1
|
Halbout M, Kopp PA. The Human Thyroid-Derived CI-huThyrEC Cell Line Expresses the Thyrotropin (TSH) Receptor and Thyroglobulin but Lacks Other Essential Characteristics of Thyroid Follicular Cells. Biomolecules 2025; 15:375. [PMID: 40149910 PMCID: PMC11940677 DOI: 10.3390/biom15030375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Thyroid hormone synthesis requires the normal function of thyroid follicular cells and adequate nutritional intake of iodine. For in vitro studies on thyroid cell pathophysiology, the immortalized FRTL5 rat thyroid cell line and a derivative thereof, the PCCL3 cell line, are widely used. However, a permanent human thyroid cell line is currently lacking. A recent report described a cell line obtained from human thyroid cells designated as Cl-huThyrEC. Methods: Four clones of Cl-huThyrEC cells were obtained and cultured in the presence of thyroid stimulating hormone (TSH). The expression of key genes defining the thyroid follicular cell phenotype was determined by reverse-transcription PCR (RT-PCR) in FRTL5, PCCL3, and Cl-huThyrEC cells. The latter were cultured as monolayers and as organoids in Matrigel. Iodide uptake was measured and compared among the cell lines. Results: Gene expression analysis reveals that Cl-huThyrEC cells express the thyroid-restricted transcription factors (PAX8, NKX2.1, FOXE1), the TSH receptor (TSHR), and thyroglobulin (TG), but they do not express the sodium-iodide symporter (NIS), thyroid peroxidase (TPO), and pendrin (SLC26A4). In functional studies, Cl-huThyrEC cells are unable to concentrate iodide. Conclusions: Despite the expression of certain key genes that are limited or restricted to thyroid follicular cells, Cl-huThyrEC cells lack some of the essential characteristics of thyroid follicular cells, in particular, NIS. Hence, their utility as a model system for thyroid follicular cells is limited.
Collapse
Affiliation(s)
- Mathias Halbout
- Division of Endocrinology, Diabetes and Metabolism, University Hospital of Lausanne, University of Lausanne, Hôtel des Patients, Avenue de la Sallaz 08, CH-1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Peter A. Kopp
- Division of Endocrinology, Diabetes and Metabolism, University Hospital of Lausanne, University of Lausanne, Hôtel des Patients, Avenue de la Sallaz 08, CH-1011 Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
2
|
Yamauchi I, Sugawa T, Hakata T, Yoshizawa A, Kita T, Kishimoto Y, Kimura S, Sakurai A, Kosugi D, Fujita H, Okamoto K, Ueda Y, Fujii T, Taura D, Sakane Y, Yasoda A, Inagaki N. Transcriptomic landscape of hyperthyroidism in mice overexpressing thyroid-stimulating hormone. iScience 2025; 28:111565. [PMID: 39811643 PMCID: PMC11730581 DOI: 10.1016/j.isci.2024.111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/06/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Activation of thyroid-stimulating hormone receptor (TSHR) fundamentally leads to hyperthyroidism. To elucidate TSHR signaling, we conducted transcriptome analyses for hyperthyroid mice that we generated by overexpressing TSH. TSH overexpression drastically changed their thyroid transcriptome. In particular, enrichment analyses identified the cell cycle, phosphatidylinositol 3-kinase/Akt pathway, and Ras-related protein 1 pathway as possibly associated with goiter development. Regarding hyperthyroidism, Slc26a4 was exclusively upregulated with TSH overexpression among genes crucial to thyroid hormone secretion. To verify its significance, we overexpressed TSH in Slc26a4 knockout mice. TSH overexpression caused hyperthyroidism in Slc26a4 knockout mice, equivalent to that in control mice. Thus, we did not observe significant changes in known genes and pathways involved in thyroid hormone secretion with TSH overexpression. Our datasets might include candidate genes that have not yet been identified as regulators of thyroid function. Our transcriptome datasets regarding hyperthyroidism can contribute to future research on TSHR signaling.
Collapse
Affiliation(s)
- Ichiro Yamauchi
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Taku Sugawa
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takuro Hakata
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akira Yoshizawa
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomoko Kita
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Sadahito Kimura
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Aya Sakurai
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Daisuke Kosugi
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Haruka Fujita
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kentaro Okamoto
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yohei Ueda
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshihito Fujii
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Daisuke Taura
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoriko Sakane
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
- Sugawa Clinic, Nakagyo-ku, Kyoto 604-8105, Japan
| | - Akihiro Yasoda
- Clinical Research Center, National Hospital Organization Kyoto Medical Center, Fushimi-ku, Kyoto 612-8555, Japan
| | - Nobuya Inagaki
- Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-kofukai, Kita-ku, Osaka 530-8480, Japan
| |
Collapse
|
3
|
Chen P, Yao Y, Tan H, Li J. Systemic treatments for radioiodine-refractory thyroid cancers. Front Endocrinol (Lausanne) 2024; 15:1346476. [PMID: 39473507 PMCID: PMC11518755 DOI: 10.3389/fendo.2024.1346476] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/27/2024] [Indexed: 03/17/2025] Open
Abstract
Differentiated thyroid cancers (DTCs) constitute the primary histological subtype within thyroid cancer. Due to DTCs' distinctive radioiodine (RAI) uptake mechanism, standard treatment involving surgery, with or without adjunctive therapy using RAI and levothyroxine inhibition, typically yields favorable prognoses for the majority of patients with DTCs. However, this favorable outcome does not extend to individuals with decreased RAI uptake, termed radioiodine-refractory thyroid cancers (RAI-RTCs). Recent research has revealed that the genetic mutations and gene rearrangements affecting sites such as RTKs, RAS, BRAF and TERTp lead to structural and functional abnormalities in encoded proteins. These abnormalities aberrantly activate signaling pathways like the mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-hydroxykinase (PI3K) signaling pathways, resulting in thyroid cells dedifferentiation, sodium/iodide symporter (NIS) dysfunction, and consequent the RAI-refractory nature of DTCs. Targeted therapy tailored to mutations presents a promising avenue for the treatment of RAI-RTCs. Lenvatinib and sorafenib, multi-kinase inhibitors, represent the standard first-line systemic treatment options, while cabozantinib is the standard second-line treatment option, for this purpose. Furthermore, ongoing clinical trials are exploring selective kinase inhibitors, immune checkpoint inhibitors, and combination therapies. Notably, numerous clinical trials have demonstrated that selective kinase inhibitors like BRAF, MEK and mTOR inhibitors can restore RAI uptake in tumor cells. However, further validation through multicenter, large-sample, double-blinded randomized controlled trials are essential. Enhanced treatment strategies and innovative therapies are expected to benefit a broader spectrum of patients as these advancements progress.
Collapse
Affiliation(s)
| | | | - Huiwen Tan
- Division of Endocrinology and Metabolism, West China Hospital of Sichuan
University, Chengdu, China
| | - Jianwei Li
- Division of Endocrinology and Metabolism, West China Hospital of Sichuan
University, Chengdu, China
| |
Collapse
|
4
|
Bernadett L, Alexandra K, Georgina F, Erika T, András S, Ilona P, Ferenc O, Orsolya D. No Correlation between PD-L1 and NIS Expression in Lymph Node Metastatic Papillary Thyroid Carcinoma. Diagnostics (Basel) 2024; 14:1858. [PMID: 39272644 PMCID: PMC11394040 DOI: 10.3390/diagnostics14171858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Approximately 90% of thyroid cancers are differentiated thyroid cancers (DTCs), originating from follicular epithelial cells. Out of these, 90% are papillary thyroid cancer (PTC), and 10% are follicular thyroid cancer (FTC). The standard care procedure for PTC includes surgery, followed by radioiodine (RAI) ablation and thyroid-stimulating hormone (TSH) suppressive therapy. Globally, treating radioiodine-refractory DTC poses a challenge. During malignant transformation, thyroid epithelial cells often lose their ability to absorb radioiodine due to impaired membrane targeting or lack of NIS (sodium/iodide symporter) expression. Recent reports show an increase in PD-L1 (programmed death ligand 1) expression in thyroid cancer cells during dedifferentiation. However, no research exists wherein NIS and PD-L1 expression are analyzed together in thyroid cancer. Therefore, we aimed to investigate and correlate PD-L1 and NIS expression within primary tumor samples of lymph node metastatic PTC. We analyzed the expression of hNIS (human sodium/iodide symporter) and PD-L1 in primary tumor samples from metastatic PTC patients using immunohistochemistry. Immunohistochemistry analysis of PD-L1 and NIS was conducted in 89 and 86 PTC cases, respectively. Any subcellular NIS localization was counted as a positive result. PD-L1 expression was absent in 25 tumors, while 58 tumors displayed PD-L1 expression in 1-50% of their cells; in 6 tumors, over 50% of the cells tested positive for PD-L1. NIS immunohistochemistry was performed for 86 primary papillary carcinomas, with 51 out of 86 tumors showcasing NIS expression. Only in seven cases was NIS localized in the plasma membrane; in most tumors, NIS was primarily found in the intracytoplasmic membrane compartments. In the case of PD-L1 staining, cells showing linear membrane positivity of any intensity were counted as positive. The evaluation of NIS immunostaining was simpler: cells showing staining of any intensity of cytoplasmic or membranous fashion were counted as positive. The number of NIS positive cells can be further divided into cytoplasmic and membrane positive compartments. There was no observed correlation between PD-L1 and NIS expression. We can speculate that the manipulation of the PD-1/PD-L1 axis using anti-PD-L1 or anti-PD-1 antibodies could reinstate the functional expression of NIS. However, based on our study, the only conclusion that can be drawn is that there is no correlation between the percentage of NIS- or PD-L1-expressing tumor cells in the primary tumor of lymph node metastatic PTC.
Collapse
Affiliation(s)
- Lévay Bernadett
- National Institute of Oncology, Multidisciplinary Head and Neck Cancer Center, 1124 Budapest, Hungary
| | - Kiss Alexandra
- National Institute of Oncology, Multidisciplinary Head and Neck Cancer Center, 1124 Budapest, Hungary
| | - Fröhlich Georgina
- National Institute of Oncology, Budapest, Center of Radiotherapy, 1124 Budapest, Hungary
| | - Tóth Erika
- National Institute of Oncology, Department of Molecular Pathology and Surgical Pathology Center, 1124 Budapest, Hungary
| | - Slezák András
- National Institute of Oncology, Department of Molecular Pathology and Surgical Pathology Center, 1124 Budapest, Hungary
| | - Péter Ilona
- National Institute of Oncology, Department of Molecular Pathology and Surgical Pathology Center, 1124 Budapest, Hungary
| | - Oberna Ferenc
- National Institute of Oncology, Multidisciplinary Head and Neck Cancer Center, 1124 Budapest, Hungary
| | - Dohán Orsolya
- Department of Internal Medicine and Clinical Oncology, Semmelweis University, 1124 Budapest, Hungary
| |
Collapse
|
5
|
Diba Lahmidi M, Le Noc M, Dali O, Kernanec PY, Merret PE, Jaulin C, Smagulova F. Sex-specific transgenerational effects on murine thyroid gland imposed by ancestral exposure to neonicotinoid thiacloprid. Sci Rep 2024; 14:13047. [PMID: 38844538 PMCID: PMC11156953 DOI: 10.1038/s41598-024-63986-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Neonicotinoids, a relatively new widely used class of insecticide is used in agriculture to control insect populations. We examined the capacity of ancestral exposure to the neonicotinoid thiacloprid (thia) to induce transgenerational effects on thyroid tissue. Pregnant outbred Swiss female mice were exposed to thia at embryonic days E6.5 to E15.5 using 0, 0.6, and 6 mg/kg/day doses. Thyroid paraffin sections were prepared for morphology analysis. We apply ELISA method to measure T4 and TSH levels, RT-qPCR for gene expression analysis, ChIP-qPCR techniques for sperm histone H3K4me3 analysis, and immunofluorescence microscopy and western blots for protein detection. We observed an alteration in the morphology of thyroids in both males and females in the F3 generation. We observed an increase in T4 hormone in F1 females and a significant T4 level decrease in F3 males. T4 changes in F1 females were associated with a TSH increase. We found that the amount of Iodothyronine Deiodinase 1 (DIO1) (an enzyme converting T4 to T3) was decreased in both F1 and F3 generations in female thyroids. GNAS protein which is important for thyroid function has increased in female thyroids. Gene expression analysis showed that the expression of genes encoding thyroid gland development, chromatin, biosynthesis and transport factors were affected in the thyroid gland in both sexes in F1 and F3. The analysis of sperm histone H3K4me3 showed that H3K4me3 occupancy at the Dio1 locus has decreased while Thyroglobulin (Tg) and Matrix Metallopeptidase 2 (Mmp2) genes have increased H3K4me3 occupancy in the sperm of F3 mice. Besides, DNA methylation analysis of our previously published datasets showed that, in the sperm of F1 and F3 thia-derived mice, several genes related to thyroid function show consistent alterations. Our data suggest that ancestral exposure to thiacloprid affects thyroid function not only in exposed but also in indirectly exposed F3 generation.
Collapse
Affiliation(s)
- Mariam Diba Lahmidi
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Morgane Le Noc
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Ouzna Dali
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Pierre-Yves Kernanec
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Pierre-Etienne Merret
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Christian Jaulin
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France
| | - Fatima Smagulova
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, 35000, Rennes, France.
- Irset-Inserm UMR 1085, 9 Avenue du Prof. Léon Bernard, 35000, Rennes, France.
| |
Collapse
|
6
|
Lévay B, Lantos A, Sinkovics I, Slezák A, Tóth E, Dohán O. The master role of polarized NIS expression in regulating iodine metabolism in the human body. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:256-261. [PMID: 36913678 PMCID: PMC10689030 DOI: 10.20945/2359-3997000000583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 08/05/2022] [Indexed: 03/15/2023]
Abstract
Objective The aim of this study was to investigate how polarized sodium iodide symporter (NIS) expression may regulate iodide metabolism in vivo. Materials and methods Polarized NIS expression was analyzed in tissues that accumulate iodide by the use of immunohistochemistry and polyclonal antibody against the C-terminal end of human NIS (hNIS). Results Iodide absorption in the human intestine occurs via NIS expressed in the apical membrane. Iodide is secreted into the lumen of the stomach and salivary glands via NIS expressed in the basolateral membrane and then circulates back from the small intestine to the bloodstream via NIS expressed in the apical membrane. Conclusion Polarized NIS expression in the human body regulates intestinal-bloodstream recirculation of iodide, perhaps prolonging the availability of iodide in the bloodstream. This leads to more efficient iodide trapping by the thyroid gland. Understanding the regulation and manipulating gastrointestinal iodide recirculation could increase radioiodine availability during theranostic NIS applications.
