1
|
Abe K, Koizumi M, Kogai T, Ida S, Sugisawa C, Kawai M, Hasegawa T, Narumi S. Two Novel SLC5A5 Variants (Q263L and G350D) Causing Congenital Hypothyroidism. Thyroid 2025; 35:335-337. [PMID: 39895319 DOI: 10.1089/thy.2024.0716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
SLC5A5 encodes sodium-iodide symporter (NIS), which transports inorganic iodide into thyroid cells. Biallelic loss-of-function variants in SLC5A5 cause thyroid dyshormonogenesis due to iodide transport defect (ITD). We report a Japanese sibling with ITD carrying novel compound heterozygous SLC5A5 variants (p. [Gln263Leu]; [Gly350Asp]). The elder brother was diagnosed with congenital hypothyroidism (CH) through newborn screening (NBS), while the younger brother, with a negative NBS result, developed CH-related symptoms at age 3 months. We characterized the two variant NIS proteins in vitro and negligible iodide transport capacity of both proteins. These findings provide unique evidence for the structure-function relationship of the NIS protein.
Collapse
Affiliation(s)
- Kiyomi Abe
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
- Department of Pediatrics, Tokyo Saiseikai Central Hospital, Tokyo, Japan
| | - Mikiko Koizumi
- Department of Gastroenterology, Nutrition and Endocrinology, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Takahiko Kogai
- Department of Genetic Diagnosis and Laboratory Medicine, Dokkyo Medical University, Tochigi, Japan
| | - Shinobu Ida
- Department of Gastroenterology, Nutrition and Endocrinology, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Chiho Sugisawa
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Masanobu Kawai
- Department of Gastroenterology, Nutrition and Endocrinology, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Narumi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
2
|
Costa IM, Firth G, Kim J, Banu A, Pham TT, Sunassee K, Langdon S, De Santis V, Vass L, Schettino G, Fruhwirth GO, Terry SYA. In Vitro and Preclinical Systematic Dose-Effect Studies of Auger Electron- and β Particle-Emitting Radionuclides and External Beam Radiation for Cancer Treatment. Int J Radiat Oncol Biol Phys 2024; 120:1124-1134. [PMID: 38797497 PMCID: PMC7616868 DOI: 10.1016/j.ijrobp.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/09/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE Despite a rise in clinical use of radiopharmaceutical therapies, the biological effects of radionuclides and their relationship with absorbed radiation dose are poorly understood. Here, we set out to define this relationship for Auger electron emitters [99mTc]TcO4- and [123I]I- and β--particle emitter [188Re]ReO4-. Studies were carried out using genetically modified cells that permitted direct radionuclide comparisons. METHODS AND MATERIALS Triple-negative MDA-MB-231 breast cancer cells expressing the human sodium iodide symporter (hNIS) and green fluorescent protein (GFP; MDA-MB-231.hNIS-GFP) were used. In vitro radiotoxicity of [99mTc]TcO4-, [123I]I-, and [188Re]ReO4- was determined using clonogenic assays. Radionuclide uptake, efflux, and subcellular location were used to calculate nuclear absorbed doses using the Medical Internal Radiation Dose (MIRD) formalism. In vivo studies were performed using female NSG mice bearing orthotopic MDA-MB-231.hNIS-GFP tumors and compared with X-ray-treated (12.6-15 Gy) and untreated cohorts. Absorbed dose per unit activity in tumors and sodium iodide symporter-expressing organs was extrapolated to reference human adult models using OLINDA/EXM. RESULTS [99mTc]TcO4- and [123I]I- reduced the survival fraction only in hNIS-expressing cells, whereas [188Re]ReO4- reduced survival fraction in hNIS-expressing and parental cells. [123I]I- required 2.4- and 1.5-fold lower decays/cell to achieve 37% survival compared with [99mTc]TcO4- and [188Re]ReO4-, respectively, after 72 hours of incubation. Additionally, [99mTc]TcO4-, [123I]I-, and [188Re]ReO4- had superior cell killing effectiveness in vitro compared with X-rays. In vivo, X-ray led to a greater median survival compared with [188Re]ReO4- and [123I]I- (54 days vs 45 and 43 days, respectively). Unlike the X-ray cohort, no metastases were visualized in the radionuclide-treated cohorts. Extrapolated human absorbed doses of [188Re]ReO4- to a 1 g tumor were 13.8- and 11.2-fold greater than for [123I]I- in female and male models, respectively. CONCLUSIONS This work reports reference dose-effect data using cell and tumor models for [99mTc]TcO4-, [123I]I-, and [188Re]ReO4- for the first time. We further demonstrate the tumor-controlling effects of [123I]I- and [188Re]ReO4- in comparison with external beam radiation therapy.
Collapse
Affiliation(s)
- Ines M Costa
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom; Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - George Firth
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Jana Kim
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Arshiya Banu
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Truc T Pham
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Kavitha Sunassee
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Sophie Langdon
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Vittorio De Santis
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Laurence Vass
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Giuseppe Schettino
- Medical Radiation Science Group, National Physical Laboratory, Teddington, United Kingdom.
| | - Gilbert O Fruhwirth
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom.
| | - Samantha Y A Terry
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
3
|
Liang C, Wu C, Liu L, Zhong J. Update on lacrimal apparatus dysfunction associated with differentiated thyroid cancer after I-131 therapy. Int Ophthalmol 2024; 44:257. [PMID: 38909080 DOI: 10.1007/s10792-024-03192-9] [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: 02/27/2024] [Accepted: 06/15/2024] [Indexed: 06/24/2024]
Abstract
PURPOSE The most prevalent lacrimal apparatus dysfunctions associated with differentiated thyroid cancer(DTC) after I-131 therapy are dry eye and nasolacrimal duct obstruction(NLDO), leading to ocular discomfort and lower quality of life for patients. It is crucial to diagnose and manage lacrimal apparatus dysfunction associated with I-131 therapy for DTC. Therefore, this review aims to comprehensively summarize and analyze the advances in mechanisms and therapeutic options underlying lacrimal apparatus dysfunction induced by I-131 therapy for DTC. METHODS A comprehensive search of CNKI, PubMed, and Wed of Science was performed from the database to December of 2023. Key search terms were "Thyroid cancer", "I-131", "Complications", "Dry eye", "Epiphora", "Tear", "Nasolacrimal duct" and "NLDO". RESULTS The research indicates that I-131 therapy for DTC causes damage to the lacrimal glands and nasolacrimal duct system, resulting in symptoms such as dry eye, epiphora, and mucoid secretions. Moreover, recent research has focused on exploring relevant risk factors of the condition and experimental and clinical treatments. However, there is some controversy regarding the mechanisms involved, whether it is due to the passive flow of I-131 in tears, active uptake of I-131 by the sodium-iodide symporter (NIS) in the lacrimal sac and nasolacrimal duct, or secondary metabolic and hormonal disturbances caused by I-131. CONCLUSION It is crucial for early detection and preventive measures by ophthalmologists and the need for further studies to elucidate the mechanisms underlying the disease.
