transwells as well as by using anaplastic thyroid cancer cells-derived conditioned media. We found that fibroblasts acquired an activated phenotype or also known as cancer-associated fibroblast phenotype after being in contact with soluble factors secreted from anaplastic thyroid cancer cells, compared to the fibroblasts in mono-cultures. All the changes were partly mediated through Src/Akt activation. Treatment with the antioxidant N-acetyl-cysteine reversed in part the metabolic phenotype of activated fibroblasts. Remarkably, conditioned media obtained from these activated fibroblasts promoted cell proliferation and invasion of follicular thyroid cancer cell line, FTC-133 cells. Thus, a reciprocal and dynamic interaction exists between tumor and stromal cells, which results in the promotion of thyroid tumorigenesis. The present studies have advanced the understanding of the molecular basis of tumor-stroma communications, enabling identification and targeting of tumor-supportive mechanisms for novel treatment modalities.
Subject(s)
Adenocarcinoma, Follicular/pathology , Cancer-Associated Fibroblasts/metabolism , Stromal Cells/pathology , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/pathology , Carcinogenesis/pathology , Cell Communication , Cell Culture Techniques , Cell Dedifferentiation , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Culture Media, Conditioned/metabolism , Disease Progression , Humans , Neoplasm Invasiveness/pathology , Paracrine Communication , Thyroid Gland/cytology , Thyroid Gland/pathology , Tumor MicroenvironmentABSTRACT
Abstract Introduction: The link between Hashimoto's thyroiditis and thyroid carcinoma has long been a topic of controversy. Objective: The aim of our study was to determine the prevalence of thyroid carcinoma and Hashimoto's thyroiditis coexistence in histopathologic material of thyroidectomized patients. Methods: In a retrospective study, the clinicohistopathologic data of 2117 patients (1738 females/379 males), who underwent total or partial thyroidectomy for thyroid gland disorder at a single institution from the 1st of January 2005 to the 31st of December 2014 were analyzed. Results: Thyroid carcinoma was detected in 318 cases (15%) and microcarcinoma (thyroid cancer ≤10 mm in diameter) was found in permanent sections in 169 cases (8%). Hashimoto's thyroiditis was detected in 318 (15%) patients. Hashimoto's thyroiditis was significantly more often associated with thyroid carcinoma and microcarcinoma compare to benign condition (p = 0.048, p = 0.00014, respectively). Coexistence of Hashimoto's thyroiditis and thyroid carcinoma/thyroid microcarcinoma did not affect tumor size (p = 0.251, p = 0.098, respectively), or tumor multifocality (p = 0.831, p = 0.957, respectively). Bilateral thyroid microcarcinoma was significantly more often detected when Hashimoto's thyroiditis was also diagnosed (p = 0.041), but presence of Hashimoto's thyroiditis did not affect bilateral occurrence of thyroid carcinoma (p = 0.731). Conclusion: Hashimoto's thyroiditis is associated with significantly increased risk of developing thyroid carcinoma, especially thyroid microcarcinoma.
Resumo: Introdução: A relação entre a tireoidite de Hashimoto e o carcinoma de tireoide tem sido um tema de controvérsia por um longo tempo. Objetivo: Determinar a prevalência da coexistência de carcinoma de tireoide e tireoidite de Hashimoto no exame histopatológico de amostras de pacientes tireoidectomizados. Método: Em um estudo retrospectivo, foram analisados os dados clinico-histopatológicos de 2.117 pacientes (1.738 mulheres/379 homens), submetidos à tireoidectomia total ou parcial por distúrbio da glândula tireoide em uma única instituição, de 1º de janeiro de 2005 a 31 de dezembro de 2014. Resultados: O carcinoma de tireoide foi detectado em 318 casos (15%) e o microcarcinoma (câncer de tireoide ≤ 10 mm de diâmetro) foi encontrado em secções permanentes em 169 casos (8%). A tireoidite de Hashimoto foi detectada em 318 (15%) pacientes e foi associada ao carcinoma da tireoide e ao microcarcinoma com maior frequência em comparação com condições benignas (p = 0,048, p = 0,00014, respectivamente). A coexistência de tireoidite de Hashimoto e carcinoma/microcarcinoma não influenciou o tamanho do tumor (p = 0,251, p = 0,098, respectivamente) ou a multifocalidade tumoral (p = 0,831, p = 0,957, respectivamente). O microcarcinoma de tireoide bilateral foi detectado com maior frequência quando a tireoidite de Hashimoto também foi diagnosticada (p = 0,041), mas a presença de tireoidite não influenciou na ocorrência bilateral de carcinoma (p = 0,731). Conclusão: A tireoidite de Hashimoto está associada a um aumento significativo do risco do desenvolvimento de carcinoma de tireoide, especialmente microcarcinoma da tireoide.