Collapse
Affiliation(s)
- Bernadett Lévay
- National Institute of Oncology, Multidisciplinary Head and Neck Cancer Center, Budapest, Hungary,
| | - András Lantos
- National Korányi Institute of Pulmonology, Department of Pathology, Budapest, Hungary
| | - István Sinkovics
- National Institute of Oncology, Department of Nuclear Medicine, Budapest, Hungary
| | - András Slezák
- National Institute of Oncology, Department of Molecular Pathology and Surgical Pathology Center, Budapest, Hungary
| | - Erika Tóth
- National Institute of Oncology, Department of Molecular Pathology and Surgical Pathology Center, Budapest, Hungary
| | - Orsolya Dohán
- Semmelweis University, Department of Internal Medicine and Clinical Oncology, Budapest, Hungary
| |
Collapse
|
7
|
Altmann H, Cui Y, Henrich M, Schaub S, Thiel C, Moritz A, Bauer R, Bauer N. 99m-Technetium-Pertechnetat- und 99m-Technetium-Sestamibi-Szintigrafie zur Darstellung von hypofunktionellem Schilddrüsengewebe und Staging bei einem Hund mit Schilddrüsenkarzinom. TIERÄRZTLICHE PRAXIS AUSGABE K: KLEINTIERE / HEIMTIERE 2022; 50:446-456. [DOI: 10.1055/a-1966-1860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
ZusammenfassungEine 10-jährige Mischlingshündin wurde aufgrund einer zervikalen Umfangsvermehrung zur Schilddrüsen-Szintigrafie vorgestellt.Aufgrund einer zusätzlichen Hypothyreose, die einen „kalten“ Knoten vermuten ließ, und zur Metastasensuche, erfolgte neben der 99m-Technetium-Pertechnetat (Tc-Pertechnetat)-Szintigrafie eine zweite Szintigrafie mit 99m-Technetium-Sestamibi (Tc-MIBI).Zwanzig Minuten nach intravenöser Gabe von 38 MBq Tc-Pertechnetat waren ein „heißer“ zervikaler und ein intrathorakaler Knoten mit einem „Uptake“ von 8,40 und 0,25 % sichtbar. In der zweiten Szintigrafie 20 Minuten nach intravenöser Gabe von 364 MBq Tc-MIBI und 70 Minuten nach der ersten Szintigrafie zeigten die Knoten nach Abzug der Pertechnetat-Aktivität und Zerfallskorrektur einen „Uptake“ von 0,99 und 0,03 %. Zudem stellten sich in der Schilddrüsenloge beide Schilddrüsenlappen mit einer geringen Tc-MIBI-Aufnahme dar. Das Verhältnis Tc-„Uptake“/Tc-MIBI-„Uptake“ betrug für beide Läsionen 8,48 bzw. 8,33.Nach Exstirpation des zervikalen Tumors ergab die histopathologische Untersuchung atrophisches Schilddrüsengewebe, das von einem expansiv wachsenden gut differenzierten follikulären Schilddrüsenkarzinom fast vollständig verdrängt wurde.Dieser Fallbericht beschreibt Durchführung, Nutzen und rechnerische Korrektur einer aufeinanderfolgenden Pertechnetat- und MIBI-Szintigrafie, die eine Darstellung von hypofunktionellem Schilddrüsengewebe ermöglichte. Die beiden heißen Knoten waren daher und aufgrund ihrer gleichen MIBI-Stoffwechselaktivität als dystopes Gewebe/Metastasen zu werten, wodurch die Hündin in das prognostisch ungünstigere WHO-Stadium IV klassifiziert werden musste. Anders als beim Menschen sollten beim Hund zur Vermeidung einer erneuten Narkose beide Szintigrafien kurz hintereinander durchgeführt werden. Eine qualitative/visuelle Beurteilung des MIBI-Szintigramms ist daher nicht sicher möglich, sodass eine quantitative Beurteilung über den „Uptake“ nach rechnerischer Korrektur der Pertechnetat-Aktivität erfolgen muss.
Collapse
Affiliation(s)
- Hannah Altmann
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Innere Medizin, klinische Laboratoriumsdiagnostik und klinische Pathophysiologie, Justus-Liebig-Universität Gießen
| | - Yi Cui
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Innere Medizin, klinische Laboratoriumsdiagnostik und klinische Pathophysiologie, Justus-Liebig-Universität Gießen
| | - Manfred Henrich
- Institut für Veterinär-Pathologie, Fachbereich Veterinärmedizin, Justus-Liebig-Universität Gießen
| | - Sebastian Schaub
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Chirurgie, Justus-Liebig-Universität Gießen
| | - Cetina Thiel
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Chirurgie, Justus-Liebig-Universität Gießen
| | - Andreas Moritz
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Innere Medizin, klinische Laboratoriumsdiagnostik und klinische Pathophysiologie, Justus-Liebig-Universität Gießen
| | | | - Natali Bauer
- Klinikum Veterinärmedizin, Klinik für Kleintiere, Innere Medizin, klinische Laboratoriumsdiagnostik und klinische Pathophysiologie, Justus-Liebig-Universität Gießen
| |
Collapse
|
8
|
Ravera S, Nicola JP, Salazar-De Simone G, Sigworth FJ, Karakas E, Amzel LM, Bianchet MA, Carrasco N. Structural insights into the mechanism of the sodium/iodide symporter. Nature 2022; 612:795-801. [PMID: 36517601 PMCID: PMC10501339 DOI: 10.1038/s41586-022-05530-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 11/04/2022] [Indexed: 12/15/2022]
Abstract
The sodium/iodide symporter (NIS) is the essential plasma membrane protein that mediates active iodide (I-) transport into the thyroid gland, the first step in the biosynthesis of the thyroid hormones-the master regulators of intermediary metabolism. NIS couples the inward translocation of I- against its electrochemical gradient to the inward transport of Na+ down its electrochemical gradient1,2. For nearly 50 years before its molecular identification3, NIS was the molecule at the centre of the single most effective internal radiation cancer therapy: radioiodide (131I-) treatment for thyroid cancer2. Mutations in NIS cause congenital hypothyroidism, which must be treated immediately after birth to prevent stunted growth and cognitive deficiency2. Here we report three structures of rat NIS, determined by single-particle cryo-electron microscopy: one with no substrates bound; one with two Na+ and one I- bound; and one with one Na+ and the oxyanion perrhenate bound. Structural analyses, functional characterization and computational studies show the substrate-binding sites and key residues for transport activity. Our results yield insights into how NIS selects, couples and translocates anions-thereby establishing a framework for understanding NIS function-and how it transports different substrates with different stoichiometries and releases substrates from its substrate-binding cavity into the cytosol.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Juan Pablo Nicola
- Department of Clinical Biochemistry, National University of Córdoba, Córdoba, Argentina
| | | | - Fred J Sigworth
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Erkan Karakas
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - L Mario Amzel
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mario A Bianchet
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nancy Carrasco
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
9
|
Transcription Factor CREB3L1 Regulates the Expression of the Sodium/Iodide Symporter (NIS) in Rat Thyroid Follicular Cells. Cells 2022; 11:cells11081314. [PMID: 35455992 PMCID: PMC9029047 DOI: 10.3390/cells11081314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023] Open
Abstract
The transcription factor CREB3L1 is expressed in a wide variety of tissues including cartilage, pancreas, and bone. It is located in the endoplasmic reticulum and upon stimulation is transported to the Golgi where is proteolytically cleaved. Then, the N-terminal domain translocates to the nucleus to activate gene expression. In thyroid follicular cells, CREB3L1 is a downstream effector of thyrotropin (TSH), promoting the expression of proteins of the secretory pathway along with an expansion of the Golgi volume. Here, we analyzed the role of CREB3L1 as a TSH-dependent transcriptional regulator of the expression of the sodium/iodide symporter (NIS), a major thyroid protein that mediates iodide uptake. We show that overexpression and inhibition of CREB3L1 induce an increase and decrease in the NIS protein and mRNA levels, respectively. This, in turn, impacts on NIS-mediated iodide uptake. Furthermore, CREB3L1 knockdown hampers the increase the TSH-induced NIS expression levels. Finally, the ability of CREB3L1 to regulate the promoter activity of the NIS-coding gene (Slc5a5) was confirmed. Taken together, our findings highlight the role of CREB3L1 in maintaining the homeostasis of thyroid follicular cells, regulating the adaptation of the secretory pathway as well as the synthesis of thyroid-specific proteins in response to TSH stimulation.
Collapse
|
10
|
Jhiang SM, Sipos JA. Na+/I- symporter expression, function, and regulation in non-thyroidal tissues and impact on thyroid cancer therapy. Endocr Relat Cancer 2021; 28:T167-T177. [PMID: 33974556 PMCID: PMC8419015 DOI: 10.1530/erc-21-0035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/20/2021] [Indexed: 11/08/2022]
Abstract
For the past 80 years, radioiodine (131I) has been used to ablate thyroid tissue not removed by surgery or to treat differentiated thyroid cancer that has metastasized to other parts of the body. However, the Na+/I- symporter (NIS), which mediates active iodide uptake into thyroid follicular cells, is also expressed in several non-thyroidal tissues. This NIS expression permits 131I accumulation and radiation damage in these non-target tissues, which accounts for the adverse effects of radioiodine therapy. We will review the data regarding the expression, function, and regulation of NIS in non-thyroidal tissues and explain the seemingly paradoxical adverse effects induced by 131I, the self-limited gastrointestinal adverse effects in contrast to the permanent salivary dysfunction that is seen after 131I therapy. We propose that prospective studies are needed to uncover the time-course of pathological processes underlying development and progression or ultimate resolution of 131I-induced salivary ductal obstruction and nasolacrimal duct obstruction. Finally, preventive measures and early therapeutic interventions that can be applied potentially to eliminate or alleviate long-term radioiodine adverse effects will be discussed.
Collapse
Affiliation(s)
- Sissy M Jhiang
- Department of Physiology and Cell Biology, The Ohio State University, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Jennifer A. Sipos
- Division of Endocrinology and Metabolism, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| |
Collapse
|
11
|
Albano F, Tucci V, Blackshear PJ, Reale C, Roberto L, Russo F, Marotta P, Porreca I, Colella M, Mallardo M, de Felice M, Ambrosino C. ZFP36L2 Role in Thyroid Functionality. Int J Mol Sci 2021; 22:9379. [PMID: 34502288 PMCID: PMC8431063 DOI: 10.3390/ijms22179379] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/30/2021] [Accepted: 08/25/2021] [Indexed: 11/17/2022] Open
Abstract
Thyroid hormone levels are usually genetically determined. Thyrocytes produce a unique set of enzymes that are dedicated to thyroid hormone synthesis. While thyroid transcriptional regulation is well-characterized, post-transcriptional mechanisms have been less investigated. Here, we describe the involvement of ZFP36L2, a protein that stimulates degradation of target mRNAs, in thyroid development and function, by in vivo and in vitro gene targeting in thyrocytes. Thyroid-specific Zfp36l2-/- females were hypothyroid, with reduced levels of circulating free Thyroxine (cfT4) and Triiodothyronine (cfT3). Their hypothyroidism was due to dyshormonogenesis, already evident one week after weaning, while thyroid development appeared normal. We observed decreases in several thyroid-specific transcripts and proteins, such as Nis and its transcriptional regulators (Pax8 and Nkx2.1), and increased apoptosis in Zfp36l2-/- thyroids. Nis, Pax8, and Nkx2.1 mRNAs were also reduced in Zfp36l2 knock-out thyrocytes in vitro (L2KO), in which we confirmed the increased apoptosis. Finally, in L2KO cells, we showed an altered response to TSH stimulation regarding both thyroid-specific gene expression and cell proliferation and survival. This result was supported by increases in P21/WAF1 and p-P38MAPK levels. Mechanistically, we confirmed Notch1 as a target of ZFP36L2 in the thyroid since its levels were increased in both in vitro and in vivo models. In both models, the levels of Id4 mRNA, a potential inhibitor of Pax8 activity, were increased. Overall, the data indicate that the regulation of mRNA stability by ZFP36L2 is a mechanism that controls the function and survival of thyrocytes.
Collapse
Affiliation(s)
- Francesco Albano
- IEOS-CNR, 80131 Naples, Italy;
- Biogem, Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, Italy; (V.T.); (C.R.); (L.R.); (F.R.); (I.P.); (M.C.)
| | - Valeria Tucci
- Biogem, Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, Italy; (V.T.); (C.R.); (L.R.); (F.R.); (I.P.); (M.C.)
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Perry J. Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
- Departments of Medicine and Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Carla Reale
- Biogem, Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, Italy; (V.T.); (C.R.); (L.R.); (F.R.); (I.P.); (M.C.)
| | - Luca Roberto
- Biogem, Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, Italy; (V.T.); (C.R.); (L.R.); (F.R.); (I.P.); (M.C.)
| | - Filomena Russo
- Biogem, Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, Italy; (V.T.); (C.R.); (L.R.); (F.R.); (I.P.); (M.C.)
| | - Pina Marotta
- Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy;
| | - Immacolata Porreca
- Biogem, Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, Italy; (V.T.); (C.R.); (L.R.); (F.R.); (I.P.); (M.C.)
| | - Marco Colella
- Biogem, Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, Italy; (V.T.); (C.R.); (L.R.); (F.R.); (I.P.); (M.C.)
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Massimo Mallardo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Mario de Felice
- IEOS-CNR, 80131 Naples, Italy;
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Concetta Ambrosino
- Biogem, Istituto di Biologia e Genetica Molecolare, 83031 Ariano Irpino, Italy; (V.T.); (C.R.); (L.R.); (F.R.); (I.P.); (M.C.)
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| |
Collapse
|
12
|
Martín M, Modenutti CP, Gil Rosas ML, Peyret V, Geysels RC, Bernal Barquero CE, Sobrero G, Muñoz L, Signorino M, Testa G, Miras MB, Masini-Repiso AM, Calcaterra NB, Coux G, Carrasco N, Martí MA, Nicola JP. A Novel SLC5A5 Variant Reveals the Crucial Role of Kinesin Light Chain 2 in Thyroid Hormonogenesis. J Clin Endocrinol Metab 2021; 106:1867-1881. [PMID: 33912899 PMCID: PMC8208674 DOI: 10.1210/clinem/dgab283] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Indexed: 12/17/2022]
Abstract
CONTEXT Iodide transport defect (ITD) (Online Mendelian Inheritance in Man No. 274400) is an uncommon cause of dyshormonogenic congenital hypothyroidism due to loss-of-function variants in the SLC5A5 gene, which encodes the sodium/iodide symporter (NIS), causing deficient iodide accumulation in thyroid follicular cells. OBJECTIVE This work aims to determine the molecular basis of a patient's ITD clinical phenotype. METHODS The propositus was diagnosed with dyshormonogenic congenital hypothyroidism with minimal 99mTc-pertechnetate accumulation in a eutopic thyroid gland. The propositus SLC5A5 gene was sequenced. Functional in vitro characterization of the novel NIS variant was performed. RESULTS Sanger sequencing revealed a novel homozygous missense p.G561E NIS variant. Mechanistically, the G561E substitution reduces iodide uptake, because targeting of G561E NIS to the plasma membrane is reduced. Biochemical analyses revealed that G561E impairs the recognition of an adjacent tryptophan-acidic motif by the kinesin-1 subunit kinesin light chain 2 (KLC2), interfering with NIS maturation beyond the endoplasmic reticulum, and reducing iodide accumulation. Structural bioinformatic analysis suggests that G561E shifts the equilibrium of the unstructured tryptophan-acidic motif toward a more structured conformation unrecognizable to KLC2. Consistently, knockdown of Klc2 causes defective NIS maturation and consequently decreases iodide accumulation in rat thyroid cells. Morpholino knockdown of klc2 reduces thyroid hormone synthesis in zebrafish larvae leading to a hypothyroid state as revealed by expression profiling of key genes related to the hypothalamic-pituitary-thyroid axis. CONCLUSION We report a novel NIS pathogenic variant associated with dyshormonogenic congenital hypothyroidism. Detailed molecular characterization of G561E NIS uncovered the significance of KLC2 in thyroid physiology.