Collapse
Affiliation(s)
- Chunlan Liang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, 613 West Huangpu Ave, Guangzhou, 510632, China
| | - Changlin Wu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, 613 West Huangpu Ave, Guangzhou, 510632, China
| | - Lian Liu
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, 613 West Huangpu Ave, Guangzhou, 510632, China.
| | - Jingxiang Zhong
- Department of Ophthalmology, The Sixth Affiliated Hospital of Jinan University, No. 88, Changdong Road, Dongguan, 523573, China.
| |
Collapse
|
4
|
Chakrabarti M, Amzel LM, Lau AY. Sodium/Iodide Symporter Metastable Intermediates Provide Insights into Conformational Transition between Principal Thermodynamic States. J Phys Chem B 2023; 127:1540-1551. [PMID: 36758032 DOI: 10.1021/acs.jpcb.2c07604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The Sodium/Iodide Symporter (NIS), a 13-helix transmembrane protein found in the thyroid and other tissues, transports iodide, a required constituent of thyroid hormones T3 and T4. Despite extensive experimental information and clinical data, structural details of the intermediate microstates comprising the conformational transition of NIS between its inwardly and outwardly open states remain unresolved. We present data from a combination of enhanced sampling and transition path molecular dynamics (MD) simulations that elucidate the principal intermediate states comprising the inwardly to outwardly open transition of fully bound and apo NIS under an enforced ionic gradient. Our findings suggest that in both the absence and presence of bound physiological ions, NIS principally occupies a proximally inward to inwardly open state. When fully bound, NIS is also found to occupy a rare "inwardly occluded" state. The results of this work provide novel insight into the populations of NIS intermediates and the free energy landscape comprising the conformational transition, adding to a mechanistic understanding of NIS ion transport. Moreover, the knowledge gained from this approach can serve as a basis for studies of NIS mutants to target therapeutic interventions.
Collapse
Affiliation(s)
- Mayukh Chakrabarti
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., PO Box B, Frederick, Maryland 21702, United States
| | - L Mario Amzel
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, Maryland 21205, United States
| | - Albert Y Lau
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, Maryland 21205, United States
| |
Collapse
|
5
|
The sodium iodide symporter (NIS) as theranostic gene: potential role in pre-clinical therapy of extra-thyroidal malignancies. Clin Transl Imaging 2023. [DOI: 10.1007/s40336-023-00540-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
6
|
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
|
7
|
Karbownik-Lewińska M, Stępniak J, Iwan P, Lewiński A. Iodine as a potential endocrine disruptor-a role of oxidative stress. Endocrine 2022; 78:219-240. [PMID: 35726078 PMCID: PMC9584999 DOI: 10.1007/s12020-022-03107-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/02/2022] [Indexed: 12/19/2022]
Abstract
PURPOSE Iodine is an essential micronutrient required for thyroid hormone biosynthesis. However, overtreatment with iodine can unfavorably affect thyroid physiology. The aim of this review is to present the evidence that iodine-when in excess-can interfere with thyroid hormone synthesis and, therefore, can act as a potential endocrine-disrupting chemical (EDC), and that this action, as well as other abnormalities in the thyroid, occurs-at least partially-via oxidative stress. METHODS We reviewed published studies on iodine as a potential EDC, with particular emphasis on the phenomenon of oxidative stress. RESULTS This paper summarizes current knowledge on iodine excess in the context of its properties as an EDC and its effects on oxidative processes. CONCLUSION Iodine does fulfill the criteria of an EDC because it is an exogenous chemical that interferes-when in excess-with thyroid hormone synthesis. However, this statement cannot change general rules regarding iodine supply, which means that iodine deficiency should be still eliminated worldwide and, at the same time, iodine excess should be avoided. Universal awareness that iodine is a potential EDC would make consumers more careful regarding their diet and what they supplement in tablets, and-what is of great importance-it would make caregivers choose iodine-containing medications (or other chemicals) more prudently. It should be stressed that compared to iodine deficiency, iodine in excess (acting either as a potential EDC or via other mechanisms) is much less harmful in such a sense that it affects only a small percentage of sensitive individuals, whereas the former affects whole populations; therefore, it causes endemic consequences.
Collapse
Affiliation(s)
- Małgorzata Karbownik-Lewińska
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752, Lodz, Poland.
- Polish Mother's Memorial Hospital-Research Institute, 93-338, Lodz, Poland.
| | - Jan Stępniak
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752, Lodz, Poland
| | - Paulina Iwan
- Department of Oncological Endocrinology, Medical University of Lodz, 90-752, Lodz, Poland
| | - Andrzej Lewiński
- Polish Mother's Memorial Hospital-Research Institute, 93-338, Lodz, Poland
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 93-338, Lodz, Poland
| |
Collapse
|
8
|
Koumarianou P, Fernández-Méndez C, Fajardo-Delgado D, Mielu LM, Santisteban P, De la Vieja A. Basolateral Sorting of the Sodium/Iodide Symporter Is Mediated by Adaptor Protein 1 Clathrin Adaptor Complexes. Thyroid 2022; 32:1259-1270. [PMID: 35833460 PMCID: PMC9618391 DOI: 10.1089/thy.2022.0163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background: The sodium/iodide symporter (NIS) is a transmembrane protein located on the basolateral membrane of thyrocytes. Despite its physiological and clinical relevance, little is known about the mechanisms that mediate NIS subcellular sorting. In the present study, we examined NIS basolateral trafficking in vitro using non-thyroid and thyroid epithelial cells. Methods: Immunofluorescence and Western blotting were performed to analyze NIS subcellular location and function in cells grown in monolayers under unpolarized and/or polarized conditions. Strategic NIS residues were mutated, and binding of NIS to clathrin adaptor complexes was determined by immunoprecipitation. Results: We show that NIS reaches the plasma membrane (PM) through a thyrotropin-dependent mechanism 24 hours after treatment with the hormone. We demonstrate that NIS basolateral trafficking is a clathrin-mediated mechanism, in which the clathrin adaptor complexes AP-1 (A and B) sort NIS from the trans-Golgi network (TGN) and recycling endosomes (REs). Specifically, we show that the AP-1B μ1 subunit controls NIS basolateral sorting through common REs. In its absence, NIS is apically missorted but remains functional. Additionally, direct NIS basolateral transport from the TGN to the basolateral membrane is mediated by AP-1A through clathrin-coated vesicles that also carry the transferrin receptor. Loss of the μ1 subunit of AP-1A is functionally compensated by AP-1B. Furthermore, loss of both subunits diminishes NIS trafficking to the PM. Finally, we demonstrate that AP-1A binds to the L121 and LL562/563 residues on NIS, whereas AP-1B binds to L583. Conclusions: Our findings highlight the novel involvement of the clathrin-coated machinery in basolateral NIS trafficking. Given that AP-1A expression is reduced in tumors, and its expression correlates with that of NIS, these findings will help uncover new targets in thyroid cancer treatment.