Subject(s)
Humans , Male , Female , Adult , Middle Aged , Aged , Thyroid Neoplasms/epidemiology , Carcinoma, Papillary/epidemiology , Hashimoto Disease/epidemiology , Particle Size , Thyroid Gland/cytology , Thyroidectomy , Thyroid Neoplasms/pathology , Carcinoma, Papillary/pathology , Comorbidity , Prevalence , Retrospective Studies , Risk Factors , Slovakia/epidemiology , Biopsy, Fine-Needle , Hashimoto Disease/pathologyABSTRACT
INTRODUCTION: The link between Hashimoto's thyroiditis and thyroid carcinoma has long been a topic of controversy. OBJECTIVE: The aim of our study was to determine the prevalence of thyroid carcinoma and Hashimoto's thyroiditis coexistence in histopathologic material of thyroidectomized patients. METHODS: In a retrospective study, the clinicohistopathologic data of 2117 patients (1738 females/379 males), who underwent total or partial thyroidectomy for thyroid gland disorder at a single institution from the 1st of January 2005 to the 31st of December 2014 were analyzed. RESULTS: Thyroid carcinoma was detected in 318 cases (15%) and microcarcinoma (thyroid cancer ≤10mm in diameter) was found in permanent sections in 169 cases (8%). Hashimoto's thyroiditis was detected in 318 (15%) patients. Hashimoto's thyroiditis was significantly more often associated with thyroid carcinoma and microcarcinoma compare to benign condition (p=0.048, p=0.00014, respectively). Coexistence of Hashimoto's thyroiditis and thyroid carcinoma/thyroid microcarcinoma did not affect tumor size (p=0.251, p=0.098, respectively), or tumor multifocality (p=0.831, p=0.957, respectively). Bilateral thyroid microcarcinoma was significantly more often detected when Hashimoto's thyroiditis was also diagnosed (p=0.041), but presence of Hashimoto's thyroiditis did not affect bilateral occurrence of thyroid carcinoma (p=0.731). CONCLUSION: Hashimoto's thyroiditis is associated with significantly increased risk of developing thyroid carcinoma, especially thyroid microcarcinoma.
Subject(s)
Carcinoma, Papillary/epidemiology , Hashimoto Disease/epidemiology , Thyroid Neoplasms/epidemiology , Adult , Aged , Biopsy, Fine-Needle , Carcinoma, Papillary/pathology , Comorbidity , Female , Hashimoto Disease/pathology , Humans , Male , Middle Aged , Particle Size , Prevalence , Retrospective Studies , Risk Factors , Slovakia/epidemiology , Thyroid Gland/cytology , Thyroid Neoplasms/pathology , ThyroidectomyABSTRACT
Exposure to ionizing radiation greatly increases the risk of developing papillary thyroid carcinoma (PTC), especially during childhood, mainly due to gradual inactivation of DNA repair genes and DNA damages. Recent molecular characterization of PTC revealed DNA methylation deregulation of several promoters of DNA repair genes. Thus, epigenetic silencing might be a plausible mechanism for the activity loss of tumor suppressor genes in radiation-induced thyroid tumors. Herein, we investigated the impact of ionizing radiation on global methylation and CpG islands within promoter regions of homologous recombination (HR) and non-homologous end joining (NHEJ) genes, as well as its effects on gene expression, using two well-established normal differentiated thyroid cell lines (FRTL5 and PCCL3). Our data reveal that X-ray exposure promoted G2/M arrest in normal thyroid cell lines. The FRTL5 cells displayed a slower kinetics of double-strand breaks (DSB) repair and a lower long interspersed nuclear element-1 (LINE-1) methylation than the PCCL3 cells. Nevertheless, acute X-ray exposure does not alter the expression of genes involved in HR and NHEJ pathways, apart from the downregulation of Brca1 in thyroid cells. On the other hand, HR and NHEJ gene expressions were upregulated in radiation-induced senescent thyroid cells. Taken together, these data suggest that FRTL5 cells intrinsically have less efficient DNA DSB repair machinery than PCCL3 cells, as well as genomic instability, which could predispose the FRTL5 cells to unrepaired DSB lesions and, therefore, gene mutations.