Collapse
Affiliation(s)
- Mariano Martín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología–Consejo Nacional de Investigaciones Científicas y Técnicas, X5000HUA Córdoba, Argentina
| | - Carlos Pablo Modenutti
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales–Consejo Nacional de Investigaciones Científicas y Técnicas, C1428EGA Buenos Aires, Argentina
| | - Mauco Lucas Gil Rosas
- Departamento de Ciencias Biológicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, S2000EZP Rosario, Argentina
- Instituto de Biología Molecular y Celular de Rosario–Consejo Nacional de Investigaciones Científicas y Técnicas, S2000EZP Rosario, Argentina
| | - Victoria Peyret
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología–Consejo Nacional de Investigaciones Científicas y Técnicas, X5000HUA Córdoba, Argentina
| | - Romina Celeste Geysels
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología–Consejo Nacional de Investigaciones Científicas y Técnicas, X5000HUA Córdoba, Argentina
| | - Carlos Eduardo Bernal Barquero
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología–Consejo Nacional de Investigaciones Científicas y Técnicas, X5000HUA Córdoba, Argentina
| | - Gabriela Sobrero
- Programa Provincial de Pesquisa Neonatal, Hospital de Niños de la Santísima Trinidad de Córdoba, X5014AKK Córdoba, Argentina
| | - Liliana Muñoz
- Programa Provincial de Pesquisa Neonatal, Hospital de Niños de la Santísima Trinidad de Córdoba, X5014AKK Córdoba, Argentina
| | - Malvina Signorino
- Programa Provincial de Pesquisa Neonatal, Hospital de Niños de la Santísima Trinidad de Córdoba, X5014AKK Córdoba, Argentina
| | - Graciela Testa
- Programa Provincial de Pesquisa Neonatal, Hospital de Niños de la Santísima Trinidad de Córdoba, X5014AKK Córdoba, Argentina
| | - Mirta Beatriz Miras
- Programa Provincial de Pesquisa Neonatal, Hospital de Niños de la Santísima Trinidad de Córdoba, X5014AKK Córdoba, Argentina
| | - Ana María Masini-Repiso
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología–Consejo Nacional de Investigaciones Científicas y Técnicas, X5000HUA Córdoba, Argentina
| | - Nora Beatriz Calcaterra
- Departamento de Ciencias Biológicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, S2000EZP Rosario, Argentina
- Instituto de Biología Molecular y Celular de Rosario–Consejo Nacional de Investigaciones Científicas y Técnicas, S2000EZP Rosario, Argentina
| | - Gabriela Coux
- Departamento de Ciencias Biológicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, S2000EZP Rosario, Argentina
- Instituto de Biología Molecular y Celular de Rosario–Consejo Nacional de Investigaciones Científicas y Técnicas, S2000EZP Rosario, Argentina
| | - Nancy Carrasco
- Department of Cellular and Molecular Physiology, Yale School of Medicine, 06510 New Haven, Connecticut, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, 37232 Nashville, Tennessee, USA
| | - Marcelo Adrián Martí
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales–Consejo Nacional de Investigaciones Científicas y Técnicas, C1428EGA Buenos Aires, Argentina
| | - Juan Pablo Nicola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología–Consejo Nacional de Investigaciones Científicas y Técnicas, X5000HUA Córdoba, Argentina
| |
Collapse
|
13
|
Cai X, Wang R, Tan J, Meng Z, Li N. Mechanisms of regulating NIS transport to the cell membrane and redifferentiation therapy in thyroid cancer. Clin Transl Oncol 2021; 23:2403-2414. [PMID: 34100218 DOI: 10.1007/s12094-021-02655-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/28/2021] [Indexed: 11/29/2022]
Abstract
Iodine is an essential constituent of thyroid hormone. Active iodide accumulation in the thyroid is mediated by the sodium iodide symporter (NIS), comprising the first step in thyroid hormone biosynthesis, which relies on the functional expression of NIS on the cell membrane. The retention of NIS expressed in differentiated thyroid cancer (DTC) cells allows further treatment with post-operative radioactive iodine (RAI) therapy. However, compared with normal thyroid tissue, differentiated thyroid tumors usually show a decrease in the active iodide conveyance and NIS is generally retained within the cells, indicating that posttranslational protein transfer to the plasma membrane is abnormal. In recent years, through in vitro studies and studies of patients with DTC, various methods have been tested to increase the transport rate of NIS to the cell membrane and increase the absorption of iodine. An in-depth understanding of the mechanism of NIS transport to the plasma membrane could lead to improvements in RAI therapy. Therefore, in this review, we discuss the current knowledge concerning the post-translational mechanisms that regulate NIS transport to the cell membrane and the current status of redifferentiation therapy for patients with RAI-refractory (RAIR)-DTC.
Collapse
Affiliation(s)
- X Cai
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - R Wang
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - J Tan
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Z Meng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - N Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| |
Collapse
|
14
|
Geysels RC, Peyret V, Martín M, Nazar M, Reale C, Bernal Barquero CE, Miranda L, Martí MA, Vito P, Masini-Repiso AM, Nicola JP. The Transcription Factor NF-κB Mediates Thyrotropin-Stimulated Expression of Thyroid Differentiation Markers. Thyroid 2021; 31:299-314. [PMID: 32935630 DOI: 10.1089/thy.2020.0208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) transcription factor is a key regulator of cell survival, proliferation, and gene expression. Although activation of NF-κB signaling in thyroid follicular cells after thyrotropin (TSH) receptor (TSHR) engagement has been reported, the downstream signaling leading to NF-κB activation remains unexplored. Here, we sought to elucidate the mechanisms that regulate NF-κB signaling activation in response to TSH stimulation. Methods: Fisher rat-derived thyroid cell lines and primary cultures of NF-κB essential modulator (NEMO)-deficient mice thyrocytes were used as models. Signaling pathways leading to the activation of NF-κB were investigated by using chemical inhibitors and phospho-specific antibodies. Luciferase reporter gene assays and site-directed mutagenesis were used to monitor NF-κB-dependent gene transcriptional activity and the expression of thyroid differentiation markers was assessed by reverse transcription quantitative polymerase chain reaction and Western blot, respectively. Chromatin immunoprecipitation (ChIP) was carried out to investigate NF-κB subunit p65 DNA binding, and small interfering RNA (siRNA)-mediated gene knockdown approaches were used for studying gene function. Results: Using thyroid cell lines, we observed that TSH treatment leads to protein kinase C (PKC)-mediated canonical NF-κB p65 subunit nuclear expression. Moreover, TSH stimulation phosphorylated the kinase TAK-1, and its knockdown abolished TSH-induced NF-κB transcriptional activity. TSH induced the transcriptional activity of the NF-κB subunit p65 in a protein kinase A (PKA)-dependent phosphorylation at Ser-276. In addition, p65 phosphorylation at Ser-276 induced acetyl transferase p300 recruitment, leading to its acetylation on Lys-310 and thereby enhancing its transcriptional activity. Evaluation of the role played by NF-κB in thyroid physiology demonstrated that the canonical NF-κB inhibitor BAY 11-7082 reduced TSH-induced expression of thyroid differentiation markers. The involvement of NF-κB signaling in thyroid physiology was confirmed by assessing the TSH-induced gene expression in primary cultures of NEMO-deficient mice thyrocytes. ChIP and the knockdown experiments revealed that p65 is a nuclear effector of TSH actions, inducing the transcripcional expression of thyroid differentiation markers. Conclusions: Taken together, our results point to NF-κB being a pivotal mediator in the TSH-induced thyroid follicular cell differentiation, a relevant finding with potential physiological and pathophysiological implications.
Collapse
Affiliation(s)
- Romina Celeste Geysels
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Victoria Peyret
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Mariano Martín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Magalí Nazar
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Carla Reale
- Biogem Consortium, Ariano Irpino, Italy
- Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| | - Carlos Eduardo Bernal Barquero
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Lucas Miranda
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales-Consejo Nacional de Investigaciones Científicas y Técnicas (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Marcelo Adrián Martí
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales-Consejo Nacional de Investigaciones Científicas y Técnicas (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Pasquale Vito
- Biogem Consortium, Ariano Irpino, Italy
- Dipartimento di Scienze e Tecnologie, Università degli Studi del Sannio, Benevento, Italy
| | - Ana María Masini-Repiso
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| | - Juan Pablo Nicola
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Córdoba, Argentina
| |
Collapse
|
15
|
Ziros PG, Renaud CO, Chartoumpekis DV, Bongiovanni M, Habeos IG, Liao XH, Refetoff S, Kopp PA, Brix K, Sykiotis GP. Mice Hypomorphic for Keap1, a Negative Regulator of the Nrf2 Antioxidant Response, Show Age-Dependent Diffuse Goiter with Elevated Thyrotropin Levels. Thyroid 2021; 31:23-35. [PMID: 32689903 PMCID: PMC7840308 DOI: 10.1089/thy.2020.0044] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Familial nontoxic multinodular goiter (MNG) is a rare disease. One of the associated genes is Kelch-like ECH-associated protein 1 (KEAP1), which encodes the main inhibitor of nuclear factor erythroid 2-related transcription factor 2 (Nrf2), a central mediator of antioxidant responses. The association of KEAP1 with familial MNG is based on only two loss-of-function mutations identified in two families, only one of which included proper phenotyping and adequate demonstration of co-segregation of the phenotype and the mutation. There is no experimental evidence from model organisms to support that decreased Keap1 levels can, indeed, cause goiter. This study used mice hypomorphic for Keap1 to test whether decreased Keap1 expression can cause goiter, and to characterize the activation status of Nrf2 in their thyroid. Methods: C57BL/6J Keap1flox/flox (Keap1 knock-down [Keap1KD]) mice were studied at 3 and 12 months of age. Plasma and thyroid glands were harvested for evaluation of thyroid function tests and for gene and protein expression by real-time polymerase chain reaction and immunoblotting, respectively. Results: Keap1KD mice showed diffuse goiter that began to develop in early adult life and became highly prominent and penetrant with age. The goiter was characterized by a markedly increased size of thyroid follicles, most notably of the colloid compartment, and by absence of thyroid nodules or hyperplasia. Keap1KD mice also showed decreased T4 levels in early adult life that were eventually well compensated over time by increased thyrotropin (TSH) levels. Nrf2 was activated in the thyroid of Keap1KD mice. Despite a known stimulatory effect of Nrf2 on thyroglobulin (Tg) gene transcription and Tg protein abundance, the expression levels were decreased in the thyroid of Keap1KD mice. No clear patterns were observed in the expression profiles of other thyroid hormone synthesis-specific factors, with the exception of Tg-processing and Tg-degrading cathepsins, including an increase in mature forms of cathepsins D, L, and S. Conclusions: Keap1KD mice develop age-dependent diffuse goiter with elevated TSH levels. The precise mechanism accounting for the thyroidal phenotype remains to be elucidated, but it may involve enhanced Tg solubilization and excessive lysosomal Tg degradation.
Collapse
Affiliation(s)
- Panos G. Ziros
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Cédric O. Renaud
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Dionysios V. Chartoumpekis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, Patras, Greece
| | | | - Ioannis G. Habeos
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, Patras, Greece
| | - Xiao-Hui Liao
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Samuel Refetoff
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, and University of Chicago, Chicago, Illinois, USA
- Department of Committee on Genetics, University of Chicago, Chicago, Illinois, USA
| | - Peter A. Kopp
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Gerasimos P. Sykiotis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
16
|
Zhang CX, Zhang JX, Yang L, Zhang CR, Cheng F, Zhang RJ, Fang Y, Wang Z, Wu FY, Li PZ, Liang J, Li R, Song HD. Novel Compound Heterozygous Pathogenic Mutations of SLC5A5 in a Chinese Patient With Congenital Hypothyroidism. Front Endocrinol (Lausanne) 2021; 12:620117. [PMID: 33815280 PMCID: PMC8018529 DOI: 10.3389/fendo.2021.620117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/05/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Defects in the human sodium/iodide symporter (SLC5A5) gene have been reported to be one of the causes of congenital hypothyroidism (CH). We aimed to identify SLC5A5 mutations in Chinese patients with CH and to evaluate the function of the mutation. METHODS Two hundred and seventy-three patients with primary CH were screened for mutations in SLC5A5 using next-generation sequencing. We investigated the expression and cellular localization of the novel compound heterozygous mutation in SLC5A5. The functional activity of the mutants was further examined in vitro. RESULTS In 273 patients with CH, two previously undescribed pathogenic mutations p.Gly51AlafsTer45 (G51fs) and p.Gly421Arg (G421R) in a compound heterozygous state in SLC5A5 were identified in a pediatric patient. G51fs was located in the first intercellular loop connecting transmembrane segment I and II, whereas G421R was in the transmembrane segment (TMS) XI. G51fs and G421R resulted in a truncated NIS and reduced protein expression, respectively. In vitro experiments further showed that the normal function of iodine transport of sodium-iodide symporter (NIS) mutants was markedly impaired. CONCLUSION The undescribed compound heterozygous mutation of SLC5A5 was discovered in a Chinese CH patient. The mutation led to significantly reduced NIS expression and impaired iodide transport function accompanied by the impaired location of the NIS on the plasma membrane. Our study thus provides further insights into the roles of SLC5A5 in CH pathogenesis.
Collapse
Affiliation(s)
- Cao-Xu Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun-Xiu Zhang
- Department of Endocrinology, Maternal and Child Health Institute of Bozhou, Bozhou, China
| | - Liu Yang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chang-Run Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Cheng
- Department of Laboratory Medicine, Fujian Children’s Hospital, Fujian Provincial Maternity and Children’s Hospital, Fuzhou, China
| | - Rui-Jia Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Fang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng-Yao Wu
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei-Zhang Li
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Liang
- Department of Endocrinology, The Central Hospital of Xuzhou Affiliated to Xuzhou Medical College, Xuzhou, China
- *Correspondence: Huai-Dong Song, ; Rui Li, ; Jun Liang,
| | - Rui Li
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Huai-Dong Song, ; Rui Li, ; Jun Liang,
| | - Huai-Dong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Huai-Dong Song, ; Rui Li, ; Jun Liang,
| |
Collapse
|
17
|
Concilio SC, Suksanpaisan L, Pham L, Peng KW, Russell SJ. Improved Noninvasive In Vivo Tracking of AAV-9 Gene Therapy Using the Perchlorate-Resistant Sodium Iodide Symporter from Minke Whale. Mol Ther 2020; 29:236-243. [PMID: 33038323 PMCID: PMC7791078 DOI: 10.1016/j.ymthe.2020.09.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 01/12/2023] Open
Abstract
The sodium iodide symporter (NIS) is widely used as a reporter gene to noninvasively monitor the biodistribution and durability of vector-mediated gene expression via gamma scintigraphy, single-photon emission computed tomography (SPECT), and positron-emission tomography (PET). However, the approach is limited by background signal due to radiotracer uptake by endogenous NIS-expressing tissues. In this study, using the SPECT tracer pertechnetate (99mTcO4) and the PET tracer tetrafluoroborate (B18F4), in combination with the NIS inhibitor perchlorate, we compared the transport properties of human NIS and minke whale (Balaenoptera acutorostrata scammoni) NIS in vitro and in vivo. Based on its relative resistance to perchlorate, the NIS protein from minke whale appeared to be the superior candidate reporter gene. SPECT and PET imaging studies in nude mice challenged with NIS-encoding adeno-associated virus (AAV)-9 vectors confirmed that minke whale NIS, in contrast to human and endogenous mouse NIS, continues to function as a reliable reporter even when background radiotracer uptake by endogenous NIS is blocked by perchlorate.