Collapse
Affiliation(s)
- Petrina Koumarianou
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid (UAM), Spain
| | - Celia Fernández-Méndez
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid (UAM), Spain
| | - Dánae Fajardo-Delgado
- Unidad de Tumores Endocrinos (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Lidia Mirella Mielu
- Unidad de Tumores Endocrinos (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Ciber de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, Madrid (UAM), Spain
- Ciber de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Address correspondence to: Pilar Santisteban, PhD, Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, C/Arturo Duperier 4, Madrid 28029, Spain
| | - Antonio De la Vieja
- Unidad de Tumores Endocrinos (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Ciber de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Antonio De la Vieja, PhD, Unidad de Tumores Endocrinos (UFIEC), Instituto de Salud Carlos III (ISCIII), Ctra. Majadahonda-Pozuelo km 2,2., Majadahonda (Madrid) 28220, Spain
| |
Collapse
|
9
|
Arnhold J, Malle E. Halogenation Activity of Mammalian Heme Peroxidases. Antioxidants (Basel) 2022; 11:antiox11050890. [PMID: 35624754 PMCID: PMC9138014 DOI: 10.3390/antiox11050890] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/10/2022] Open
Abstract
Mammalian heme peroxidases are fascinating due to their unique peculiarity of oxidizing (pseudo)halides under physiologically relevant conditions. These proteins are able either to incorporate oxidized halides into substrates adjacent to the active site or to generate different oxidized (pseudo)halogenated species, which can take part in multiple (pseudo)halogenation and oxidation reactions with cell and tissue constituents. The present article reviews basic biochemical and redox mechanisms of (pseudo)halogenation activity as well as the physiological role of heme peroxidases. Thyroid peroxidase and peroxidasin are key enzymes for thyroid hormone synthesis and the formation of functional cross-links in collagen IV during basement membrane formation. Special attention is directed to the properties, enzymatic mechanisms, and resulting (pseudo)halogenated products of the immunologically relevant proteins such as myeloperoxidase, eosinophil peroxidase, and lactoperoxidase. The potential role of the (pseudo)halogenated products (hypochlorous acid, hypobromous acid, hypothiocyanite, and cyanate) of these three heme peroxidases is further discussed.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Medical Faculty, Institute of Medical Physics and Biophysics, Leipzig University, 04107 Leipzig, Germany
- Correspondence: (J.A.); or (E.M.)
| | - Ernst Malle
- Gottfried Schatz Research Center, Division of Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
- Correspondence: (J.A.); or (E.M.)
| |
Collapse
|
10
|
Reyna-Neyra A, Jung L, Chakrabarti M, Suárez MX, Amzel LM, Carrasco N. The Iodide Transport Defect-Causing Y348D Mutation in the Na +/I - Symporter Renders the Protein Intrinsically Inactive and Impairs Its Targeting to the Plasma Membrane. Thyroid 2021; 31:1272-1281. [PMID: 33779310 PMCID: PMC8377515 DOI: 10.1089/thy.2020.0931] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background: The sodium/iodide (Na+/I-) symporter (NIS) mediates active transport of I- into the thyroid gland. Mutations in the SLC5A5 gene, which encodes NIS, cause I- transport defects (ITDs)-which, if left untreated, lead to congenital hypothyroidism and consequent cognitive and developmental deficiencies. The ITD-causing NIS mutation Y348D, located in transmembrane segment (TMS) 9, was reported in three Sudanese patients. Methods: We generated cDNAs coding for Y348D NIS and mutants with other hydrophilic and hydrophobic amino acid substitutions at position 348 and transfected them into cells. The activity of the resulting mutants was quantitated by radioiodide transport assays. NIS glycosylation was investigated by Western blotting after endoglycosidase H (Endo H) and PNGase-F glycosidase treatment. Subcellular localization of the mutant proteins was ascertained by flow cytometry analysis, cell surface biotinylation, and immunofluorescence. The intrinsic activity of Y348D was studied by measuring radioiodide transport in membrane vesicles prepared from Y348D-NIS-expressing cells. Our NIS homology models and molecular dynamics simulations were used to identify residues that interact with Y348 and investigate possible interactions between Y348 and the membrane. The sequences of several Slc5 family transporters were aligned, and a phylogenetic tree was generated in ClustalX. Results: Cells expressing Y348D NIS transport no I-. Furthermore, Y348D NIS is only partially glycosylated, is retained intracellularly, and is intrinsically inactive. Hydrophilic residues other than Asp at position 348 also yield NIS proteins that fail to be targeted to the plasma membrane (PM), whereas hydrophobic residues at this position, which we show do not interact with the membrane, rescue PM targeting and function. Conclusions: Y348D NIS does not reach the PM and is intrinsically inactive. Hydrophobic amino acid substitutions at position 348, however, preserve NIS activity. Our findings are consistent with our homology model's prediction that Y348 should face the side opposite the TMS9 residues that coordinate Na+ and participate in Na+ transport, and with the notion that Y348 interacts only with hydrophobic residues. Hydrophilic or charged residues at position 348 have deleterious effects on NIS PM targeting and activity, whereas a hydrophobic residue at this position rescues NIS activity.
Collapse
Affiliation(s)
- Andrea Reyna-Neyra
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Lara Jung
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Mayukh Chakrabarti
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mikel X. Suárez
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - L. Mario Amzel
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nancy Carrasco
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Address correspondence to: Nancy Carrasco, MD, Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 2215 Garland Avenue, 707 Light Hall, Nashville, TN 37232-0615, USA
| |
Collapse
|
11
|
Wen G, Eder K, Ringseis R. Resveratrol Alleviates the Inhibitory Effect of Tunicamycin-Induced Endoplasmic Reticulum Stress on Expression of Genes Involved in Thyroid Hormone Synthesis in FRTL-5 Thyrocytes. Int J Mol Sci 2021; 22:ijms22094373. [PMID: 33922129 PMCID: PMC8122728 DOI: 10.3390/ijms22094373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 11/19/2022] Open
Abstract
Recently, ER stress induced by tunicamycin (TM) was reported to inhibit the expression of key genes involved in thyroid hormone synthesis, such as sodium/iodide symporter (NIS), thyroid peroxidase (TPO) and thyroglobulin (TG), and their regulators such as thyrotropin receptor (TSHR), thyroid transcription factor-1 (TTF-1), thyroid transcription factor-2 (TTF-2) and paired box gene 8 (PAX-8), in FRTL-5 thyrocytes. The present study tested the hypothesis that resveratrol (RSV) alleviates this effect of TM in FRTL-5 cells. While treatment of FRTL-5 cells with TM alone (0.1 µg/mL) for 48 h strongly induced the ER stress-sensitive genes heat shock protein family A member 5 (HSPA5) and DNA damage inducible transcript 3 (DDIT3) and repressed NIS, TPO, TG, TSHR, TTF-1, TTF-2 and PAX-8, combined treatment with TM (0.1 µg/mL) and RSV (10 µM) for 48 h attenuated this effect of TM. In conclusion, RSV alleviates TM-induced ER stress and attenuates the strong impairment of expression of genes involved in thyroid hormone synthesis and their regulators in FRTL-5 thyrocytes exposed to TM-induced ER stress. Thus, RSV may be useful for the treatment of specific thyroid disorders, provided that strategies with improved oral bioavailability of RSV are applied.