Subject(s)
BRCA1 Protein/genetics , DNA Methylation/radiation effects , DNA Repair/radiation effects , Gene Expression Regulation/radiation effects , Long Interspersed Nucleotide Elements/genetics , Thyroid Gland/cytology , Animals , Cell Line , Cellular Senescence/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/radiation effects , Homologous Recombination/radiation effects , Kinetics , Phenotype , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Thyroid Gland/metabolism , Thyroid Gland/radiation effects , Up-Regulation/radiation effectsABSTRACT
Iodide has direct effects on thyroid function. Several iodinated lipids are biosynthesized by the thyroid and they were postulated as intermediaries in the action of iodide. Among them, 2-iodohexadecanal (2-IHDA) has been identified and proposed to play a role in thyroid autoregulation. The aim of this study was to compare the effect of iodide and 2-IHDA on thyroid cell physiology. For this purpose, FRTL-5 thyroid cells were incubated with the two compounds during 24 or 48 h and several thyroid parameters were evaluated such as: iodide uptake, intracellular calcium and H2O2 levels. To further explore the molecular mechanism involved in 2-IHDA action, transcript and protein levels of genes involved in thyroid hormone biosynthesis, as well as the transcriptional expression of these genes were evaluated in the presence of iodide and 2-IHDA. The results obtained indicate that 2-IHDA reproduces the action of excess iodide on the "Wolff-Chaikoff" effect as well as on thyroid specific genes transcription supporting its role in thyroid autoregulation.
Subject(s)
Aldehydes/pharmacology , Thyroid Gland/cytology , Thyroid Gland/physiology , Animals , Calcium/metabolism , Cell Line , Deoxyglucose/metabolism , Gene Expression Regulation/drug effects , Hydrogen Peroxide/metabolism , Iodides/metabolism , Organ Specificity/genetics , Promoter Regions, Genetic/genetics , Rats , Thyroid Hormones/biosynthesis , Transcription Factors/metabolism , Transcription, Genetic/drug effectsABSTRACT
Immunoreactive proteins in follicular cells, fibroblasts and endothelial cells were assessed in canine thyroid carcinomas and healthy thyroid glands. No differences were detected in thyrotropin receptor and thyroglobulin staining between cancer and normal tissues, but expression was higher in follicular cells than in fibroblasts. Fibroblast growth factor-2 staining was more intense in healthy follicular cells than in those of carcinomas. Follicular cells in carcinomas presented two- to three-fold greater staining intensity of thyroid transcription factor-1 and proliferating cell nuclear antigen, respectively, than healthy cells, and a similar trend was found for the latter antigen in fibroblasts. Vascular endothelial growth factor staining was more intense in the endothelial cells of tumours than in those of normal tissues. In conclusion, greater expression of factors related to proliferation and angiogenesis was demonstrated in several cell types within thyroid carcinomas compared to healthy tissues, which may represent mechanisms of tumour progression in this disease.