Collapse
Affiliation(s)
- Susanna C Concilio
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Linh Pham
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Imanis Life Sciences, LLC, Rochester, MN 55901, USA
| | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA; Imanis Life Sciences, LLC, Rochester, MN 55901, USA.
| |
Collapse
|
18
|
Concilio SC, Zhekova HR, Noskov SY, Russell SJ. Inter-species variation in monovalent anion substrate selectivity and inhibitor sensitivity in the sodium iodide symporter (NIS). PLoS One 2020; 15:e0229085. [PMID: 32084174 PMCID: PMC7034854 DOI: 10.1371/journal.pone.0229085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
The sodium iodide symporter (NIS) transports iodide, which is necessary for thyroid hormone production. NIS also transports other monovalent anions such as tetrafluoroborate (BF4-), pertechnetate (TcO4-), and thiocyanate (SCN-), and is competitively inhibited by perchlorate (ClO4-). However, the mechanisms of substrate selectivity and inhibitor sensitivity are poorly understood. Here, a comparative approach was taken to determine whether naturally evolved NIS proteins exhibit variability in their substrate transport properties. The NIS proteins of thirteen animal species were initially assessed, and three species from environments with differing iodide availability, freshwater species Danio rerio (zebrafish), saltwater species Balaenoptera acutorostrata scammoni (minke whale), and non-aquatic mammalian species Homo sapiens (human) were studied in detail. NIS genes from each of these species were lentivirally transduced into HeLa cells, which were then characterized using radioisotope uptake assays, 125I- competitive substrate uptake assays, and kinetic assays. Homology models of human, minke whale and zebrafish NIS were used to evaluate sequence-dependent impact on the organization of Na+ and I- binding pockets. Whereas each of the three proteins that were analyzed in detail concentrated iodide to a similar degree, their sensitivity to perchlorate inhibition varied significantly: minke whale NIS was the least impacted by perchlorate inhibition (IC50 = 4.599 μM), zebrafish NIS was highly sensitive (IC50 = 0.081 μM), and human NIS showed intermediate sensitivity (IC50 = 1.566 μM). Further studies with fifteen additional substrates and inhibitors revealed similar patterns of iodide uptake inhibition, though the degree of 125I- uptake inhibition varied with each compound. Kinetic analysis revealed whale NIS had the lowest Km-I and the highest Vmax-I. Conversely, zebrafish NIS had the highest Km and lowest Vmax. Again, human NIS was intermediate. Molecular modeling revealed a high degree of conservation in the putative ion binding pockets of NIS proteins from different species, which suggests the residues responsible for the observed differences in substrate selectivity lie elsewhere in the protein. Ongoing studies are focusing on residues in the extracellular loops of NIS as determinants of anion specificity. These data demonstrate significant transport differences between the NIS proteins of different species, which may be influenced by the unique physiological needs of each organism. Our results also identify naturally-existing NIS proteins with significant variability in substrate transport kinetics and inhibitor sensitivity, which suggest that the affinity and selectivity of NIS for certain substrates can be altered for biotechnological and clinical applications. Further examination of interspecies differences may improve understanding of the substrate transport mechanism.
Collapse
Affiliation(s)
- Susanna C. Concilio
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Hristina R. Zhekova
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Sergei Y. Noskov
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
19
|
Proteomic Analysis of Iodinated Contrast Agent-Induced Perturbation of Thyroid Iodide Uptake. J Clin Med 2020; 9:jcm9020329. [PMID: 31979418 PMCID: PMC7073936 DOI: 10.3390/jcm9020329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 11/17/2022] Open
Abstract
(1) Background: We recently showed that iodinated contrast media (ICM) reduced thyroid uptake of iodide independently of free iodide through a mechanism different from that of NaI and involving a dramatic and long-lasting decrease in Na/I symporter expression. The present study aimed at comparing the response of the thyroid to ICM and NaI using a quantitative proteomic approach. (2) Methods: Scintiscans were performed on ICM-treated patients. Micro Single-Photon Emission Computed Tomography (microSPECT/CT) imaging was used to assess thyroid uptakes in ICM- or NaI-treated mice and their response to recombinant human thyroid-stimulating hormone. Total thyroid iodide content and proteome was determined in control, NaI-, or ICM-treated animals. (3) Results: The inhibitory effect of ICM in patients was selectively observed on thyroids but not on salivary glands for up to two months after a systemic administration. An elevated level of iodide was observed in thyroids from NaI-treated mice but not in those from ICM animals. Exposure of the thyroid to NaI modulates 15 cellular pathways, most of which are also affected by ICM treatment (including the elF4 and P706SK cell signaling pathway and INSR identified as an upstream activator in both treatments). In addition, ICM modulates 16 distinct pathways and failed to affect thyroid iodide content. Finally, administration of ICM reduces thyroid-stimulating hormone (TSH) receptor expression which results in a loss of TSH-induced iodide uptake by the thyroid. (4) Conclusions: Common intracellular mechanisms are involved in the ICM- and NaI-induced reduction of iodide uptake. However, ICM fails to affect thyroid iodide content which suggests that the modulation of these common pathways is triggered by separate effectors. ICM also modulates numerous distinct pathways which may account for its long-lasting effect on thyroid uptake. These observations may have implications in the management of patients affected by differentiated thyroid carcinomas who have been exposed to ICM. They also provide the basis for the utilization of ICM-based compounds in radioprotection of the thyroid.
Collapse
|
20
|
Rathod M, Chatterjee S, Dutta S, Kalraiya R, Bhattacharyya D, De A. Mannose glycosylation is an integral step for NIS localization and function in human breast cancer cells. J Cell Sci 2019; 132:jcs.232058. [PMID: 31455607 DOI: 10.1242/jcs.232058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022] Open
Abstract
Chasing an intriguing biological question on the disparity of sodium iodide symporter (NIS, officially known as SLC5A5) expression and function in the clinical scenario of breast cancer, this study addresses key molecular defects involved. NIS in cancer patients has primarily been recorded to be a cytoplasmic protein, thus limiting the scope for targeted radio-iodine therapy. We developed NIS transgene-overexpressing MCF-7 breast cancer cells, and found a few clonal derivatives that show predominant expression of NIS in the plasma membrane. The majority of clones, however, showed cytosolic NIS expression over long passages. Cells expressing membranous NIS show unperturbed dynamic trafficking of NIS through secretory pathway organelles when compared to cells expressing cytoplasmic NIS or to parental cells. Further, treatment of cells expressing membranous NIS with specific glycosylation inhibitors highlighted the importance of inherent glycosylation processing and an 84 gene signature glycosylation RT-Profiler array revealed that clones expressing NIS in their membrane cluster separately compared to the other cells. We further confirm a role of three differentially expressed genes, i.e. MAN1B1, MAN1A1 and MAN2A1, in regulating NIS localization by RNA interference. Thus, this study shows the important role of mannosidase in N-glycosylation processing in order to correctly traffic NIS to the plasma membrane in breast cancer cells.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Maitreyi Rathod
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Sushmita Chatterjee
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India
| | - Shruti Dutta
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India
| | - Rajiv Kalraiya
- Glycobiology Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Dibyendu Bhattacharyya
- Cell Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India.,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| | - Abhijit De
- Molecular Functional Imaging Laboratory, ACTREC, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India .,Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India
| |
Collapse
|
21
|
Thompson RJ, Fletcher A, Brookes K, Nieto H, Alshahrani MM, Mueller JW, Fine NH, Hodson DJ, Boelaert K, Read ML, Smith VE, McCabe CJ. Dimerization of the Sodium/Iodide Symporter. Thyroid 2019; 29:1485-1498. [PMID: 31310151 PMCID: PMC6797079 DOI: 10.1089/thy.2019.0034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background: The ability of thyroid follicular epithelial cells to accumulate iodide via the sodium/iodide symporter (NIS) is exploited to successfully treat most thyroid cancers, although a subset of patients lose functional NIS activity and become unresponsive to radioiodide therapy, with poor clinical outcome. Our knowledge of NIS regulation remains limited, however. While numerous membrane proteins are functionally regulated via dimerization, there is little definitive evidence of NIS dimerization, and whether this might impact upon radioiodide uptake and treatment success is entirely unknown. We hypothesized that NIS dimerizes and that dimerization is a prerequisite for iodide uptake. Methods: Coimmunoprecipitation, proximity ligation, and Förster resonance energy transfer (FRET) assays were used to assess NIS:NIS interaction. To identify residues involved in dimerization, a homology model of NIS structure was built based on the crystal structure of the dimeric bacterial protein vSGLT. Results: Abundant cellular NIS dimerization was confirmed in vitro via three discrete methodologies. FRET and proximity ligation assays demonstrated that while NIS can exist as a dimer at the plasma membrane (PM), it is also apparent in other cellular compartments. Homology modeling revealed one key potential site of dimeric interaction, with six residues <3Å apart. In particular, NIS residues Y242, T243, and Q471 were identified as critical to dimerization. Individual mutation of residues Y242 and T243 rendered NIS nonfunctional, while abrogation of Q471 did not impact radioiodide uptake. FRET data show that the putative dimerization interface can tolerate the loss of one, but not two, of these three clustered residues. Conclusions: We show for the first time that NIS dimerizes in vitro, and we identify the key residues via which this happens. We hypothesize that dimerization of NIS is critical to its trafficking to the PM and may therefore represent a new mechanism that would need to be considered in overcoming therapeutic failure in patients with thyroid cancer.
Collapse
Affiliation(s)
- Rebecca J. Thompson
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Alice Fletcher
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Katie Brookes
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Hannah Nieto
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Mohammed M. Alshahrani
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Jonathan W. Mueller
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Nicholas H.F. Fine
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - David J. Hodson
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Kristien Boelaert
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Martin L. Read
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Vicki E. Smith
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
- Vicki E. Smith, PhD, Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham Health Partners, Birmingham B15 2TT, United Kingdom
| | - Christopher J. McCabe
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
- Address correspondence to: Christopher J. McCabe, PhD, Institute of Metabolism and Systems Research, Centre for Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham Health Partners, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
22
|
Boutagy NE, Ravera S, Papademetris X, Onofrey JA, Zhuang ZW, Wu J, Feher A, Stacy MR, French BA, Annex BH, Carrasco N, Sinusas AJ. Noninvasive In Vivo Quantification of Adeno-Associated Virus Serotype 9-Mediated Expression of the Sodium/Iodide Symporter Under Hindlimb Ischemia and Neuraminidase Desialylation in Skeletal Muscle Using Single-Photon Emission Computed Tomography/Computed Tomography. Circ Cardiovasc Imaging 2019; 12:e009063. [PMID: 31296047 DOI: 10.1161/circimaging.119.009063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND We propose micro single-photon emission computed tomography/computed tomography imaging of the hNIS (human sodium/iodide symporter) to noninvasively quantify adeno-associated virus 9 (AAV9)-mediated gene expression in a murine model of peripheral artery disease. METHODS AAV9-hNIS (2×1011 viral genome particles) was injected into nonischemic or ischemic gastrocnemius muscles of C57Bl/6J mice following unilateral hindlimb ischemia ± the α-sialidase NA (neuraminidase). Control nonischemic limbs were injected with phosphate buffered saline or remained noninjected. Twelve mice underwent micro single-photon emission computed tomography/computed tomography imaging after serial injection of pertechnetate (99mTcO4-), a NIS substrate, up to 28 days after AAV9-hNIS injection. Twenty four animals were euthanized at selected times over 1 month for ex vivo validation. Forty-two animals were imaged with 99mTcO4- ± the selective NIS inhibitor perchlorate on day 10, to ascertain specificity of radiotracer uptake. Tissue was harvested for ex vivo validation. A modified version of the U-Net deep learning algorithm was used for image quantification. RESULTS As quantitated by standardized uptake value, there was a gradual temporal increase in 99mTcO4- uptake in muscles treated with AAV9-hNIS. Hindlimb ischemia, NA, and hindlimb ischemia plus NA increased the magnitude of 99mTcO4- uptake by 4- to 5-fold compared with nonischemic muscle treated with only AAV9-hNIS. Perchlorate treatment significantly reduced 99mTcO4- uptake in AAV9-hNIS-treated muscles, demonstrating uptake specificity. The imaging results correlated well with ex vivo well counting (r2=0.9375; P<0.0001) and immunoblot analysis of NIS protein (r2=0.65; P<0.0001). CONCLUSIONS Micro single-photon emission computed tomography/computed tomography imaging of hNIS-mediated 99mTcO4- uptake allows for accurate in vivo quantification of AAV9-driven gene expression, which increases under ischemic conditions or neuraminidase desialylation in skeletal muscle.
Collapse
Affiliation(s)
- Nabil E Boutagy
- Department of Medicine, Section of Cardiovascular Medicine, Yale Translational Research Imaging Center (N.E.B., Z.W.Z., A.F., M.R.S., A.J.S.), Yale School of Medicine, New Haven, CT
| | - Silvia Ravera
- Department of Cellular and Molecular Physiology (S.R., N.C.), Yale School of Medicine, New Haven, CT
| | - Xenophon Papademetris
- Department of Radiology and Biomedical Imaging (X.P., J.A.O., J.W., A.J.S.), Yale School of Medicine, New Haven, CT
| | - John A Onofrey
- Department of Radiology and Biomedical Imaging (X.P., J.A.O., J.W., A.J.S.), Yale School of Medicine, New Haven, CT
| | - Zhen W Zhuang
- Department of Medicine, Section of Cardiovascular Medicine, Yale Translational Research Imaging Center (N.E.B., Z.W.Z., A.F., M.R.S., A.J.S.), Yale School of Medicine, New Haven, CT
| | - Jing Wu
- Department of Radiology and Biomedical Imaging (X.P., J.A.O., J.W., A.J.S.), Yale School of Medicine, New Haven, CT
| | - Attila Feher
- Department of Medicine, Section of Cardiovascular Medicine, Yale Translational Research Imaging Center (N.E.B., Z.W.Z., A.F., M.R.S., A.J.S.), Yale School of Medicine, New Haven, CT
| | - Mitchel R Stacy
- Department of Medicine, Section of Cardiovascular Medicine, Yale Translational Research Imaging Center (N.E.B., Z.W.Z., A.F., M.R.S., A.J.S.), Yale School of Medicine, New Haven, CT
| | - Brent A French
- Department of Biomedical Engineering (B.A.F., B.H.A.), University of Virginia, Charlottesville
- Division of Cardiovascular Medicine, Department of Medicine (B.A.F., B.H.A.), University of Virginia, Charlottesville
| | - Brian H Annex
- Department of Biomedical Engineering (B.A.F., B.H.A.), University of Virginia, Charlottesville
- Division of Cardiovascular Medicine, Department of Medicine (B.A.F., B.H.A.), University of Virginia, Charlottesville
| | - Nancy Carrasco
- Department of Cellular and Molecular Physiology (S.R., N.C.), Yale School of Medicine, New Haven, CT
| | - Albert J Sinusas
- Department of Medicine, Section of Cardiovascular Medicine, Yale Translational Research Imaging Center (N.E.B., Z.W.Z., A.F., M.R.S., A.J.S.), Yale School of Medicine, New Haven, CT
- Department of Radiology and Biomedical Imaging (X.P., J.A.O., J.W., A.J.S.), Yale School of Medicine, New Haven, CT
| |
Collapse
|
23
|
Wen G, Fischer J, Most E, Eder K, Ringseis R. Decreased All- trans Retinoic Acid-Induced Expression of Sodium-Iodide Transporter in Mammary Epithelial Cells Caused by Conjugated Linoleic Acid Isomers. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:4493-4504. [PMID: 30938528 DOI: 10.1021/acs.jafc.9b00673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Expression of sodium-iodide symporter (NIS) is stimulated by sterol-regulatory-element-binding transcription factors (SREBFs) in mammary epithelial MCF-7 cells. Because conjugated linoleic acid (CLA) isomers have been shown to inhibit transcriptional activity of SREBFs in the mammary gland, the hypothesis was tested that CLA isomers inhibit NIS expression induced by all- trans retinoic acid (ATRA) in MCF-7 cells through inhibiting SREBF activity. c9t11-CLA and t10c12-CLA decreased ATRA-induced NIS-mRNA expression from 1.00 (ATRA alone) to 0.80 ± 0.12 (200 μM c9t11-CLA, P < 0.05) and 0.62 ± 0.10 (200 μM t10c12-CLA, P < 0.05), NIS-protein expression from 1.00 (ATRA alone) to 0.77 ± 0.08 (200 μM c9t11-CLA, P < 0.05) and 0.63 ± 0.05 (200 μM t10c12-CLA, P < 0.05), and NIS-promoter activity from 1.00 (ATRA alone) to 0.74 ± 0.13 (200 μM c9t11-CLA, P < 0.05) and 0.76 ± 0.13 (200 μM t10c12-CLA, P < 0.05); however, c9t11-CLA and t10c12-CLA increased the mRNA levels of SREBF isoforms and their target genes. In contrast, the mRNA expression of peroxisome-proliferator-activated receptor γ (PPARG) was strongly induced by ATRA alone but decreased by CLA isomers from 1.00 (ATRA alone) to 0.80 ± 0.06 (200 μM c9t11-CLA, P < 0.05) and 0.86 ± 0.06 (200 μM t10c12-CLA, P < 0.05). Overexpression of PPARγ in MCF-7 cells increased basal NIS-promoter activity, and treatment with the PPARγ ligand troglitazone stimulated ATRA-induced NIS-promoter activity. In conclusion, the results suggest that CLA isomers exert their effect on the expression of NIS by decreasing PPARG expression in MCF-7 cells.