Collapse
|
12
|
De la Vieja A, Riesco-Eizaguirre G. Radio-Iodide Treatment: From Molecular Aspects to the Clinical View. Cancers (Basel) 2021; 13:cancers13050995. [PMID: 33673669 PMCID: PMC7957486 DOI: 10.3390/cancers13050995] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary This year marks the 80th commemoration of the first time that radio-iodide treatment (RAI) was used. RAI is one of the most effective targeted internal radiation anticancer therapies ever devised and it has been used for many decades, however, a thorough understanding of the underlying molecular mechanisms involved could greatly improve the success of this therapy. This is an in-depth innovative review focusing on the molecular mechanisms underlying radio-iodide therapy in thyroid cancer and how the alteration of these mechanisms affects the results in the clinic. Abstract Thyroid radio-iodide therapy (RAI) is one of the oldest known and used targeted therapies. In thyroid cancer, it has been used for more than eight decades and is still being used to improve thyroid tumor treatment to eliminate remnants after thyroid surgery, and tumor metastases. Knowledge at the molecular level of the genes/proteins involved in the process has led to improvements in therapy, both from the point of view of when, how much, and how to use the therapy according to tumor type. The effectiveness of this therapy has spread into other types of targeted therapies, and this has made sodium/iodide symporter (NIS) one of the favorite theragnostic tools. Here we focus on describing the molecular mechanisms involved in radio-iodide therapy and how the alteration of these mechanisms in thyroid tumor progression affects the diagnosis and results of therapy in the clinic. We analyze basic questions when facing treatment, such as: (1) how the incorporation of radioiodine in normal, tumor, and metastatic thyroid cells occurs and how it is regulated; (2) the pros and cons of thyroid hormonal deprivation vs. recombinant human Thyroid Stimulating Hormone (rhTSH) in radioiodine residence time, treatment efficacy, thyroglobulin levels and organification, and its influence on diagnostic imaging tests and metastasis treatment; and (3) the effect of stunning and the possible causes. We discuss the possible incorporation of massive sequencing data into clinical practice, and we conclude with a socioeconomical and clinical vision of the above aspects.
Collapse
Affiliation(s)
- Antonio De la Vieja
- Endocrine Tumors Unit (Unidad Funcional de Investigación en Enfermedades Endocrinas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
- Correspondence: ; Tel.: +34-918223270
| | - Garcilaso Riesco-Eizaguirre
- Departamento de Endocrinología y Nutrición, Hospital Universitario de Móstoles, 28935 Madrid, Spain
- Molecular Endocrinology Group, Faculty of Medicine, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Serrano-Nascimento C, Morillo-Bernal J, Rosa-Ribeiro R, Nunes MT, Santisteban P. Impaired Gene Expression Due to Iodine Excess in the Development and Differentiation of Endoderm and Thyroid Is Associated with Epigenetic Changes. Thyroid 2020; 30:609-620. [PMID: 31801416 DOI: 10.1089/thy.2018.0658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Thyroid hormone (TH) synthesis is essential for the control of development, growth, and metabolism in vertebrates and depends on a sufficient dietary iodine intake. Importantly, both iodine deficiency and iodine excess (IE) impair TH synthesis, causing serious health problems especially during fetal/neonatal development. While it is known that IE disrupts thyroid function by inhibiting thyroid gene expression, its effects on thyroid development are less clear. Accordingly, this study sought to investigate the effects of IE during the embryonic development/differentiation of endoderm and the thyroid gland. Methods: We used the murine embryonic stem (ES) cell model of in vitro directed differentiation to assess the impact of IE on the generation of endoderm and thyroid cells. Additionally, we subjected endoderm and thyroid explants obtained during early gestation to IE and evaluated gene and protein expression of endodermal markers in both models. Results: ES cells were successfully differentiated into endoderm cells and, subsequently, into thyrocytes expressing the specific thyroid markers Tshr, Slc5a5, Tpo, and Tg. IE exposure decreased the messenger RNA (mRNA) levels of the main endoderm markers Afp, Crcx4, Foxa1, Foxa2, and Sox17 in both ES cell-derived endoderm cells and embryonic explants. Interestingly, IE also decreased the expression of the main thyroid markers in ES cell-derived thyrocytes and thyroid explants. Finally, we demonstrate that DNA methyltransferase expression was increased by exposure to IE, and this was accompanied by hypermethylation and hypoacetylation of histone H3, pointing to an association between the gene repression triggered by IE and the observed epigenetic changes. Conclusions: These data establish that IE treatment is deleterious for embryonic endoderm and thyroid gene expression.
Collapse
Affiliation(s)
- Caroline Serrano-Nascimento
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAM, Madrid, Spain
- CIBERONC Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Ensino e Pesquisa, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jesús Morillo-Bernal
- Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAM, Madrid, Spain
- CIBERONC Instituto de Salud Carlos III, Madrid, Spain
| | - Rafaela Rosa-Ribeiro
- Instituto de Ensino e Pesquisa, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Maria Tereza Nunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas "Alberto Sols," CSIC-UAM, Madrid, Spain
- CIBERONC Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
16
|
Zhekova HR, Sakuma T, Johnson R, Concilio SC, Lech PJ, Zdravkovic I, Damergi M, Suksanpaisan L, Peng KW, Russell SJ, Noskov S. Mapping of Ion and Substrate Binding Sites in Human Sodium Iodide Symporter (hNIS). J Chem Inf Model 2020; 60:1652-1665. [PMID: 32134653 DOI: 10.1021/acs.jcim.9b01114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The human sodium iodide symporter (hNIS) is a theranostic reporter gene which concentrates several clinically approved SPECT and PET radiotracers and plays an essential role for the synthesis of thyroid hormones as an iodide transporter in the thyroid gland. Development of hNIS mutants which could enhance translocation of the desired imaging ions is currently underway. Unfortunately, it is hindered by lack of understanding of the 3D organization of hNIS and its relation to anion transport. There are no known crystal structures of hNIS in any of its conformational states. Homology modeling can be very effective in such situations; however, the low sequence identity between hNIS and relevant secondary transporters with available experimental structures makes the choice of a template and the generation of 3D models nontrivial. Here, we report a combined application of homology modeling and molecular dynamics refining of the hNIS structure in its semioccluded state. The modeling was based on templates from the LeuT-fold protein family and was done with emphasis on the refinement of the substrate-ion binding pocket. The consensus model developed in this work is compared to available biophysical and biochemical experimental data for a number of different LeuT-fold proteins. Some functionally important residues contributing to the formation of putative binding sites and permeation pathways for the cotransported Na+ ions and I- substrate were identified. The model predictions were experimentally tested by generation of mutant versions of hNIS and measurement of relative (to WT hNIS) 125I- uptake of 35 hNIS variants.