Subject(s)
Carcinoma/veterinary , Dog Diseases/pathology , Immunohistochemistry/veterinary , Thyroid Gland/metabolism , Thyroid Neoplasms/veterinary , Animals , Biomarkers , Carcinoma/pathology , Case-Control Studies , Dogs , Female , Gene Expression Regulation, Neoplastic , Male , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Thyroglobulin/genetics , Thyroglobulin/metabolism , Thyroid Gland/cytology , Thyroid Gland/pathology , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/metabolismABSTRACT
PURPOSE OF REVIEW: Thyroid hormones are essential for normal development, growth, and metabolism. Their synthesis occurs in thyroid follicles and requires an adequate iodide supply and a sequence of regulated biochemical steps. The uptake of iodide into thyrocytes is well characterized, but its efflux at the apical membrane is poorly understood. This review discusses potential mechanisms underlying iodide efflux with emphasis on recent developments and controversies. RECENT FINDINGS: The functional characterization of pendrin (PDS/SLC26A4), a multifunctional anion exchanger, suggested that it could be involved in mediating iodide efflux. This is supported by the phenotype of patients with Pendred syndrome (deafness, goiter, partial iodide organification defect), which is caused by biallelic mutations in the SLC26A4 gene, as well as functional studies. However, apical iodide efflux is also possible in the absence of pendrin, implicating the presence of at least another channel. Recently, Anoctamin 1 (TMEM16A), a calcium-activated anion channel has been identified at the apical membrane of thyrocytes and functional studies suggest that it may play a predominant role in mediating iodide efflux. SUMMARY: Anoctamin and pendrin are two plausible candidates as mediators of apical iodide efflux. Their relative affinity for iodide and their exact physiological role await, however, further characterization.
Subject(s)
Chloride Channels/physiology , Iodides/metabolism , Membrane Transport Proteins/physiology , Neoplasm Proteins/physiology , Thyroid Gland/metabolism , Animals , Anoctamin-1 , Chloride Channels/genetics , Goiter, Nodular , Hearing Loss, Sensorineural , Humans , Membrane Transport Proteins/genetics , Neoplasm Proteins/genetics , Sulfate Transporters , Thyroid Gland/cytology , Thyroid Hormones/biosynthesisABSTRACT
UNLABELLED: It is well known that pituitary TSH exerts the major task in the regulation of thyroid function. However, this gland is capable of certain degree of autonomy, independently of TSH control. Iodine plays an important role in thyroid physiology and biochemistry. The thyroid is capable of producing different iodolipids such as 2-iodohexadecanal (2-IHDA). It was shown that this iodolipid mimic some of the inhibitory effects of excess iodide on several thyroid parameters. OBJECTIVES: To identify the miRNAs regulated by 2-IHDA in rat thyroid cells and likely characterize their role in thyroid cell proliferation and function. RESULTS: FRTL-5 cells were grown in the presence of TSH and treated with 2-IHDA. Among the miRNAs up-regulated by 2-IHDA we focused on miR-let-7f and miR-138. When we transfected the miRNAs, miR-let-7f but not miR-138 overexpression inhibited proliferation of FRTL 5 cells, while miR-let-7f inhibition restored cell growth in 2-IHDA treated cultures. Analysis of cell cycle by flow cytometric DNA analysis revealed that miR-let-7f inhibition reduced the percentage of 2-IHDA treated cells in G1 phase and an increased of the percentage of cells in S phase was observed upon anti-let-7f transfection. The expresion of Cyclin D1 and Cyclin D3 were reduced after the transfection of miR-let-7f and miR-138, respectively. In in vivo studies we observed that miR-let-7f and miR-138 were up regulated by 2-IHDA during goiter involution. CONCLUSION: These results suggest that the inhibitory effects of 2-IHDA on FRTL-5 thyroid cell proliferation are mediated in part through the induction of let-7f microRNA.
Subject(s)
Aldehydes/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Thyroid Gland/cytology , Thyroid Gland/drug effects , Animals , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Rats , Thyrotropin/pharmacology , Up-RegulationABSTRACT
UNLABELLED: Although thyroid gland function is mainly under the control of pituitary TSH, other factors, such as iodine, play a role in this process. The thyroid is capable of producing different iodolipids such as 6-iodo-deltalactone and 2-iodohexadecanal (2-IHDA). It was shown that these iodolipids mimic some of the inhibitory effects of excess iodide on several thyroid parameters. OBJECTIVES: To study the effect of 2-IHDA on cell proliferation and apoptosis in FRTL-5 cells. RESULTS: FRTL-5 cells were grown in the presence of TSH and treated with increasing concentrations of KI and 2-IHDA (0.5, 5, 10 and 33 µM) for 24, 48 and 72 h. Whereas KI inhibited cell proliferation only at 33 µM after 72 h of treatment, 2-IHDA inhibited in a time and concentration dependent manner. Analysis of cell cycle by flow cytometric DNA analysis revealed an accumulation of cells in G1 phase induced by 2-IHDA. The expression of cyclin A, cyclin D1 and cyclin D3 were reduced after treatment with 2-IHDA whereas CDK4 and CDK6 proteins were not modified. 2-IHDA induced a dynamic change in cytoplasmic to nuclear accumulation of p21 and p27 causing these proteins to be accumulated mostly in the nucleus. We also observed evidence of a pro-apoptotic effect of 2-IHDA at highest concentrations. No significant effect of KI was observed. CONCLUSION: These results suggest that the inhibitory effects of 2-IHDA on FRTL-5 thyroid cell proliferation are mediated by cell cycle arrest in G1/S phase and cell death by apoptosis.