Collapse
Affiliation(s)
- Gaiping Wen
- Institute of Animal Nutrition and Nutrition Physiology , Justus-Liebig-University Giessen , Heinrich-Buff-Ring 26-32 , 35392 Giessen , Germany
| | - Julia Fischer
- Institute of Animal Nutrition and Nutrition Physiology , Justus-Liebig-University Giessen , Heinrich-Buff-Ring 26-32 , 35392 Giessen , Germany
| | - Erika Most
- Institute of Animal Nutrition and Nutrition Physiology , Justus-Liebig-University Giessen , Heinrich-Buff-Ring 26-32 , 35392 Giessen , Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology , Justus-Liebig-University Giessen , Heinrich-Buff-Ring 26-32 , 35392 Giessen , Germany
| | - Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology , Justus-Liebig-University Giessen , Heinrich-Buff-Ring 26-32 , 35392 Giessen , Germany
| |
Collapse
|
24
|
Dong J, Cong Z, You M, Fu Y, Wang Y, Wang Y, Fu H, Wei L, Chen J. Effects of perinatal di (2-ethylhexyl) phthalate exposure on thyroid function in rat offspring. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 67:53-60. [PMID: 30716676 DOI: 10.1016/j.etap.2019.01.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/14/2018] [Accepted: 01/23/2019] [Indexed: 06/09/2023]
Abstract
Di (2-ethylhexyl) phthalate (DEHP) is a commonly used plasticizer in industry and displays the characteristics of an endocrine disruptor. Disorders of the maternal thyroid hormone (TH) during pregnancy can cause adverse effects on the fetus. We investigated the effects and possible mechanism of perinatal DEHP exposure on the thyroid function of pups. Pregnant female Wistar rats were randomly divided into four groups and received doses of DEHP of 0, 30, 300, 750 mg/kg/day by gavage at from gestational day (GD) 0 to postnatal day (PN) 21. The concentration of serum THs and the ultrastructure of thyroid follicular cells in the offspring were examined. Related protein level and gene expression of thyroid proteins in pups were analyzed by western blotting and real-time PCR. We found that DEHP significantly reduced total thyroxine (TT4) and increased thyroid stimulating hormone (TSH) in pups, while total triiodothyronine (TT3) showed no change. Thyroid follicular cells ultrastructure was damaged in DEHP exposed pups as viewed by electron microscopy. Furthermore, exposure to DEHP significantly increased protein and mRNA levels of thyroid transcription factor 1 (TTF-1), paired box 8 (PAX8), sodium iodide symporter (NIS) and thyroid peroxidase (TPO) in pups. In addition, levels of deiodinases of pups were also affected. These findings indicated that DEHP can disrupt thyroid function by damaging thyroid follicles and affecting TTF-1, PAX8, NIS, TPO and the deiodinase protein family.
Collapse
Affiliation(s)
- Jing Dong
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, People's Republic of China
| | - Zhangzhao Cong
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, People's Republic of China
| | - Mingdan You
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, People's Republic of China
| | - Yuanyuan Fu
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, People's Republic of China
| | - Yi Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, People's Republic of China
| | - Yuan Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, People's Republic of China
| | - Hui Fu
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, People's Republic of China
| | - Lingling Wei
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, People's Republic of China
| | - Jie Chen
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, People's Republic of China.
| |
Collapse
|
25
|
Waugh DT. Fluoride Exposure Induces Inhibition of Sodium/Iodide Symporter (NIS) Contributing to Impaired Iodine Absorption and Iodine Deficiency: Molecular Mechanisms of Inhibition and Implications for Public Health. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E1086. [PMID: 30917615 PMCID: PMC6466022 DOI: 10.3390/ijerph16061086] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 12/24/2022]
Abstract
The sodium iodide symporter (NIS) is the plasma membrane glycoprotein that mediates active iodide transport in the thyroid and other tissues, such as the salivary, gastric mucosa, rectal mucosa, bronchial mucosa, placenta and mammary glands. In the thyroid, NIS mediates the uptake and accumulation of iodine and its activity is crucial for the development of the central nervous system and disease prevention. Since the discovery of NIS in 1996, research has further shown that NIS functionality and iodine transport is dependent on the activity of the sodium potassium activated adenosine 5'-triphosphatase pump (Na+, K+-ATPase). In this article, I review the molecular mechanisms by which F inhibits NIS expression and functionality which in turn contributes to impaired iodide absorption, diminished iodide-concentrating ability and iodine deficiency disorders. I discuss how NIS expression and activity is inhibited by thyroglobulin (Tg), tumour necrosis factor alpha (TNF-α), transforming growth factor beta 1 (TGF-β1), interleukin 6 (IL-6) and Interleukin 1 beta (IL-1β), interferon-γ (IFN-γ), insulin like growth factor 1 (IGF-1) and phosphoinositide 3-kinase (PI3K) and how fluoride upregulates expression and activity of these biomarkers. I further describe the crucial role of prolactin and megalin in regulation of NIS expression and iodine homeostasis and the effect of fluoride in down regulating prolactin and megalin expression. Among many other issues, I discuss the potential conflict between public health policies such as water fluoridation and its contribution to iodine deficiency, neurodevelopmental and pathological disorders. Further studies are warranted to examine these associations.
Collapse
Affiliation(s)
- Declan Timothy Waugh
- EnviroManagement Services, 11 Riverview, Doherty's Rd, Bandon, Co. Cork, P72 YF10, Ireland.
| |
Collapse
|
26
|
Zhao Y, Zhong L, Yi H. A review on the mechanism of iodide metabolic dysfunction in differentiated thyroid cancer. Mol Cell Endocrinol 2019; 479:71-77. [PMID: 30287400 DOI: 10.1016/j.mce.2018.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 12/27/2022]
Abstract
The incidence of differentiated thyroid cancer (DTC) has been increasing rapidly worldwide, and the risk factors remain unclear. With the growing number of patients with DTC, the related issues have been gradually highlighted. 131Iodide (131I) is an important treatment for DTC and has the potential to reduce the risk of recurrence. 131I is also an effective treatment for distant metastases of thyroid carcinoma. However, iodide metabolism dysfunction in metastatic foci causes patients to lose the opportunity of 131I treatment. This article reviews the related mechanisms of iodide metabolism dysfunction in DTC cells and summarizes the clinical transformation progression.
Collapse
Affiliation(s)
- Yinlong Zhao
- Department of Nuclear Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, 130041, PR China.
| | - Lili Zhong
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, 130041, PR China.
| | - Heqing Yi
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang, 310021, PR China.
| |
Collapse
|
27
|
Ziros PG, Habeos IG, Chartoumpekis DV, Ntalampyra E, Somm E, Renaud CO, Bongiovanni M, Trougakos IP, Yamamoto M, Kensler TW, Santisteban P, Carrasco N, Ris-Stalpers C, Amendola E, Liao XH, Rossich L, Thomasz L, Juvenal GJ, Refetoff S, Sykiotis GP. NFE2-Related Transcription Factor 2 Coordinates Antioxidant Defense with Thyroglobulin Production and Iodination in the Thyroid Gland. Thyroid 2018; 28:780-798. [PMID: 29742982 PMCID: PMC5994681 DOI: 10.1089/thy.2018.0018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND The thyroid gland has a special relationship with oxidative stress. While generation of oxidative substances is part of normal iodide metabolism during thyroid hormone synthesis, the gland must also defend itself against excessive oxidation in order to maintain normal function. Antioxidant and detoxification enzymes aid thyroid cells to maintain homeostasis by ameliorating oxidative insults, including during exposure to excess iodide, but the factors that coordinate their expression with the cellular redox status are not known. The antioxidant response system comprising the ubiquitously expressed NFE2-related transcription factor 2 (Nrf2) and its redox-sensitive cytoplasmic inhibitor Kelch-like ECH-associated protein 1 (Keap1) defends tissues against oxidative stress, thereby protecting against pathologies that relate to DNA, protein, and/or lipid oxidative damage. Thus, it was hypothesized that Nrf2 should also have important roles in maintaining thyroid homeostasis. METHODS Ubiquitous and thyroid-specific male C57BL6J Nrf2 knockout (Nrf2-KO) mice were studied. Plasma and thyroids were harvested for evaluation of thyroid function tests by radioimmunoassays and of gene and protein expression by real-time polymerase chain reaction and immunoblotting, respectively. Nrf2-KO and Keap1-KO clones of the PCCL3 rat thyroid follicular cell line were generated using CRISPR/Cas9 technology and were used for gene and protein expression studies. Software-predicted Nrf2 binding sites on the thyroglobulin enhancer were validated by site-directed in vitro mutagenesis and chromatin immunoprecipitation. RESULTS The study shows that Nrf2 mediates antioxidant transcriptional responses in thyroid cells and protects the thyroid from oxidation induced by iodide overload. Surprisingly, it was also found that Nrf2 has a dramatic impact on both the basal abundance and the thyrotropin-inducible intrathyroidal abundance of thyroglobulin (Tg), the precursor protein of thyroid hormones. This effect is mediated by cell-autonomous regulation of Tg gene expression by Nrf2 via its direct binding to two evolutionarily conserved antioxidant response elements in an upstream enhancer. Yet, despite upregulating Tg levels, Nrf2 limits Tg iodination both under basal conditions and in response to excess iodide. CONCLUSIONS Nrf2 exerts pleiotropic roles in the thyroid gland to couple cell stress defense mechanisms to iodide metabolism and the thyroid hormone synthesis machinery, both under basal conditions and in response to excess iodide.
Collapse
Affiliation(s)
- Panos G. Ziros
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ioannis G. Habeos
- Department of Internal Medicine, Division of Endocrinology, School of Medicine, University of Patras, Patras, Greece
| | | | - Eleni Ntalampyra
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Emmanuel Somm
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Cédric O. Renaud
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Massimo Bongiovanni
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Thomas W. Kensler
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid, CIBERONC (ISCIII), Madrid, Spain
| | - Nancy Carrasco
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Carrie Ris-Stalpers
- Women's and Children's Clinic, Department of Obstetrics and Gynaecology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Elena Amendola
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli, Federico II, Naples, Italy
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Luciano Rossich
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - Lisa Thomasz
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - Guillermo J. Juvenal
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
- Department of Committee on Genetics, The University of Chicago, Chicago, Illinois
| | - Gerasimos P. Sykiotis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
28
|
Trøan G, Pihlava JM, Brandt-Kjelsen A, Salbu B, Prestløkken E. Heat-treated rapeseed expeller press cake with extremely low glucosinolate content reduce transfer of iodine to cow milk. Anim Feed Sci Technol 2018. [DOI: 10.1016/j.anifeedsci.2018.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
29
|
Yaglova NV, Sledneva YP, Nazimova SV, Obernikhin SS, Yaglov VV. Sex Differences in the Production of SLC5A5, Thyroid Peroxidase, and Thyroid Hormones in Pubertal Rats Exposed to Endocrine Disruptor Dichlorodiphenyltrichloroethane (DDT) during Postnatal Ontogeny. Bull Exp Biol Med 2018; 164:430-433. [PMID: 29500802 DOI: 10.1007/s10517-018-4005-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Indexed: 11/29/2022]
Abstract
Sex differences in the expression of iodide transporter SLC5A5 and thyroid peroxidase in thyroid follicular epithelium and thyroid serum profile were assessed in pubertal rats exposed to endocrine disruptor DDT starting from the first postnatal day. It was found that exposure to DDT reduced expression of SLC5A5 in peripheral regions of thyroid lobes in males and in central regions in females. The most pronounced sex differences were observed in thyroid peroxidase expression that remained sensitive to thyroid stimulating hormone regulation in males and lost sensitivity to pituitary stimulation in females after exposure to disruptor, which determines more pronounced hypothyroidism in females.
Collapse
Affiliation(s)
- N V Yaglova
- Research Institute of Human Morphology, Moscow, Russia.
| | - Yu P Sledneva
- Research Institute of Human Morphology, Moscow, Russia
| | - S V Nazimova
- Research Institute of Human Morphology, Moscow, Russia
| | | | - V V Yaglov
- Research Institute of Human Morphology, Moscow, Russia
| |
Collapse
|
30
|
A Simple Protocol for High Efficiency Protein Isolation After RNA Isolation from Mouse Thyroid and Other Very Small Tissue Samples. Methods Mol Biol 2018. [PMID: 27613051 DOI: 10.1007/978-1-4939-3756-1_25] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
As a dedicated hormone-secreting organ, the thyroid gland possesses a complement of proteostatic systems, including antioxidant, unfolded protein, and autophagic responses. The vast majority of animal investigations of thyroid physiology and, more recently, proteostasis, have utilized as model the rat, rather than the mouse. This is due to the very small size of the thyroid gland in the latter, with a total weight of ~2 mg (~1 mg per thyroid lobe). However, this strategy has limited the utilization of genetic approaches, such as taking advantage of the various transgenic and knockout mouse models. Here, we describe a simple and highly efficient protocol for the simultaneous isolation of mRNA, micro-RNA and 150-200 μg of protein from as little as 1 mg of mouse thyroid tissue, the average weight of one of the two thyroid lobes, thus preserving the other lobe for immunohistochemical or other analyses. While our workflow is similar to other protocols published in the literature and/or proposed by commercial reagent providers, we have introduced a key modification that addresses efficiently the most challenging step of the protein isolation process: the solubilization of the protein pellet after RNA extraction and protein precipitation. We demonstrate the feasibility of our approach and its utility for downstream analyses (including Western blotting) that facilitate the comparative study of proteostatic pathways in the mouse thyroid. We have also successfully applied this protocol on samples from mouse liver, brown and white adipose tissue, as well as from rodent cell lines.