Collapse
Affiliation(s)
- Hristina R Zhekova
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Toshie Sakuma
- Imanis Life Sciences, Rochester, Minnesota 55901, United States.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Ryan Johnson
- Imanis Life Sciences, Rochester, Minnesota 55901, United States
| | - Susanna C Concilio
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55902, United States.,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Patrycja J Lech
- Imanis Life Sciences, Rochester, Minnesota 55901, United States
| | - Igor Zdravkovic
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mirna Damergi
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | - Kah-Whye Peng
- Imanis Life Sciences, Rochester, Minnesota 55901, United States.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Stephen J Russell
- Imanis Life Sciences, Rochester, Minnesota 55901, United States.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota 55902, United States
| | - Sergei Noskov
- Centre for Molecular Simulation, Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
17
|
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
|
18
|
Targovnik HM, Scheps KG, Rivolta CM. Defects in protein folding in congenital hypothyroidism. Mol Cell Endocrinol 2020; 501:110638. [PMID: 31751626 DOI: 10.1016/j.mce.2019.110638] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/21/2019] [Accepted: 11/01/2019] [Indexed: 12/26/2022]
Abstract
Primary congenital hypothyroidism (CH) is the most common endocrine disease in children and one of the most common preventable causes of both cognitive and motor deficits. CH is a heterogeneous group of thyroid disorders in which inadequate production of thyroid hormone occurs due to defects in proteins involved in the gland organogenesis (dysembryogenesis) or in multiple steps of thyroid hormone biosynthesis (dyshormonogenesis). Dysembryogenesis is associated with genes responsible for the development or growth of thyroid cells: such as NKX2-1, FOXE1, PAX8, NKX2-5, TSHR, TBX1, CDCA8, HOXD3 and HOXB3 resulting in agenesis, hypoplasia or ectopia of thyroid gland. Nevertheless, the etiology of the dysembryogenesis remains unknown for most cases. In contrast, the majority of patients with dyshormonogenesis has been linked to mutations in the SLC5A5, SLC26A4, SLC26A7, TPO, DUOX1, DUOX2, DUOXA1, DUOXA2, IYD or TG genes, which usually originate goiter. About 800 genetic mutations have been reported to cause CH in patients so far, including missense, nonsense, in-frame deletion and splice-site variations. Many of these mutations are implicated in specific domains, cysteine residues or glycosylation sites, affecting the maturation of nascent proteins that go through the secretory pathway. Consequently, misfolded proteins are permanently entrapped in the endoplasmic reticulum (ER) and are translocated to the cytosol for proteasomal degradation by the ER-associated degradation (ERAD) machinery. Despite of all these remarkable advances in the field of the CH pathogenesis, several points on the development of this disease remain to be elucidated. The continuous study of thyroid gene mutations with the application of new technologies will be useful for the understanding of the intrinsic mechanisms related to CH. In this review we summarize the present status of knowledge on the disorders in the protein folding caused by thyroid genes mutations.
Collapse
Affiliation(s)
- Héctor M Targovnik
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina.
| | - Karen G Scheps
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Carina M Rivolta
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Microbiología, Inmunología, Biotecnología y Genética/Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| |
Collapse
|
19
|
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
|
20
|
Chai W, Ye F, Zeng L, Li Y, Yang L. HMGB1-mediated autophagy regulates sodium/iodide symporter protein degradation in thyroid cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:325. [PMID: 31331356 PMCID: PMC6647330 DOI: 10.1186/s13046-019-1328-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/15/2019] [Indexed: 11/11/2022]
Abstract
Background Sodium/iodide symporter (NIS)-mediated iodide uptake plays an important physiological role in regulating thyroid gland function, as well as in diagnosing and treating Graves’ disease and thyroid cancer. High-mobility group box 1 (HMGB1), a highly conserved nuclear protein, is a positive regulator of autophagy conferring resistance to chemotherapy, radiotherapy and immunotherapy in cancer cells. Here the authors intended to identify the role of HMGB1 in Hank’s balanced salt solution (HBSS)-induced autophagy, explore NIS protein degradation through a autophagy-lysosome pathway in thyroid cancer cells and elucidate the possible molecular mechanisms. Methods Immunohistochemical staining and reverse transcription-polymerase chain reaction (RT-PCR) were performed for detecting the expression of HMGB1 in different tissues. HMGB1 was knocked down by lentiviral transfection in FTC-133/TPC-1 cells. Autophagic markers LC3-II, p62, Beclin1 and autophagosomal formation were employed for evaluating HMGB1-mediated autophagy in HBSS-treated cells by Western blot, immunofluorescence and electron microscopy. Western blot, quantitative RT-PCR and gamma counter analysis were performed for detecting NIS expression and iodide uptake in HMGB1-knockdown cells after different treatments. The reactive oxygen species (ROS) level, ROS-mediated LC3-II expression and HMGB1 cytosolic translocation were detected by fluorospectrophotometer, flow cytometry, Western blot and immunofluorescence. HMGB1-mediated AMPK, mTOR and p70S6K phosphorylation (p-AMPK, p-mTOR & p-p70S6K) were detected by Western blot. Furthermore, a nude murine model with transplanted tumor was employed for examining the effect of HMGB1-mediated autophagy on imaging and biodistribution of 99mTcO4−. NIS, Beclin1, p-AMPK and p-mTOR were detected by immunohistochemical staining and Western blot in transplanted tumor samples. Results HMGB1 was a critical regulator of autophagy-mediated NIS degradation in HBSS-treated FTC-133/TPC-1 cells. And HMGB1 up-regulation was rather prevalent in thyroid cancer tissues and closely correlated with worse overall lymph node metastasis and clinical stage. HMGB1-knockdown dramatically suppressed autophagy, NIS degradation and boosted iodide uptake in HBSS-treated cells. Moreover, HBSS enhanced ROS-sustained autophagy and promoted the cytosolic translocation of HMGB1. A knockdown of HMGB1 suppressed LC3-II conversion and NIS degradation via an AMPK/mTOR-dependent signal pathway through a regulation of ROS generation, rather than ATP. Furthermore, these data were further supported by our in vivo experiment of xenografts formed by HMGB1 knockdown cells reverting the uptake of 99mTcO4− as compared with control shRNA-transfected cells in hunger group. Conclusions Acting as a critical regulator of autophagy-mediated NIS degradation via ROS/AMPK/mTOR pathway, HMGB1is a potential intervention target of radioiodine therapy in thyroid cancer. Electronic supplementary material The online version of this article (10.1186/s13046-019-1328-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenwen Chai
- Department of Nuclear Medicine, Hunan Cancer Hospital, Changsha, Hunan, 410008, People's Republic of China
| | - Fanghua Ye
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Li Zeng
- Department of Nuclear Medicine, Hunan Cancer Hospital, Changsha, Hunan, 410008, People's Republic of China
| | - Yanling Li
- Department of Nuclear Medicine, Hunan Cancer Hospital, Changsha, Hunan, 410008, People's Republic of China
| | - Liangchun Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
21
|
Wang J, Hallinger DR, Murr AS, Buckalew AR, Lougee RR, Richard AM, Laws SC, Stoker TE. High-throughput screening and chemotype-enrichment analysis of ToxCast phase II chemicals evaluated for human sodium-iodide symporter (NIS) inhibition. ENVIRONMENT INTERNATIONAL 2019; 126:377-386. [PMID: 30826616 PMCID: PMC9082575 DOI: 10.1016/j.envint.2019.02.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/14/2019] [Accepted: 02/07/2019] [Indexed: 05/26/2023]
Abstract
In support of the Endocrine Disruptor Screening Program (EDSP), the U.S.EPA's Office of Research and Development (ORD) is developing high-throughput screening (HTS) approaches to identify chemicals that alter target sites in the thyroid hormone (TH) pathway. The sodium iodide symporter (NIS) is a transmembrane glycoprotein that mediates iodide uptake into the thyroid as the initial step of TH biosynthesis. Previously, we screened 293 ToxCast chemicals (ph1v2) using a HEK293T cell line expressing human NIS in parallel radioactive iodide uptake (RAIU) and cell viability assays to identify potential environmental NIS inhibitors. Here, we expanded NIS inhibitor screening for a set of 768 ToxCast Phase II (ph2) chemicals, and applied a novel computational toxicology approach based on the ToxPrint chemotype to identify chemical substructures associated with NIS inhibition. Following single-concentration screening (at 1 × 10-4 M with a 20% inhibition cutoff), 235 samples (228 chemicals) were further tested in multiple-concentration (1 × 10-9 - 1 × 10-4 M) format in both RAIU and cell viability assays. The 167 chemicals that exhibited significant RAIU inhibition were then prioritized using combined RAIU and cell viability responses that were normalized relative to the known NIS inhibitor sodium perchlorate. Some of the highest ranked chemicals, such as PFOS, tributyltin chloride, and triclocarban, have been previously reported to be thyroid disruptors. In addition, several novel chemicals were identified as potent NIS inhibitors. The present results were combined with the previous ph1v2 screening results to produce two sets of binary hit-calls for 1028 unique chemicals, consisting of 273 positives exhibiting significant RAIU inhibition, and 63 positives following application of a cell viability filter. A ToxPrint chemotype-enrichment analysis identified >20 distinct chemical substructural features, represented in >60% of the active chemicals, as significantly enriched in each NIS inhibition hit-call space. A shared set of 9 chemotypes enriched in both hit-call sets indicates stable chemotype signals (insensitive to cytotoxicity filters) that can help guide structure-activity relationship (SAR) investigations and inform future research.