Subject(s)
Aldehydes/pharmacology , Cell Cycle Checkpoints/drug effects , Thyroid Gland/cytology , Thyrotropin/pharmacology , Animals , Apoptosis , Cell Line , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cyclins/metabolism , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Rats , Thyroid Gland/drug effectsABSTRACT
Iodide uptake by thyroid cells is mediated by a transmembrane glycoprotein known as the Na+/I--symporter (NIS). NIS-mediated iodide uptake plays important physiological role in thyroid gland function, as well as in diagnostic and treatment of Graves' disease and thyroid cancer. Although different studies investigated the transcriptional mechanisms of NIS expression, there is no report on the NIS post-translational regulation related to NIS protein degradation in thyroid cells. Recently, our group showed that AMP-activated protein kinase (AMPK) plays a pivotal role in the rat thyroid gland, downregulating iodide uptake, NIS protein, and mRNA content. Since several studies demonstrated that AMPK regulates post-transcriptional mechanisms, such as autophagy-mediated processes in different tissues, we hypothesized that AMPK activation could also regulate NIS protein degradation through the lysosome pathway in thyroid cells. Rat follicular thyroid PCCL3 cells cultivated in Ham's F12 supplemented with 5% calf serum and hormones were exposed to the AMPK pharmacological activator 5-aminoimidazole-4 carboxamide ribonucleoside (AICAR), in the presence or absence of Bafilomycin A1 or MG132 for 24 h. Treatment of PCCL3 cells with Bafilomycin A1 fully prevented the decrease of iodide uptake and NIS protein content mediated by AMPK activation. In contrast, the treatment with MG132 was unable to prevent the effects of AMPK activation on NIS. Our results show that AMPK activation significantly induces NIS protein degradation through a lysosome-mediated mechanism.
Subject(s)
AMP-Activated Protein Kinases/metabolism , Lysosomes/metabolism , Symporters/metabolism , Thyroid Gland/cytology , AMP-Activated Protein Kinases/genetics , Animals , Enzyme Activation , Iodides/metabolism , Lysosomes/enzymology , Proteolysis , Rats , Symporters/genetics , Thyroid Gland/enzymology , Thyroid Gland/metabolism , Thyrotropin/metabolismABSTRACT
BACKGROUND: Excess iodine inhibits thyroid follicular cell proliferation associated with TGFß pathway activation, although thyroid cancers are frequently refractory to TGFß signaling. The TGFß pathway is predicted to be regulated by miR-17-92 cluster microRNAs. MicroRNAs are small noncoding RNAs that inhibit target mRNA translation and have emerged as potent modulators of tumorigenesis. Although the BRAF(V600E) mutation is the most prevalent alteration in thyroid cancer, the impact of iodine intake on BRAF-mediated oncogenesis remains unclear. Therefore, the aim of this study was to investigate the influence of high iodine on miR-17-92 transcriptional regulation and expression in thyroid cells expressing activated BRAF. METHODS: Rat thyroid follicular cells that conditionally express BRAF(V600E) under doxycycline stimulation (PC-BRAF(V600E)-6) were derived from the PCCl3 line. These cells were treated with doxycycline for two days, in the absence or presence of 10 µM sodium iodide. The thyroid cancer cell lines BCPAP and KTC2 were also analyzed. Expression of the miR-17-92 cluster and Notch1 was analyzed by quantitative polymerase chain reaction, and expression of these genes was modulated by anti-miR or anti-Notch1 siRNAs transfection. Protein expression was assessed by Western blot. Luciferase assays were used to quantify Smad4 3'-UTR/miR-19 interaction and Notch signaling activation. TGFß responsiveness was evaluated by cell cycle analysis of TGFß-treated cells. RESULTS: High iodine blocked BRAF(V600E)-induced upregulation of miR-17-92, including miR-19a/b. miR-17-92 promoter region analysis revealed a putative binding site for Hes1, a transcription factor responsive to Notch signaling. Notch-1 overexpression resulted in miR-19 upregulation in normal thyroid cells, while Notch-1 knockdown blocked BRAF-induced miR-19 expression. Moreover, in anaplastic thyroid cancer cells, Notch-1 knockdown reduced miR-19. Expression of BRAF(V600E) decreased Smad4 protein in normal thyroid cells. Smad4 was validated as a miR-19 target by luciferase assays, which revealed reduced luminescence associated with miR-19 interaction in Smad4 3'-UTR. Iodine treatment restored Smad4 levels in BRAF-activated cells, resulting in enhanced G1-cell cycle arrest in response to TGFß. Moreover, this effect was mimicked in papillary thyroid cancer cells treated with anti-miR-19. CONCLUSION: High iodine abrogates BRAF(V600E)-induced activation of miR-19, a newly identified Smad4 regulator, through Notch pathway inhibition and restores responsiveness to TGFß signaling. Our results indicate that iodine exerts protective effects in thyroid cells, attenuating acute BRAF oncogene-mediated microRNA deregulation.