Collapse
|
31
|
Carvalho DP, Dupuy C. Thyroid hormone biosynthesis and release. Mol Cell Endocrinol 2017; 458:6-15. [PMID: 28153798 DOI: 10.1016/j.mce.2017.01.038] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/07/2017] [Accepted: 01/23/2017] [Indexed: 12/31/2022]
Abstract
Thyroid hormones (TH) 3,5,3',5'- tetraiodothyronine or thyroxine (T4) and 3,5,3'- triiodothyronine (T3) contain iodine atoms as part of their structure, and their synthesis occur in the unique structures called thyroid follicles. Iodide reaches thyroid cells through the bloodstream that supplies the basolateral plasma membrane of thyrocytes, where it is avidly taken up through the sodium/iodide symporter (NIS). Thyrocytes are also specialized in the secretion of the high molecular weight protein thyroglobulin (TG) in the follicular lumen. The iodination of the tyrosyl residues of TG preceeds TH biosynthesis, which depends on the interaction of iodide, TG, hydrogen peroxide (H2O2) and thyroid peroxidase (TPO) at the apical plasma membrane of thyrocytes. Thyroid hormone biosynthesis is under the tonic control of thyrotropin (TSH), while the iodide recycling ability is very important for normal thyroid function. We discuss herein the biochemical aspects of TH biosynthesis and release, highlighting the novel molecules involved in the process.
Collapse
Affiliation(s)
- Denise P Carvalho
- Biophysics Institute of Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Corinne Dupuy
- Université Paris-Saclay, Orsay, France; UMR 8200 CNRS, Villejuif, France; Institut de Cancérologie Gustave Roussy, Villejuif, Ile-de-France, France
| |
Collapse
|
32
|
García IA, Torres Demichelis V, Viale DL, Di Giusto P, Ezhova Y, Polishchuk RS, Sampieri L, Martinez H, Sztul E, Alvarez C. CREB3L1-mediated functional and structural adaptation of the secretory pathway in hormone-stimulated thyroid cells. J Cell Sci 2017; 130:4155-4167. [PMID: 29093023 DOI: 10.1242/jcs.211102] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/11/2017] [Indexed: 01/22/2023] Open
Abstract
Many secretory cells increase the synthesis and secretion of cargo proteins in response to specific stimuli. How cells couple increased cargo load with a coordinate rise in secretory capacity to ensure efficient transport is not well understood. We used thyroid cells stimulated with thyrotropin (TSH) to demonstrate a coordinate increase in the production of thyroid-specific cargo proteins and ER-Golgi transport factors, and a parallel expansion of the Golgi complex. TSH also increased expression of the CREB3L1 transcription factor, which alone caused amplified transport factor levels and Golgi enlargement. Furthermore, CREB3L1 potentiated the TSH-induced increase in Golgi volume. A dominant-negative CREB3L1 construct hampered the ability of TSH to induce Golgi expansion, implying that this transcription factor contributes to Golgi expansion. Our findings support a model in which CREB3L1 acts as a downstream effector of TSH to regulate the expression of cargo proteins, and simultaneously increases the synthesis of transport factors and the expansion of the Golgi to synchronize the rise in cargo load with the amplified capacity of the secretory pathway.
Collapse
Affiliation(s)
- Iris A García
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Vanina Torres Demichelis
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Diego L Viale
- Laboratorio de Neuro y Citogenetica Molecular, Centro de Estudios en Salud y Medio Ambiente, Escuela de Ciencia y Tecnologi-Universidad Nacional de San Martiń-CONICET, Buenos Aires, B1650 WAB, Argentina
| | - Pablo Di Giusto
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Yulia Ezhova
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei, 34, 80078, Pozzuoli (NA), Italy
| | - Roman S Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei, 34, 80078, Pozzuoli (NA), Italy
| | - Luciana Sampieri
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Hernán Martinez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Elizabeth Sztul
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233-2008, USA
| | - Cecilia Alvarez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| |
Collapse
|
33
|
Kang HS, Kumar D, Liao G, Lichti-Kaiser K, Gerrish K, Liao XH, Refetoff S, Jothi R, Jetten AM. GLIS3 is indispensable for TSH/TSHR-dependent thyroid hormone biosynthesis and follicular cell proliferation. J Clin Invest 2017; 127:4326-4337. [PMID: 29083325 DOI: 10.1172/jci94417] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/12/2017] [Indexed: 12/12/2022] Open
Abstract
Deficiency in Krüppel-like zinc finger transcription factor GLI-similar 3 (GLIS3) in humans is associated with the development of congenital hypothyroidism. However, the functions of GLIS3 in the thyroid gland and the mechanism by which GLIS3 dysfunction causes hypothyroidism are unknown. In the current study, we demonstrate that GLIS3 acts downstream of thyroid-stimulating hormone (TSH) and TSH receptor (TSHR) and is indispensable for TSH/TSHR-mediated proliferation of thyroid follicular cells and biosynthesis of thyroid hormone. Using ChIP-Seq and promoter analysis, we demonstrate that GLIS3 is critical for the transcriptional activation of several genes required for thyroid hormone biosynthesis, including the iodide transporters Nis and Pds, both of which showed enhanced GLIS3 binding at their promoters. The repression of cell proliferation of GLIS3-deficient thyroid follicular cells was due to the inhibition of TSH-mediated activation of the mTOR complex 1/ribosomal protein S6 (mTORC1/RPS6) pathway as well as the reduced expression of several cell division-related genes regulated directly by GLIS3. Consequently, GLIS3 deficiency in a murine model prevented the development of goiter as well as the induction of inflammatory and fibrotic genes during chronic elevation of circulating TSH. Our study identifies GLIS3 as a key regulator of TSH/TSHR-mediated thyroid hormone biosynthesis and proliferation of thyroid follicular cells and uncovers a mechanism by which GLIS3 deficiency causes neonatal hypothyroidism and prevents goiter development.
Collapse
Affiliation(s)
| | | | - Grace Liao
- 1, Immunity, Inflammation and Disease Laboratory
| | | | - Kevin Gerrish
- 3, Molecular Genomics Core, National Institute of Environmental Health Sciences (NIEHS), NIH, Research Triangle Park, North Carolina, USA
| | | | - Samuel Refetoff
- 4, Department of Medicine, and.,5, Department of Pediatrics and Committee on Genetics, The University of Chicago, Chicago, Illinois, USA
| | - Raja Jothi
- 2, Epigenetics and Stem Cell Biology Laboratory, and
| | | |
Collapse
|
34
|
Ravera S, Reyna-Neyra A, Ferrandino G, Amzel LM, Carrasco N. The Sodium/Iodide Symporter (NIS): Molecular Physiology and Preclinical and Clinical Applications. Annu Rev Physiol 2017; 79:261-289. [PMID: 28192058 DOI: 10.1146/annurev-physiol-022516-034125] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Active iodide (I-) transport in both the thyroid and some extrathyroidal tissues is mediated by the Na+/I- symporter (NIS). In the thyroid, NIS-mediated I- uptake plays a pivotal role in thyroid hormone (TH) biosynthesis. THs are key during embryonic and postembryonic development and critical for cell metabolism at all stages of life. The molecular characterization of NIS in 1996 and the use of radioactive I- isotopes have led to significant advances in the diagnosis and treatment of thyroid cancer and provide the molecular basis for studies aimed at extending the use of radioiodide treatment in extrathyroidal malignancies. This review focuses on the most recent findings on I- homeostasis and I- transport deficiency-causing NIS mutations, as well as current knowledge of the structure/function properties of NIS and NIS regulatory mechanisms. We also discuss employing NIS as a reporter gene using viral vectors and stem cells in imaging, diagnostic, and therapeutic procedures.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut 06510;
| | - Andrea Reyna-Neyra
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut 06510;
| | - Giuseppe Ferrandino
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut 06510;
| | - L Mario Amzel
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Nancy Carrasco
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut 06510;
| |
Collapse
|
35
|
An extremely high dietary iodide supply forestalls severe hypothyroidism in Na +/I - symporter (NIS) knockout mice. Sci Rep 2017; 7:5329. [PMID: 28706256 PMCID: PMC5509730 DOI: 10.1038/s41598-017-04326-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 05/12/2017] [Indexed: 12/27/2022] Open
Abstract
The sodium/iodide symporter (NIS) mediates active iodide (I−) accumulation in the thyroid, the first step in thyroid hormone (TH) biosynthesis. Mutations in the SLC5A5 gene encoding NIS that result in a non-functional protein lead to congenital hypothyroidism due to I− transport defect (ITD). ITD is a rare autosomal disorder that, if not treated promptly in infancy, can cause mental retardation, as the TH decrease results in improper development of the nervous system. However, in some patients, hypothyroidism has been ameliorated by unusually large amounts of dietary I−. Here we report the first NIS knockout (KO) mouse model, obtained by targeting exons 6 and 7 of the Slc5a5 gene. In NIS KO mice, in the thyroid, stomach, and salivary gland, NIS is absent, and hence there is no active accumulation of the NIS substrate pertechnetate (99mTcO4−). NIS KO mice showed undetectable serum T4 and very low serum T3 levels when fed a diet supplying the minimum I− requirement for rodents. These hypothyroid mice displayed oxidative stress in the thyroid, but not in the brown adipose tissue or liver. Feeding the mice a high-I− diet partially rescued TH biosynthesis, demonstrating that, at high I− concentrations, I− enters the thyroid through routes other than NIS.
Collapse
|
36
|
Alotaibi H, Tuzlakoğlu-Öztürk M, Tazebay UH. The Thyroid Na+/I- Symporter: Molecular Characterization and Genomic Regulation. Mol Imaging Radionucl Ther 2017; 26:92-101. [PMID: 28117294 PMCID: PMC5283716 DOI: 10.4274/2017.26.suppl.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Iodide (I-) is an essential constituent of the thyroid hormones triiodothyronine (T3) and thyroxine (T4), and the iodide concentrating mechanism of the thyroid gland is essential for the synthesis of these hormones. In addition, differential uptake of iodine isotopes (radioiodine) is a key modality for the diagnosis and therapy of thyroid cancer. The sodium dependent iodide transport activity of the thyroid gland is mainly attributed to the functional expression of the Na+/I- Symporter (NIS) localized at the basolateral membrane of thyrocytes. In this paper, we review and summarize current data on molecular characterization, on structure and function of NIS protein, as well as on the transcriptional regulation of NIS encoding gene in the thyroid gland. We also propose that a better and more precise understanding of NIS gene regulation at the molecular level in both healthy and malignant thyroid cells may lead to the identification of small molecule candidates. These could then be translated into clinical practice for better induction and more effective modulation of radioiodine uptake in dedifferentiated thyroid cancer cells and in their distant metastatic lesions.
Collapse
Affiliation(s)
| | | | - Uygar Halis Tazebay
- Gebze Technical University, Department of Molecular Biology and Genetics, Kocaeli, Turkey, Phone: +90 262 605 25 22, E-mail:
| |
Collapse
|
37
|
Niwa O, Barcellos-Hoff MH, Globus RK, Harrison JD, Hendry JH, Jacob P, Martin MT, Seed TM, Shay JW, Story MD, Suzuki K, Yamashita S. ICRP Publication 131: Stem Cell Biology with Respect to Carcinogenesis Aspects of Radiological Protection. Ann ICRP 2016; 44:7-357. [PMID: 26637346 DOI: 10.1177/0146645315595585] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This report provides a review of stem cells/progenitor cells and their responses to ionising radiation in relation to issues relevant to stochastic effects of radiation that form a major part of the International Commission on Radiological Protection's system of radiological protection. Current information on stem cell characteristics, maintenance and renewal, evolution with age, location in stem cell 'niches', and radiosensitivity to acute and protracted exposures is presented in a series of substantial reviews as annexes concerning haematopoietic tissue, mammary gland, thyroid, digestive tract, lung, skin, and bone. This foundation of knowledge of stem cells is used in the main text of the report to provide a biological insight into issues such as the linear-no-threshold (LNT) model, cancer risk among tissues, dose-rate effects, and changes in the risk of radiation carcinogenesis by age at exposure and attained age. Knowledge of the biology and associated radiation biology of stem cells and progenitor cells is more developed in tissues that renew fairly rapidly, such as haematopoietic tissue, intestinal mucosa, and epidermis, although all the tissues considered here possess stem cell populations. Important features of stem cell maintenance, renewal, and response are the microenvironmental signals operating in the niche residence, for which a well-defined spatial location has been identified in some tissues. The identity of the target cell for carcinogenesis continues to point to the more primitive stem cell population that is mostly quiescent, and hence able to accumulate the protracted sequence of mutations necessary to result in malignancy. In addition, there is some potential for daughter progenitor cells to be target cells in particular cases, such as in haematopoietic tissue and in skin. Several biological processes could contribute to protecting stem cells from mutation accumulation: (a) accurate DNA repair; (b) rapidly induced death of injured stem cells; (c) retention of the DNA parental template strand during divisions in some tissue systems, so that mutations are passed to the daughter differentiating cells and not retained in the parental cell; and (d) stem cell competition, whereby undamaged stem cells outcompete damaged stem cells for residence in the niche. DNA repair mainly occurs within a few days of irradiation, while stem cell competition requires weeks or many months depending on the tissue type. The aforementioned processes may contribute to the differences in carcinogenic radiation risk values between tissues, and may help to explain why a rapidly replicating tissue such as small intestine is less prone to such risk. The processes also provide a mechanistic insight relevant to the LNT model, and the relative and absolute risk models. The radiobiological knowledge also provides a scientific insight into discussions of the dose and dose-rate effectiveness factor currently used in radiological protection guidelines. In addition, the biological information contributes potential reasons for the age-dependent sensitivity to radiation carcinogenesis, including the effects of in-utero exposure.