Collapse
Affiliation(s)
- Jun Wang
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA; Oak Ridge Institute for Science and Education, U.S. Department of Energy, Oak Ridge, TN 37831, USA
| | - Daniel R Hallinger
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Ashley S Murr
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Angela R Buckalew
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Ryan R Lougee
- Oak Ridge Institute for Science and Education, U.S. Department of Energy, Oak Ridge, TN 37831, USA; National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Ann M Richard
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Susan C Laws
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA.
| | - Tammy E Stoker
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA.
| |
Collapse
|
22
|
Pro-tumorigenic non-pump function of sodium iodide symporter: A reimagined Trojan horse? Oncotarget 2019; 10:688-689. [PMID: 30774768 PMCID: PMC6366818 DOI: 10.18632/oncotarget.26596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 01/09/2019] [Indexed: 11/25/2022] Open
|
23
|
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
|
24
|
Martín M, Modenutti CP, Peyret V, Geysels RC, Darrouzet E, Pourcher T, Masini-Repiso AM, Martí MA, Carrasco N, Nicola JP. A Carboxy-Terminal Monoleucine-Based Motif Participates in the Basolateral Targeting of the Na+/I- Symporter. Endocrinology 2019; 160:156-168. [PMID: 30496374 PMCID: PMC6936561 DOI: 10.1210/en.2018-00603] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 11/20/2018] [Indexed: 12/26/2022]
Abstract
The Na+/iodide (I-) symporter (NIS), a glycoprotein expressed at the basolateral plasma membrane of thyroid follicular cells, mediates I- accumulation for thyroid hormonogenesis and radioiodide therapy for differentiated thyroid carcinoma. However, differentiated thyroid tumors often exhibit lower I- transport than normal thyroid tissue (or even undetectable I- transport). Paradoxically, the majority of differentiated thyroid cancers show intracellular NIS expression, suggesting abnormal targeting to the plasma membrane. Therefore, a thorough understanding of the mechanisms that regulate NIS plasma membrane transport would have multiple implications for radioiodide therapy. In this study, we show that the intracellularly facing carboxy-terminus of NIS is required for the transport of the protein to the plasma membrane. Moreover, the carboxy-terminus contains dominant basolateral information. Using internal deletions and site-directed mutagenesis at the carboxy-terminus, we identified a highly conserved monoleucine-based sorting motif that determines NIS basolateral expression. Furthermore, in clathrin adaptor protein (AP)-1B-deficient cells, NIS sorting to the basolateral plasma membrane is compromised, causing the protein to also be expressed at the apical plasma membrane. Computer simulations suggest that the AP-1B subunit σ1 recognizes the monoleucine-based sorting motif in NIS carboxy-terminus. Although the mechanisms by which NIS is intracellularly retained in thyroid cancer remain elusive, our findings may open up avenues for identifying molecular targets that can be used to treat radioiodide-refractory thyroid tumors that express NIS intracellularly.
Collapse
Affiliation(s)
- 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
| | - Carlos Pablo Modenutti
- 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
- Correspondence: Juan Pablo Nicola, PhD, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Centro de Investigaciones en Bioquímica Clínica e Inmunología–Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI–CONICET), Haya de la Torre y Medina Allende, Córdoba X5000HUA, Argentina. E-mail:
| | - 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
| | - 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
| | - Elisabeth Darrouzet
- Laboratoire Transporteurs en Imagerie et Radiothérapie en Oncologie, Faculté de Médecine, Université de Nice Sophia Antipolis–Université Côte d’Azur, Nice, France
- Laboratoire Transporteurs en Imagerie et Radiothérapie en Oncologie, Faculté de Médecine, Commissariat à l’Energie Atomique, Nice, France
| | - Thierry Pourcher
- Laboratoire Transporteurs en Imagerie et Radiothérapie en Oncologie, Faculté de Médecine, Université de Nice Sophia Antipolis–Université Côte d’Azur, Nice, France
- Laboratoire Transporteurs en Imagerie et Radiothérapie en Oncologie, Faculté de Médecine, Commissariat à l’Energie Atomique, Nice, France
| | - 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
| | - 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
| | - Nancy Carrasco
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - 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
|
25
|
Martín M, Geysels RC, Peyret V, Bernal Barquero CE, Masini-Repiso AM, Nicola JP. Implications of Na +/I - Symporter Transport to the Plasma Membrane for Thyroid Hormonogenesis and Radioiodide Therapy. J Endocr Soc 2018; 3:222-234. [PMID: 30620007 PMCID: PMC6316985 DOI: 10.1210/js.2018-00100] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023] Open
Abstract
Iodine is a crucial component of thyroid hormones; therefore, a key requirement for thyroid hormone biosynthesis is that iodide (I−) be actively accumulated in the thyroid follicular cell. The ability of the thyroid epithelia to concentrate I− is ultimately dependent on functional Na+/ I− symporter (NIS) expression at the plasma membrane. Underscoring the significance of NIS for thyroid physiology, loss-of-function mutations in the NIS-coding SLC5A5 gene cause an I− transport defect, resulting in dyshormonogenic congenital hypothyroidism. Moreover, I− accumulation in the thyroid cell constitutes the cornerstone for radioiodide ablation therapy for differentiated thyroid carcinoma. However, differentiated thyroid tumors often exhibit reduced (or even undetectable) I− transport compared with normal thyroid tissue, and they are diagnosed as cold nodules on thyroid scintigraphy. Paradoxically, immunohistochemistry analysis revealed that cold thyroid nodules do not express NIS or express normal, or even higher NIS levels compared with adjacent normal tissue, but NIS is frequently intracellularly retained, suggesting the presence of posttranslational abnormalities in the transport of the protein to the plasma membrane. Ultimately, a thorough comprehension of the mechanisms that regulate NIS transport to the plasma membrane would have multiple implications for radioiodide therapy, opening the possibility to identify new molecular targets to treat radioiodide-refractory thyroid tumors. Therefore, in this review, we discuss the current knowledge regarding posttranslational mechanisms that regulate NIS transport to the plasma membrane under physiological and pathological conditions affecting the thyroid follicular cell, a topic of great interest in the thyroid cancer field.