Subject(s)
MicroRNAs/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Sodium Iodide/pharmacology , Thyroid Gland/drug effects , Transforming Growth Factor beta/pharmacology , Animals , Rats , Thyroid Gland/cytology , Thyroid Gland/metabolismABSTRACT
The extracellular-matrix protein laminin forms polymers both in vivo and in vitro. Acidification of pH leads to the formation of an artificial polymer with biomimetic properties, named polylaminin (polyLM). Follicle cells in the thyroid are in close contact with laminin, but their response to this important extracellular signal is still poorly understood. PCCL3 thyroid follicular cells cultured on glass, on regular laminin (LM) or on laminin previously polymerized in acidic pH (polyLM) showed different cell morphologies and propensities to proliferate, as well as differences in the organization of their actin cytoskeleton. On polyLM, cells displayed a typical epithelial morphology and radially organized actin fibers; whereas on LM, they spread irregularly on the substrate, lost cell contacts, and developed thick actin fibers extending through the entire cytoplasm. Iodide uptake decreased similarly in response to both laminin substrates, in comparison to glass. On both the LM and polyLM substrates, the expression of the sodium iodide symporter (NIS) decreased slightly but not significantly. NIS showed dotted immunostaining at the plasma membrane in the cells cultured on glass; on polyLM, NIS was observed mainly in the perinuclear region, and more diffusely throughout the cytoplasm on the LM substrate. Additionally, polyLM specifically favored the maintenance of cell polarity in culture. These findings indicate that PCCL3 cells can discriminate between LM and polyLM and that they respond to the latter by better preserving the phenotype observed in the thyroid tissue.
Subject(s)
Laminin/pharmacology , Peptides/pharmacology , Thyroid Gland/drug effects , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Biological Transport , Cell Line , Cell Polarity/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression , Hydrogen-Ion Concentration , Peptides/chemistry , Polymerization , Rats , Rats, Inbred F344 , Sodium Iodide/metabolism , Symporters/genetics , Symporters/metabolism , Thyroid Gland/cytology , Thyroid Gland/metabolismABSTRACT
Hexachlorobenzene (HCB) is an organochlorine pesticide widely distributed in the environment. We have previously shown that chronic HCB exposure triggers apoptosis in rat thyroid follicular cells. This study was carried out to investigate the molecular mechanism by which the pesticide causes apoptosis in FRTL-5 rat thyroid cells exposed to HCB (0.005, 0.05, 0.5, and 5µM) for 2, 6, 8, 24, and 48h. HCB treatment lowered cell viability and induced apoptotic cell death in a dose- and time-dependent manner, as demonstrated by morphological nuclear changes and the increase of DNA fragmentation. The pesticide increased activation of caspases-3, -8, and full-length caspase-10 processing. HCB induced mitochondrial membrane depolarization, release of cytochrome c and apoptosis-inducing factor (AIF), from the mitochondria to the cytosol, and AIF nuclear translocation. Cell death was accompanied by an increase in reactive oxygen species (ROS) generation. Blocking of ROS production, with a radical scavenger (Trolox), resulted in inhibition of AIF nuclear translocation and returned cells survival to control levels, demonstrating that ROS are critical mediators of HCB-induced apoptosis. The pesticide increased ERK1/2, JNK, and p38 phosphorylation in a time- and dose-dependent manner. However, when FRTL-5 cells were treated with specific MAPK inhibitors, only blockade of MEK1/2 with PD98059 prevented cell loss of viability, as well as caspase-3 activation. In addition, we demonstrated that HCB-induced production of ROS has a critical role in ERK1/2 activation. These results demonstrate for the first time that HCB induces apoptosis in FRTL-5 cells, by ROS-mediated ERK1/2 activation, through caspase-dependent and -independent pathways.