Collapse
|
38
|
Wen G, Pachner LI, Gessner DK, Eder K, Ringseis R. Sterol regulatory element-binding proteins are regulators of the sodium/iodide symporter in mammary epithelial cells. J Dairy Sci 2016; 99:9211-9226. [PMID: 27614840 DOI: 10.3168/jds.2016-11174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 07/17/2016] [Indexed: 12/29/2022]
Abstract
The sodium/iodide symporter (NIS), which is essential for iodide concentration in the thyroid, is reported to be transcriptionally regulated by sterol regulatory element-binding proteins (SREBP) in rat FRTL-5 thyrocytes. The SREBP are strongly activated after parturition and throughout lactation in the mammary gland of cattle and are important for mammary epithelial cell synthesis of milk lipids. In this study, we tested the hypothesis that the NIS gene is regulated also by SREBP in mammary epithelial cells, in which NIS is functionally expressed during lactation. Regulation of NIS expression and iodide uptake was investigated by means of inhibition, silencing, and overexpression of SREBP and by reporter gene and DNA-binding assays. As a mammary epithelial cell model, the human MCF-7 cell line, a breast adenocarcinoma cell line, which shows inducible expression of NIS by all-trans retinoic acid (ATRA), and unlike bovine mammary epithelial cells, is widely used to investigate the regulation of mammary gland NIS and NIS-specific iodide uptake, was used. Inhibition of SREBP maturation by treatment with 25-hydroxycholesterol (5 µM) for 48h reduced ATRA (1 µM)-induced mRNA concentration of NIS and iodide uptake in MCF-7 cells by approximately 20%. Knockdown of SREBP-1c and SREBP-2 by RNA interference decreased the mRNA and protein concentration of NIS by 30 to 50% 48h after initiating knockdown, whereas overexpression of nuclear SREBP (nSREBP)-1c and nSREBP-2 increased the expression of NIS in MCF-7 cells by 45 to 60%, respectively, 48h after initiating overexpression. Reporter gene experiments with varying length of NIS promoter reporter constructs revealed that the NIS 5'-flanking region is activated by nSREBP-1c and nSREBP-2 approximately 1.5- and 4.5-fold, respectively, and activation involves a SREBP-binding motif (SRE) at -38 relative to the transcription start site of the NIS gene. Gel shift assays using oligonucleotides spanning either the wild-type or the mutated SRE at -38 of the NIS 5'-flanking region showed that in vitro-translated nSREBP-1c and nSREBP-2 bind only the wild-type but not the mutated SRE at -38 of NIS. Collectively, the present results from cell culture experiments with human mammary epithelial MCF-7 cells and from genetic studies show for the first time that the NIS gene and iodide uptake are regulated by SREBP in cultured human mammary epithelial cells. Future studies are necessary to clarify if the regulation of NIS expression and iodide uptake by SREBP also applies to the lactating bovine mammary epithelium.
Collapse
Affiliation(s)
- G Wen
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring 26-32, 35392 Gießen, Germany
| | - L I Pachner
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring 26-32, 35392 Gießen, Germany
| | - D K Gessner
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring 26-32, 35392 Gießen, Germany
| | - K Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring 26-32, 35392 Gießen, Germany
| | - R Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Heinrich-Buff-Ring 26-32, 35392 Gießen, Germany.
| |
Collapse
|
39
|
Abstract
The sodium/iodide symporter (NIS) mediates active I(-) transport in the thyroid-the first step in thyroid hormone biosynthesis-with a 2 Na(+): 1 I(-) stoichiometry. The two Na(+) binding sites (Na1 and Na2) and the I(-) binding site interact allosterically: when Na(+) binds to a Na(+) site, the affinity of NIS for the other Na(+) and for I(-) increases significantly. In all Na(+)-dependent transporters with the same fold as NIS, the side chains of two residues, S353 and T354 (NIS numbering), were identified as the Na(+) ligands at Na2. To understand the cooperativity between the substrates, we investigated the coordination at the Na2 site. We determined that four other residues-S66, D191, Q194, and Q263-are also involved in Na(+) coordination at this site. Experiments in whole cells demonstrated that these four residues participate in transport by NIS: mutations at these positions result in proteins that, although expressed at the plasma membrane, transport little or no I(-) These residues are conserved throughout the entire SLC5 family, to which NIS belongs, suggesting that they serve a similar function in the other transporters. Our findings also suggest that the increase in affinity that each site displays when an ion binds to another site may result from changes in the dynamics of the transporter. These mechanistic insights deepen our understanding not only of NIS but also of other transporters, including many that, like NIS, are of great medical relevance.
Collapse
|
40
|
Ortiga-Carvalho TM, Chiamolera MI, Pazos-Moura CC, Wondisford FE. Hypothalamus-Pituitary-Thyroid Axis. Compr Physiol 2016; 6:1387-428. [PMID: 27347897 DOI: 10.1002/cphy.c150027] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The hypothalamus-pituitary-thyroid (HPT) axis determines the set point of thyroid hormone (TH) production. Hypothalamic thyrotropin-releasing hormone (TRH) stimulates the synthesis and secretion of pituitary thyrotropin (thyroid-stimulating hormone, TSH), which acts at the thyroid to stimulate all steps of TH biosynthesis and secretion. The THs thyroxine (T4) and triiodothyronine (T3) control the secretion of TRH and TSH by negative feedback to maintain physiological levels of the main hormones of the HPT axis. Reduction of circulating TH levels due to primary thyroid failure results in increased TRH and TSH production, whereas the opposite occurs when circulating THs are in excess. Other neural, humoral, and local factors modulate the HPT axis and, in specific situations, determine alterations in the physiological function of the axis. The roles of THs are vital to nervous system development, linear growth, energetic metabolism, and thermogenesis. THs also regulate the hepatic metabolism of nutrients, fluid balance and the cardiovascular system. In cells, TH actions are mediated mainly by nuclear TH receptors (210), which modify gene expression. T3 is the preferred ligand of THR, whereas T4, the serum concentration of which is 100-fold higher than that of T3, undergoes extra-thyroidal conversion to T3. This conversion is catalyzed by 5'-deiodinases (D1 and D2), which are TH-activating enzymes. T4 can also be inactivated by conversion to reverse T3, which has very low affinity for THR, by 5-deiodinase (D3). The regulation of deiodinases, particularly D2, and TH transporters at the cell membrane control T3 availability, which is fundamental for TH action. © 2016 American Physiological Society. Compr Physiol 6:1387-1428, 2016.
Collapse
Affiliation(s)
- Tania M Ortiga-Carvalho
- Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Maria I Chiamolera
- Department of Medicine, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Carmen C Pazos-Moura
- Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Fredic E Wondisford
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
41
|
Leoni SG, Sastre-Perona A, De la Vieja A, Santisteban P. Selenium Increases Thyroid-Stimulating Hormone-Induced Sodium/Iodide Symporter Expression Through Thioredoxin/Apurinic/Apyrimidinic Endonuclease 1-Dependent Regulation of Paired Box 8 Binding Activity. Antioxid Redox Signal 2016; 24:855-66. [PMID: 26650895 DOI: 10.1089/ars.2014.6228] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
AIMS The sodium-iodide symporter (NIS) mediates the uptake of I(-) by the thyroid follicular cell and is essential for thyroid hormone biosynthesis. Nis expression is stimulated by thyroid-stimulating hormone (TSH) and also requires paired box 8 (Pax8) to bind to its promoter. Pax8 binding activity depends on its redox state by a mechanism involving thioredoxin/thioredoxin reductase-1 (Txn/TxnRd1) reduction of apurinic/apyrimidinic endonuclease 1 (Ape1). In this study, we investigate the role of Se in Nis expression. RESULTS Selenium increases TSH-induced Nis expression and activity in rat thyroid cells. The stimulatory effect of Se occurs at the transcriptional level and is only observed for Nis promoters containing a Pax8 binding site in the Nis upstream enhancer, suggesting that Pax8 is involved in this effect. In fact, Se increases Pax8 expression and its DNA-binding capacity, and in Pax8-silenced rat thyroid cells, Nis is not Se responsive. By inhibiting Ape1 and TxnRd1 functions, we found that both enzymes are crucial for TSH and TSH plus Se stimulation of Pax8 activity and mediate the Nis response to Se treatment. INNOVATION We describe that Se increases Nis expression and activity. We demonstrate that this effect is dependent on the redox functions of Ape1 and Txn/TxnRd1 through control of the DNA binding activity of Pax8. CONCLUSION Nis expression is controlled by Txn/Ape1 through a TSH/Se-dependent mechanism. These findings open a new field of study regarding the regulation of Nis activity in thyroid cells. Antioxid. Redox Signal. 24, 855-866.
Collapse
Affiliation(s)
- Suzana G Leoni
- 1 Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid , Madrid, Spain .,2 Instituto de Salud Carlos III, Unidad de Tumores Endocrinos, Unidad Funcional de Investigación en Enfermedades Crónicas (UFIEC) , Majadahonda (Madrid), Spain
| | - Ana Sastre-Perona
- 1 Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid , Madrid, Spain
| | - Antonio De la Vieja
- 2 Instituto de Salud Carlos III, Unidad de Tumores Endocrinos, Unidad Funcional de Investigación en Enfermedades Crónicas (UFIEC) , Majadahonda (Madrid), Spain
| | - Pilar Santisteban
- 1 Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid , Madrid, Spain
| |
Collapse
|
42
|
Chung T, Youn H, Yeom CJ, Kang KW, Chung JK. Glycosylation of Sodium/Iodide Symporter (NIS) Regulates Its Membrane Translocation and Radioiodine Uptake. PLoS One 2015; 10:e0142984. [PMID: 26599396 PMCID: PMC4658105 DOI: 10.1371/journal.pone.0142984] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/29/2015] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Human sodium/iodide symporter (hNIS) protein is a membrane glycoprotein that transports iodide ions into thyroid cells. The function of this membrane protein is closely regulated by post-translational glycosylation. In this study, we measured glycosylation-mediated changes in subcellular location of hNIS and its function of iodine uptake. METHODS HeLa cells were stably transfected with hNIS/tdTomato fusion gene in order to monitor the expression of hNIS. Cellular localization of hNIS was visualized by confocal microscopy of the red fluorescence of tdTomato. The expression of hNIS was evaluated by RT-PCR and immunoblot analysis. Functional activity of hNIS was estimated by radioiodine uptake. Cyclic AMP (cAMP) and tunicamycin were used to stimulate and inhibit glycosylation, respectively. In vivo images were obtained using a Maestro fluorescence imaging system. RESULTS cAMP-mediated Glycosylation of NIS resulted in increased expression of hNIS, stimulating membrane translocation, and enhanced radioiodine uptake. In contrast, inhibition of glycosylation by treatment with tunicamycin dramatically reduced membrane translocation of intracellular hNIS, resulting in reduced radioiodine uptake. In addition, our hNIS/tdTomato fusion reporter successfully visualized cAMP-induced hNIS expression in xenografted tumors from mouse model. CONCLUSIONS These findings clearly reveal that the membrane localization of hNIS and its function of iodine uptake are glycosylation-dependent, as our results highlight enhancement of NIS expression and glycosylation with subsequent membrane localization after cAMP treatment. Therefore, enhancing functional NIS by the increasing level of glycosylation may be suggested as a promising therapeutic strategy for cancer patients who show refractory response to conventional radioiodine treatment.
Collapse
Affiliation(s)
- Taemoon Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
- Cancer Imaging Center, Seoul National University Hospital, Seoul, Korea
- * E-mail: (HY); (JKC)
| | - Chan Joo Yeom
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - June-Key Chung
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Tumor Microenvironment Global Core Research Center, Seoul National University, Seoul, Korea
- * E-mail: (HY); (JKC)
| |
Collapse
|
43
|
Nicola JP, Reyna-Neyra A, Saenger P, Rodriguez-Buritica DF, Gamez Godoy JD, Muzumdar R, Amzel LM, Carrasco N. Sodium/Iodide Symporter Mutant V270E Causes Stunted Growth but No Cognitive Deficiency. J Clin Endocrinol Metab 2015. [PMID: 26204134 PMCID: PMC4596044 DOI: 10.1210/jc.2015-1824] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
CONTEXT Iodide (I(-)), an essential constituent of the thyroid hormones, is actively accumulated in the thyroid by the Na(+)/I(-) symporter (NIS), a key plasma membrane protein encoded by the slc5a5 gene. Mutations in slc5a5 cause I(-) transport defects (ITDs), autosomal-recessive disorders in which I(-) accumulation is totally or partially impaired, leading to congenital hypothyroidism. The characterization of NIS mutants has yielded significant insights into the molecular mechanism of NIS. OBJECTIVE This study aimed to determine the basis of a patient's ITD clinical phenotype, by sequencing her slc5a5 gene. DESIGN Genomic DNA was purified and the slc5a5 gene sequence determined. Functional in vitro studies were performed to characterize the V270E NIS mutant. PATIENT The index patient was diagnosed with hypothyroidism with minimal radioiodide uptake in a normally located, although enlarged, thyroid gland. RESULTS We identified a new NIS mutation: V270E. The patient had the compound heterozygous NIS mutation R124H/V270E. R124H NIS has been characterized previously. We show that V270E markedly reduces I(-) uptake via a pronounced (but not total) impairment of the protein's plasma membrane targeting. Remarkably, V270E is intrinsically active. Therefore, a negative charge at position 270 interferes with NIS cell surface trafficking. The patient's minimal I(-) uptake enabled sufficient thyroid hormone biosynthesis to prevent cognitive impairment. CONCLUSIONS A nonpolar residue at position 270, which all members of the SLC5A family have, is required for NIS plasma membrane targeting.
Collapse
Affiliation(s)
- Juan Pablo Nicola
- Department of Cellular and Molecular Physiology (J.P.N., A.R.-N., N.C.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Pediatrics (P.S., J.D.G.G.), Winthrop-University Hospital, Mineola, New York 11501; Department of Genetics (D.F.R.-B.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Department of Pediatrics (R.M.), Albert Einstein College of Medicine, Bronx, New York 10467; and Department of Biophysics and Biophysical Chemistry (L.M.A.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Andrea Reyna-Neyra
- Department of Cellular and Molecular Physiology (J.P.N., A.R.-N., N.C.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Pediatrics (P.S., J.D.G.G.), Winthrop-University Hospital, Mineola, New York 11501; Department of Genetics (D.F.R.-B.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Department of Pediatrics (R.M.), Albert Einstein College of Medicine, Bronx, New York 10467; and Department of Biophysics and Biophysical Chemistry (L.M.A.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Paul Saenger
- Department of Cellular and Molecular Physiology (J.P.N., A.R.-N., N.C.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Pediatrics (P.S., J.D.G.G.), Winthrop-University Hospital, Mineola, New York 11501; Department of Genetics (D.F.R.-B.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Department of Pediatrics (R.M.), Albert Einstein College of Medicine, Bronx, New York 10467; and Department of Biophysics and Biophysical Chemistry (L.M.A.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - David F Rodriguez-Buritica
- Department of Cellular and Molecular Physiology (J.P.N., A.R.-N., N.C.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Pediatrics (P.S., J.D.G.G.), Winthrop-University Hospital, Mineola, New York 11501; Department of Genetics (D.F.R.-B.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Department of Pediatrics (R.M.), Albert Einstein College of Medicine, Bronx, New York 10467; and Department of Biophysics and Biophysical Chemistry (L.M.A.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - José David Gamez Godoy
- Department of Cellular and Molecular Physiology (J.P.N., A.R.-N., N.C.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Pediatrics (P.S., J.D.G.G.), Winthrop-University Hospital, Mineola, New York 11501; Department of Genetics (D.F.R.-B.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Department of Pediatrics (R.M.), Albert Einstein College of Medicine, Bronx, New York 10467; and Department of Biophysics and Biophysical Chemistry (L.M.A.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Radhika Muzumdar
- Department of Cellular and Molecular Physiology (J.P.N., A.R.-N., N.C.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Pediatrics (P.S., J.D.G.G.), Winthrop-University Hospital, Mineola, New York 11501; Department of Genetics (D.F.R.-B.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Department of Pediatrics (R.M.), Albert Einstein College of Medicine, Bronx, New York 10467; and Department of Biophysics and Biophysical Chemistry (L.M.A.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - L Mario Amzel
- Department of Cellular and Molecular Physiology (J.P.N., A.R.-N., N.C.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Pediatrics (P.S., J.D.G.G.), Winthrop-University Hospital, Mineola, New York 11501; Department of Genetics (D.F.R.-B.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Department of Pediatrics (R.M.), Albert Einstein College of Medicine, Bronx, New York 10467; and Department of Biophysics and Biophysical Chemistry (L.M.A.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Nancy Carrasco
- Department of Cellular and Molecular Physiology (J.P.N., A.R.-N., N.C.), Yale University School of Medicine, New Haven, Connecticut 06510; Department of Pediatrics (P.S., J.D.G.G.), Winthrop-University Hospital, Mineola, New York 11501; Department of Genetics (D.F.R.-B.), University of Alabama at Birmingham, Birmingham, Alabama 35294; Department of Pediatrics (R.M.), Albert Einstein College of Medicine, Bronx, New York 10467; and Department of Biophysics and Biophysical Chemistry (L.M.A.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
44
|
Arriagada AA, Albornoz E, Opazo MC, Becerra A, Vidal G, Fardella C, Michea L, Carrasco N, Simon F, Elorza AA, Bueno SM, Kalergis AM, Riedel CA. Excess iodide induces an acute inhibition of the sodium/iodide symporter in thyroid male rat cells by increasing reactive oxygen species. Endocrinology 2015; 156:1540-51. [PMID: 25594695 PMCID: PMC5393323 DOI: 10.1210/en.2014-1371] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Na+/I- symporter (NIS) mediates iodide (I-) uptake in the thyroid gland, the first and rate-limiting step in the biosynthesis of the thyroid hormones. The expression and function of NIS in thyroid cells is mainly regulated by TSH and by the intracellular concentration of I-. High doses of I- for 1 or 2 days inhibit the synthesis of thyroid hormones, a process known as the Wolff-Chaikoff effect. The cellular mechanisms responsible for this physiological response are mediated in part by the inhibition of I- uptake through a reduction of NIS expression. Here we show that inhibition of I- uptake occurs as early as 2 hours or 5 hours after exposure to excess I- in FRTL-5 cells and the rat thyroid gland, respectively. Inhibition of I- uptake was not due to reduced NIS expression or altered localization in thyroid cells. We observed that incubation of FRTL-5 cells with excess I- for 2 hours increased H2O2 generation. Furthermore, the inhibitory effect of excess I- on NIS-mediated I- transport could be recapitulated by H2O2 and reverted by reactive derived oxygen species scavengers. The data shown here support the notion that excess I- inhibits NIS at the cell surface at early times by means of a posttranslational mechanism that involves reactive derived oxygen species.