Collapse
Affiliation(s)
- 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, Córdoba, Argentina
| | - 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, 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, Córdoba, Argentina
| | - 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, Córdoba, Argentina
| | - 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, 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, Córdoba, Argentina
| |
Collapse
|
26
|
Ząbczyńska M, Kozłowska K, Pocheć E. Glycosylation in the Thyroid Gland: Vital Aspects of Glycoprotein Function in Thyrocyte Physiology and Thyroid Disorders. Int J Mol Sci 2018; 19:E2792. [PMID: 30227620 PMCID: PMC6163523 DOI: 10.3390/ijms19092792] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/07/2018] [Accepted: 09/14/2018] [Indexed: 02/08/2023] Open
Abstract
The key proteins responsible for hormone synthesis in the thyroid are glycosylated. Oligosaccharides strongly affect the function of glycosylated proteins. Both thyroid-stimulating hormone (TSH) secreted by the pituitary gland and TSH receptors on the surface of thyrocytes contain N-glycans, which are crucial to their proper activity. Thyroglobulin (Tg), the protein backbone for synthesis of thyroid hormones, is a heavily N-glycosylated protein, containing 20 putative N-glycosylated sites. N-oligosaccharides play a role in Tg transport into the follicular lumen, where thyroid hormones are produced, and into thyrocytes, where hyposialylated Tg is degraded. N-glycans of the cell membrane transporters sodium/iodide symporter and pendrin are necessary for iodide transport. Some changes in glycosylation result in abnormal activity of the thyroid and alteration of the metabolic clearance rate of hormones. Alteration of glycan structures is a pathological process related to the progression of chronic diseases such as thyroid cancers and autoimmunity. Thyroid carcinogenesis is accompanied by changes in sialylation and fucosylation, β1,6-branching of glycans, the content and structure of poly-LacNAc chains, as well as O-GlcNAcylation, while in thyroid autoimmunity the main processes affected are sialylation and fucosylation. The glycobiology of the thyroid gland is an intensively studied field of research, providing new data helpful in understanding the role of the sugar component in thyroid protein biology and disorders.
Collapse
Affiliation(s)
- Marta Ząbczyńska
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| | - Kamila Kozłowska
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| | - Ewa Pocheć
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland.
| |
Collapse
|
27
|
Feng F, Yehia L, Ni Y, Chang YS, Jhiang SM, Eng C. A Nonpump Function of Sodium Iodide Symporter in Thyroid Cancer via Cross-talk with PTEN Signaling. Cancer Res 2018; 78:6121-6133. [PMID: 30217930 DOI: 10.1158/0008-5472.can-18-1954] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/15/2018] [Accepted: 09/11/2018] [Indexed: 01/21/2023]
Abstract
The sodium iodide symporter (NIS) is a classical iodide pump typically localized within the cell plasma membrane in thyroid cells, where NIS expression is believed to ensure success of mainstay radioiodide therapy in thyroid cancers. Although radioiodide uptake is generally reduced in thyroid cancer tissue, intracellular nonmembranous NIS has been reported to increase, suggesting that NIS serves a pump-independent function. Thyroid cancer is one of the major component cancers of Cowden syndrome, a subset of which is caused by germline mutations in PTEN In this study, we explored the noncanonical tumorigenic role of NIS in thyroid cancer cells in relation to PTEN signaling. PTEN knockdown in thyroid cancer cell lines stabilized intracellular NIS protein by promoting an interaction with NIS-LARG (leukemia-associated RhoA guanine exchange factor). Increased protein levels of cytoplasmic NIS enhanced RhoA activation and resulted in a promigration tumorigenic phenotype. Inhibition of NIS glycosylation through activation of the PI3K/AKT/mTOR signaling pathway contributed to mislocalization of NIS in the cytoplasm, facilitating its nonpump tumorigenic function through an interaction with LARG, which predominantly localized in the cytoplasm. Moreover, PTEN or PI3K/AKT/mTOR signaling could affect DPAGT1, a glycosylating enzyme involved in the initial step of N-linked glycosylation, to inhibit glycosylation of NIS. In summary, our results elucidate a pump-independent, protumorigenic role for NIS in thyroid cancer via its cross-talk with PTEN signaling.Significance: A novel pump-independent protumorigenic role of nonmembranous NIS challenges the presumption that radioiodine treatment of thyroid cancer is ineffective when transmembrane NIS is not expressed. Cancer Res; 78(21); 6121-33. ©2018 AACR.
Collapse
Affiliation(s)
- Fang Feng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lamis Yehia
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Ying Ni
- Center for Clinical Genomics, Cleveland Clinic, Cleveland, Ohio
| | - Yi Seok Chang
- Department of Physiology and Cell Biology, and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Sissy Meihua Jhiang
- Department of Physiology and Cell Biology, and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio. .,Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Germline High Risk Cancer Focus Group, CASE Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
28
|
Wang J, Hallinger DR, Murr AS, Buckalew AR, Simmons SO, Laws SC, Stoker TE. High-Throughput Screening and Quantitative Chemical Ranking for Sodium-Iodide Symporter Inhibitors in ToxCast Phase I Chemical Library. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:5417-5426. [PMID: 29611697 PMCID: PMC6697091 DOI: 10.1021/acs.est.7b06145] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Thyroid uptake of iodide via the sodium-iodide symporter (NIS) is the first step in the biosynthesis of thyroid hormones that are critical for health and development in humans and wildlife. Despite having long been a known target of endocrine disrupting chemicals such as perchlorate, information regarding NIS inhibition activity is still unavailable for the vast majority of environmental chemicals. This study applied a previously validated high-throughput approach to screen for NIS inhibitors in the ToxCast phase I library, representing 293 important environmental chemicals. Here 310 blinded samples were screened in a tiered-approach using an initial single-concentration (100 μM) radioactive-iodide uptake (RAIU) assay, followed by 169 samples further evaluated in multi-concentration (0.001 μM-100 μM) testing in parallel RAIU and cell viability assays. A novel chemical ranking system that incorporates multi-concentration RAIU and cytotoxicity responses was also developed as a standardized method for chemical prioritization in current and future screenings. Representative chemical responses and thyroid effects of high-ranking chemicals are further discussed. This study significantly expands current knowledge of NIS inhibition potential in environmental chemicals and provides critical support to U.S. EPA's Endocrine Disruptor Screening Program (EDSP) initiative to expand coverage of thyroid molecular targets, as well as the development of thyroid adverse outcome pathways (AOPs).