Subject(s)
Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Hexachlorobenzene/toxicity , Reactive Oxygen Species/metabolism , Thyroid Gland/drug effects , Animals , Caspases/metabolism , Cell Line , Enzyme Activation , Mitochondria/drug effects , Phosphorylation , Rats , Thyroid Gland/cytology , Thyroid Gland/metabolismABSTRACT
Rab1b belongs to the Rab-GTPase family that regulates membrane trafficking and signal transduction systems able to control diverse cellular activities, including gene expression. Rab1b is essential for endoplasmic reticulum-Golgi transport. Although it is ubiquitously expressed, its mRNA levels vary among different tissues. This work aims to characterize the role of the high Rab1b levels detected in some secretory tissues. We report that, in HeLa cells, an increase in Rab1b levels induces changes in Golgi size and gene expression. Significantly, analyses applied to selected genes, KDELR3, GM130 (involved in membrane transport), and the proto-oncogene JUN, indicate that the Rab1b increase acts as a molecular switch to control the expression of these genes at the transcriptional level, resulting in changes at the protein level. These Rab1b-dependent changes require the activity of p38 mitogen-activated protein kinase and the cAMP-responsive element-binding protein consensus binding site in those target promoter regions. Moreover, our results reveal that, in a secretory thyroid cell line (FRTL5), Rab1b expression increases in response to thyroid-stimulating hormone (TSH). Additionally, changes in Rab1b expression in FRTL5 cells modify the specific TSH response. Our results show, for the first time, that changes in Rab1b levels modulate gene transcription and strongly suggest that a Rab1b increase is required to elicit a secretory response.
Subject(s)
Golgi Apparatus/metabolism , Thyroid Gland/metabolism , Transcription, Genetic , rab1 GTP-Binding Proteins/genetics , Biological Transport , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Gene Expression Regulation/drug effects , Golgi Apparatus/ultrastructure , HeLa Cells , Humans , Proto-Oncogene Mas , Signal Transduction , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyrotropin/metabolism , Thyrotropin/pharmacology , rab1 GTP-Binding Proteins/metabolismABSTRACT
MicroRNAs (miRNA) are small non-coding RNAs involved in post-transcriptional gene regulation that have crucial roles in several types of tumors, including papillary thyroid carcinoma (PTC). miR-146b-5p is overexpressed in PTCs and is regarded as a relevant diagnostic marker for this type of cancer. A computational search revealed that miR-146b-5p putatively binds to the 3' untranslated region (UTR) of SMAD4, an important member of the transforming growth factor ß (TGF-ß) signaling pathway. The TGF-ß pathway is a negative regulator of thyroid follicular cell growth, and the mechanism by which thyroid cancer cells evade its inhibitory signal remains unclear. We questioned whether the modulation of the TGF-ß pathway by miR-146b-5p can contribute to thyroid tumorigenesis. Luciferase reporter assay confirmed the direct binding of miR-146b-5p on the SMAD4 3'UTR. Specific inhibition of miR-146b-5p with a locked nucleic acid-modified anti-miR-146b oligonucleotide significantly increased SMAD4 levels in the human papillary carcinoma cell lines, TPC-1 and BCPAP. Moreover, suppression of miR-146b-5p increased the cellular response to the TGF-ß anti-proliferative signal, significantly decreasing the proliferation rate. The overexpression of miR-146b-5p in normal rat follicular PCCL3 cells decreased SMAD4 levels and disrupted TGF-ß signal transduction. MiR-146b-5p overexpression in PCCL3 cells also significantly increased cell proliferation in the absence of thyroid-stimulating hormone and conferred resistance to TGF-ß-mediated cell-cycle arrest. Additionally, the activation of thyroid most common oncogenes RET/PTC3 and BRAF in PCCL3 cells upregulated miR-146b-5p expression. Our results confirm the oncogenic role of miR-146b-5p in thyroid follicular cells and contribute to knowledge regarding the modulation of TGF-ß signal transduction by miRNAs in PTCs.