Collapse
Affiliation(s)
- Alejandro A Arriagada
- Facultad de Ciencias Biológicas y Medicina (A.A.A., E.A., M.C.O., A.B., G.V., F.S., A.A.E., C.A.R.), Universidad Andrés Bello, República 217, Piso 4, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy (A.A.A., E.A., M.C.O., A.B., G.V., C.F., L.M., F.S., A.A.E., S.M.B., A.M.K., C.A.R.), Departamento de Endocrinología (C.F.) and Departamento de Reumatología (A.M.K.), Facultad de Medicina, and Departamento de Genética Molecular y Microbiología (S.M.B., A.M.K.), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331010 Santiago, Chile; Center for Molecular Studies of the Cell (L.M.), ICBM, Facultad de Medicina, Universidad De Chile, 6640750 Santiago, Chile; Department of Cellular and Molecular Physiology (N.C.), Yale School of Medicine, New Haven, Connecticut 06520; and INSERM Unité Mixte de Recherche 1064 (S.M.B., A.M.K., C.A.R.), 44000 Nantes, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Salzano M, Russo E, Salzano S, Bifulco M, Vitale M. Ras oncoprotein disrupts the TSH/CREB signaling upstream adenylyl cyclase in human thyroid cell. J Cell Physiol 2014; 229:2137-41. [PMID: 24819468 DOI: 10.1002/jcp.24672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/05/2014] [Accepted: 05/09/2014] [Indexed: 01/27/2023]
Abstract
Activating mutations in RAS genes and p21 Ras overactivation are common occurrences in a variety of human tumors. p21 Ras oncoproteins deregulate a number of signaling pathways, dedifferentiating the thyroid cell, and negatively regulating the expression of thyroid specific genes. In rat thyroid cells, Ras oncoproteins inhibit the TSH pathway by reducing PKA activity and thus the expression of thyroid specific genes, while in mouse melanocytes, Ras oncoproteins reduce the αMSH-stimulated cAMP signaling by increasing the expression of the phosphodiesterase-4B. Given these cell-dependent differences, we investigated if and how the TSH/CREB pathway is modulated by Ras oncoprotein in a human thyroid cell line. CREB phosphorylation was stimulated by TSH and forskolin in TAD-2 cells. Ras(V12) expression negatively regulated the TSH-stimulated CREB phosphorylation but was ineffective on forskolin-stimulated CREB phosphorylation. Phosphodiesterase inhibition by IBMX enhanced TSH-stimulated CREB phosphorylation, but did not restore TSH-stimulated CREB phosphorylation inhibited by Ras oncoprotein. These data indicate that Ras oncoprotein disrupts the TSH/CREB pathway, upstream adenylyl cyclase, and highlight the existence of mechanisms of interaction between Ras and the cAMP pathway different in human and in rat thyroid cells.
Collapse
Affiliation(s)
- Marcella Salzano
- Department of Medicine and Surgery, University of Salerno, Baronissi 84081, Salerno, Italy; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
| | | | | | | | | |
Collapse
|
47
|
Plantinga TS, Heinhuis B, Gerrits D, Netea MG, Joosten LAB, Hermus ARMM, Oyen WJG, Schweppe RE, Haugen BR, Boerman OC, Smit JWA, Netea-Maier RT. mTOR Inhibition promotes TTF1-dependent redifferentiation and restores iodine uptake in thyroid carcinoma cell lines. J Clin Endocrinol Metab 2014; 99:E1368-75. [PMID: 24712572 PMCID: PMC5393487 DOI: 10.1210/jc.2014-1171] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CONCEPT Redifferentiation of thyroid carcinoma cells has the potential to increase the efficacy of radioactive iodine therapy in treatment-refractory, nonmedullary thyroid carcinoma (TC), leading to an improved disease outcome. Mammalian target of rapamycin (mTOR) is a key regulator of cell fate affecting survival and differentiation, with autophagy and inflammation as prominent downstream pathways. METHODS The effects of mTOR inhibition were studied for its redifferentiation potential of the human TC cell lines BC-PAP, FTC133, and TPC1 by assessment of mRNA and protein expression of thyroid-specific genes and by performance of iodine uptake assays. RESULTS In thyroid transcription factor 1 (TTF1)-expressing cell lines, mTOR inhibition promoted redifferentiation of TC cells by the up-regulation of human sodium-iodine symporter mRNA and protein expression. Furthermore, these cells exhibited markedly elevated iodine uptake capacity. Surprisingly, this redifferentiation process was not mediated by autophagy induced during mTOR inhibition or by inflammatory mediators but through transcriptional effects at the level of TTF1 expression. Accordingly, small interfering RNA inhibition of TTF1 completely abrogated the induction of human sodium-iodine symporter by mTOR inhibition. CONCLUSION The present study has identified the TTF1-dependent molecular mechanisms through which the inhibition of mTOR leads to the redifferentiation of TC cells and subsequently to increased radioactive iodine uptake.
Collapse
Affiliation(s)
- Theo S Plantinga
- Departments of Internal Medicine (T.S.P., B.H., M.G.N., L.A.B.J., A.R.M.M.H., J.W.A.S., R.T.N.-M.) and Nuclear Medicine (D.G., W.J.G.O., O.C.B.) and Division of Endocrinology (T.S.P., A.R.M.M.H., J.W.A.S., R.T.N.-M.), Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; and Division of Endocrinology, Diabetes, and Metabolism (R.E.S., B.R.H.), University of Colorado Denver, Aurora, Colorado 80045
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Elisei R, Agate L, Viola D, Matrone A, Biagini A, Molinaro E. How to manage patients with differentiated thyroid cancer and a rising serum thyroglobulin level. Endocrinol Metab Clin North Am 2014; 43:331-44. [PMID: 24891165 DOI: 10.1016/j.ecl.2014.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Serum thyroglobulin (sTg) is the marker for monitoring persistence/recurrence of differentiated thyroid cancer, in patients without sTg antibodies. Patients with undetectable basal sTg or peak sTg <2 ng/mL are cured with low risk to recur. Newly detectable level of sTg indicates the recurrence. The significance of increasing sTg in patients treated with emithyroidectomy or total-thyroidectomy but not ablated with radioiodine is undefined. A doubling time <1 year may be a poor prognostic factor, but this is more relevant in cases with high levels of sTg. Because of its sensitivity, neck ultrasound should be performed at any visit, especially when an increased sTg is seen.
Collapse
Affiliation(s)
- Rossella Elisei
- Endocrinology Unit, Department of Clinical and Experimental Medicine, WHO Collaborating Center for the Study and Treatment of Thyroid Diseases and Other Endocrine and Metabolic Disorders, University of Pisa, Via Paradisa 2, Pisa 56124, Italy.
| | - Laura Agate
- Endocrinology Unit, Department of Clinical and Experimental Medicine, WHO Collaborating Center for the Study and Treatment of Thyroid Diseases and Other Endocrine and Metabolic Disorders, University of Pisa, Via Paradisa 2, Pisa 56124, Italy
| | - David Viola
- Endocrinology Unit, Department of Clinical and Experimental Medicine, WHO Collaborating Center for the Study and Treatment of Thyroid Diseases and Other Endocrine and Metabolic Disorders, University of Pisa, Via Paradisa 2, Pisa 56124, Italy
| | - Antonio Matrone
- Endocrinology Unit, Department of Clinical and Experimental Medicine, WHO Collaborating Center for the Study and Treatment of Thyroid Diseases and Other Endocrine and Metabolic Disorders, University of Pisa, Via Paradisa 2, Pisa 56124, Italy
| | - Agnese Biagini
- Endocrinology Unit, Department of Clinical and Experimental Medicine, WHO Collaborating Center for the Study and Treatment of Thyroid Diseases and Other Endocrine and Metabolic Disorders, University of Pisa, Via Paradisa 2, Pisa 56124, Italy
| | - Eleonora Molinaro
- Endocrinology Unit, Department of Clinical and Experimental Medicine, WHO Collaborating Center for the Study and Treatment of Thyroid Diseases and Other Endocrine and Metabolic Disorders, University of Pisa, Via Paradisa 2, Pisa 56124, Italy
| |
Collapse
|
49
|
Rauer C, Ringseis R, Rothe S, Wen G, Eder K. Sterol regulatory element-binding proteins are regulators of the rat thyroid peroxidase gene in thyroid cells. PLoS One 2014; 9:e91265. [PMID: 24625548 PMCID: PMC3953333 DOI: 10.1371/journal.pone.0091265] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 02/07/2014] [Indexed: 01/21/2023] Open
Abstract
Sterol regulatory element-binding proteins (SREBPs)-1c and -2, which were initially discovered as master transcriptional regulators of lipid biosynthesis and uptake, were recently identified as novel transcriptional regulators of the sodium-iodide symporter gene in the thyroid, which is essential for thyroid hormone synthesis. Based on this observation that SREBPs play a role for thyroid hormone synthesis, we hypothesized that another gene involved in thyroid hormone synthesis, the thyroid peroxidase (TPO) gene, is also a target of SREBP-1c and -2. Thyroid epithelial cells treated with 25-hydroxycholesterol, which is known to inhibit SREBP activation, had about 50% decreased mRNA levels of TPO. Similarly, the mRNA level of TPO was reduced by about 50% in response to siRNA mediated knockdown of both, SREBP-1 and SREBP-2. Reporter gene assays revealed that overexpression of active SREBP-1c and -2 causes a strong transcriptional activation of the rat TPO gene, which was localized to an approximately 80 bp region in the intron 1 of the rat TPO gene. In vitro- and in vivo-binding of both, SREBP-1c and SREBP-2, to this region in the rat TPO gene could be demonstrated using gel-shift assays and chromatin immunoprecipitation. Mutation analysis of the 80 bp region of rat TPO intron 1 revealed two isolated and two overlapping SREBP-binding elements from which one, the overlapping SRE+609/InvSRE+614, was shown to be functional in reporter gene assays. In connection with recent findings that the rat NIS gene is also a SREBP target gene in the thyroid, the present findings suggest that SREBPs may be possible novel targets for pharmacological modulation of thyroid hormone synthesis.
Collapse
Affiliation(s)
- Christine Rauer
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
- * E-mail:
| | - Susanne Rothe
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Gaiping Wen
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-Universität Gießen, Gießen, Germany
| |
Collapse
|
50
|
Serrano-Nascimento C, da Silva Teixeira S, Nicola JP, Nachbar RT, Masini-Repiso AM, Nunes MT. The acute inhibitory effect of iodide excess on sodium/iodide symporter expression and activity involves the PI3K/Akt signaling pathway. Endocrinology 2014; 155:1145-56. [PMID: 24424051 DOI: 10.1210/en.2013-1665] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Iodide (I(-)) is an irreplaceable constituent of thyroid hormones and an important regulator of thyroid function, because high concentrations of I(-) down-regulate sodium/iodide symporter (NIS) expression and function. In thyrocytes, activation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) cascade also inhibits NIS expression and function. Because I(-) excess and PI3K/Akt signaling pathway induce similar inhibitory effects on NIS expression, we aimed to study whether the PI3K/Akt cascade mediates the acute and rapid inhibitory effect of I(-) excess on NIS expression/activity. Here, we reported that the treatment of PCCl3 cells with I(-) excess increased Akt phosphorylation under normal or TSH/insulin-starving conditions. I(-) stimulated Akt phosphorylation in a PI3K-dependent manner, because the use of PI3K inhibitors (wortmannin or 2-(4-Morpholinyl)-8-phenyl-4H-1-benzopyran-4-one) abrogated the induction of I(-) effect. Moreover, I(-) inhibitory effect on NIS expression and function were abolished when the cells were previously treated with specific inhibitors of PI3K or Akt (Akt1/2 kinase inhibitor). Importantly, we also found that the effect of I(-) on NIS expression involved the generation of reactive oxygen species (ROS). Using the fluorogenic probes dihydroethidium and mitochondrial superoxide indicator (MitoSOX Red), we observed that I(-) excess increased ROS production in thyrocytes and determined that mitochondria were the source of anion superoxide. Furthermore, the ROS scavengers N-acetyl cysteine and 2-phenyl-1,2-benzisoselenazol-3-(2H)-one blocked the effect of I(-) on Akt phosphorylation. Overall, our data demonstrated the involvement of the PI3K/Akt signaling pathway as a novel mediator of the I(-)-induced thyroid autoregulation, linking the role of thyroid oxidative state to the Wolff-Chaikoff effect.
Collapse
Affiliation(s)
- Caroline Serrano-Nascimento
- Department of Physiology and Biophysics (C.S.-N., S.d.S.T., R.T.N., M.T.N.), Institute of Biomedical Sciences, University of São Paulo, 05508-000 São Paulo, Brazil; and Centro de Investigaciones en Bioquímica Clínica e Inmunología-Consejo Nacional de Investigaciones Científicas y Técnicas (J.P.N., A.M.M.-R.), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA Córdoba, Argentina
| | | | | | | | | | | |
Collapse
|