Subject(s)
Carcinoma, Papillary/genetics , MicroRNAs/physiology , Oncogenes , Smad4 Protein/metabolism , Thyroid Neoplasms/genetics , Transforming Growth Factor beta/metabolism , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/antagonists & inhibitors , Rats , Signal Transduction , Smad4 Protein/genetics , Thyroid Gland/cytologyABSTRACT
Iodide excess acutely downregulates NIS mRNA expression, as already demonstrated. PCCl3 cells treated or not with NaI, NaI+NaClO(4) or NaI+Methimazole, for 30 min to 24 h, were used to further explore how iodide reduces NIS gene expression. NIS mRNA expression was evaluated by Real-Time PCR; its poly(A) tail length, by RACE-PAT; its translation rate, by polysome profile; total NIS content, by Western blotting. NIS mRNA decay rate was evaluated in actinomycin-D-treated cells, incubated with or without NaI for 0-6 h. Iodide treatment caused a reduction in NIS mRNA expression, half-life, poly(A) tail length, recruitment to ribosomes, as well as NIS protein expression. Perchlorate, but not methimazole, prevented these effects. Therefore, reduced poly(A) tail length of NIS mRNA seems to be related to its decreased half-life, in addition to its translation impairment. These data provide new insights about the molecular mechanisms involved in the rapid and posttranscriptional inhibitory effect of iodide on NIS expression.
Subject(s)
Gene Expression/drug effects , RNA Processing, Post-Transcriptional , Sodium Iodide/pharmacology , Symporters/metabolism , Thyroid Gland/drug effects , Animals , Antithyroid Agents/pharmacology , Blotting, Western , Cell Line , Half-Life , Methimazole/pharmacology , Perchlorates/pharmacology , Polyribosomes/chemistry , Polyribosomes/drug effects , Polyribosomes/genetics , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Sodium Compounds/pharmacology , Symporters/antagonists & inhibitors , Symporters/genetics , Thyroid Gland/cytology , Thyroid Gland/physiologyABSTRACT
The aim of this study was to investigate the role of AMP-kinase (AMPK) in the regulation of iodide uptake by the thyroid gland. Iodide uptake was assessed in PCCL3 follicular thyroid cells exposed to the AMPK agonist 5-aminoimidazole-4-carboxamide-ribonucleoside (AICAR), and also in rat thyroid glands 24 h after a single intraperitoneal injection of AICAR. In PCCL3 cells, AICAR-induced AMPK and acetyl-CoA carboxylase (ACC) phosphorylation decreased iodide uptake in a concentration-dependent manner, while the AMPK inhibitor compound C prevented this effect. In the thyroid gland of rats injected with AICAR, AMPK and ACC phosphorylation was increased and iodide uptake was reduced by ~35%. Under conditions of increased AMPK phosphorylation/activation such as TSH deprivation or AICAR treatment, significant reductions in cellular Na(+)/I(-)-symporter (NIS) protein (~41%) and mRNA content (~65%) were observed. The transcriptional (actinomycin D) and translational (cycloheximide) inhibitors, as well as the AMPK inhibitor compound C prevented AICAR-induced reduction of NIS protein content in PCCL3 cells. The presence of TSH in the culture medium reduced AMPK phosphorylation in PCCL3 cells, while inhibition of protein kinase A (PKA) with H89 prevented this effect. Conversely, the adenylyl cyclase activator forskolin abolished the AMPK phosphorylation response induced by TSH withdrawal in PCCL3 cells. These findings demonstrate that TSH suppresses AMPK phosphorylation/activation in a cAMP-PKA-dependent manner. In summary, we provide novel evidence that AMPK is involved in the physiological regulation of iodide uptake, which is an essential step for the formation of thyroid hormones as well as for the regulation of thyroid function.