Collapse
Affiliation(s)
- Jun Wang
- Oak Ridge Institute for Science and Education, U.S. Department of Energy, Oak Ridge, TN 37831, USA
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Daniel R. Hallinger
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Ashley S. Murr
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Angela R. Buckalew
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Steven O. Simmons
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | - Susan C. Laws
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
- Corresponding authors: (Phone: 919-541-0173 Fax: 919-541-5138) and (Phone: 919-541-2783 Fax: 919-541-5138)
| | - Tammy E. Stoker
- Endocrine Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
- Corresponding authors: (Phone: 919-541-0173 Fax: 919-541-5138) and (Phone: 919-541-2783 Fax: 919-541-5138)
| |
Collapse
|
29
|
De la Vieja A, Santisteban P. Role of iodide metabolism in physiology and cancer. Endocr Relat Cancer 2018; 25:R225-R245. [PMID: 29437784 DOI: 10.1530/erc-17-0515] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/28/2022]
Abstract
Iodide (I-) metabolism is crucial for the synthesis of thyroid hormones (THs) in the thyroid and the subsequent action of these hormones in the organism. I- is principally transported by the sodium iodide symporter (NIS) and by the anion exchanger PENDRIN, and recent studies have demonstrated the direct participation of new transporters including anoctamin 1 (ANO1), cystic fibrosis transmembrane conductance regulator (CFTR) and sodium multivitamin transporter (SMVT). Several of these transporters have been found expressed in various tissues, implicating them in I- recycling. New research supports the exciting idea that I- participates as a protective antioxidant and can be oxidized to hypoiodite, a potent oxidant involved in the host defense against microorganisms. This was possibly the original role of I- in biological systems, before the appearance of TH in evolution. I- per se participates in its own regulation, and new evidence indicates that it may be antineoplastic, anti-proliferative and cytotoxic in human cancer. Alterations in the expression of I- transporters are associated with tumor development in a cancer-type-dependent manner and, accordingly, NIS, CFTR and ANO1 have been proposed as tumor markers. Radioactive iodide has been the mainstay adjuvant treatment for thyroid cancer for the last seven decades by virtue of its active transport by NIS. The rapid advancement of techniques that detect radioisotopes, in particular I-, has made NIS a preferred target-specific theranostic agent.
Collapse
Affiliation(s)
- Antonio De la Vieja
- Tumor Endocrine Unit, Chronic Disease Program (UFIEC), Instituto de Salud Carlos III, Madrid, Spain
- CiberOnc, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Santisteban
- CiberOnc, Instituto de Salud Carlos III, Madrid, Spain
- Department of Physiopathology of Endocrine a Nervous System, Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| |
Collapse
|
30
|
Wang ZF, Liu QJ, Liao SQ, Yang R, Ge T, He X, Tian CP, Liu W. Expression and correlation of sodium/iodide symporter and thyroid stimulating hormone receptor in human thyroid carcinoma. TUMORI JOURNAL 2018; 97:540-6. [DOI: 10.1177/030089161109700420] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims To investigate the expression of sodium/iodide symporter (NIS) and thyroid stimulating hormone receptor (TSHR) in human thyroid cancer. Patients and methods NIS and TSHR mRNA levels quantified by real-time PCR as well as NIS and TSHR proteins evaluated by immunohistochemistry were examined in surgical specimens including 38 benign nodules, 32 thyroid carcinomas and 36 normal thyroid samples. Results NIS and TSHR mRNA levels in thyroid carcinomas were significantly lower than in benign nodules and normal thyroid samples (P <0.001). Interestingly, we found that NIS and TSHR mRNA expression in benign nodules had similar levels to those in normal thyroid tissues. However, NIS and TSHR protein expression in benign nodules and thyroid carcinomas was stronger than in normal thyroid samples (P <0.05) but mainly located in cytoplasm. In addition, there was a significant positive correlation between NIS and TSHR in benign nodules and normal thyroid samples (r = 0.551 and 0.667, respectively, P = 0.001 and 0.000, respectively) but there was no such correlation in thyroid carcinomas (r = 0.222, P = 0.376). Conclusions In thyroid carcinomas, NIS and TSHR mRNA levels were lower but the proteins were overexpressed. The NIS protein mainly locates in the cytoplasm, which therefore lacks the ability of transporting and absorbing iodine in patients with thyroid carcinoma. In addition, there was no correlation between NIS and TSHR in thyroid cancer, which may explain why, even after TSH stimulation, 10–20% of these malignant tumors are unable to concentrate enough radioiodine for effective therapy.
Collapse
Affiliation(s)
- Zhi-Feng Wang
- Institute of Cell Biology, College of Life Sciences, Lanzhou University, Lanzhou Gansu
| | - Qin-Jiang Liu
- Department of Head and Neck Surgery, Gansu Tumor Hospital, Lanzhou Gansu, China
| | - Shi-Qi Liao
- Molecular Biology Center, Gansu Tumor Hospital, Lanzhou Gansu, China
| | - Rong Yang
- Department of Pathology, Gansu Tumor Hospital, Lanzhou Gansu, China
| | - Ting Ge
- Molecular Biology Center, Gansu Tumor Hospital, Lanzhou Gansu, China
| | - Xin He
- Department of Pathology, Gansu Tumor Hospital, Lanzhou Gansu, China
| | - Cai-Ping Tian
- Molecular Biology Center, Gansu Tumor Hospital, Lanzhou Gansu, China
| | - Wei Liu
- Department of Pathology, Gansu Tumor Hospital, Lanzhou Gansu, China
| |
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
|
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
|
33
|
Amit M, Na'ara S, Francis D, Matanis W, Zolotov S, Eisenhaber B, Eisenhaber F, Weiler Sagie M, Malkin L, Billan S, Charas T, Gil Z. Post-translational Regulation of Radioactive Iodine Therapy Response in Papillary Thyroid Carcinoma. J Natl Cancer Inst 2017; 109:4108088. [DOI: 10.1093/jnci/djx092] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 04/20/2017] [Indexed: 02/06/2023] Open
|
34
|
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
|
35
|
Gonçalves CFL, de Freitas ML, Ferreira ACF. Flavonoids, Thyroid Iodide Uptake and Thyroid Cancer-A Review. Int J Mol Sci 2017; 18:E1247. [PMID: 28604619 PMCID: PMC5486070 DOI: 10.3390/ijms18061247] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 12/27/2022] Open
Abstract
Thyroid cancer is the most common malignant tumor of the endocrine system and the incidence has been increasing in recent years. In a great part of the differentiated carcinomas, thyrocytes are capable of uptaking iodide. In these cases, the main therapeutic approach includes thyroidectomy followed by ablative therapy with radioiodine. However, in part of the patients, the capacity to concentrate iodide is lost due to down-regulation of the sodium-iodide symporter (NIS), the protein responsible for transporting iodide into the thyrocytes. Thus, therapy with radioiodide becomes ineffective, limiting therapeutic options and reducing the life expectancy of the patient. Excessive ingestion of some flavonoids has been associated with thyroid dysfunction and goiter. Nevertheless, studies have shown that some flavonoids can be beneficial for thyroid cancer, by reducing cell proliferation and increasing cell death, besides increasing NIS mRNA levels and iodide uptake. Recent data show that the flavonoids apingenin and rutin are capable of increasing NIS function and expression in vivo. Herein we review literature data regarding the effect of flavonoids on thyroid cancer, besides the effect of these compounds on the expression and function of the sodium-iodide symporter. We will also discuss the possibility of using flavonoids as adjuvants for therapy of thyroid cancer.
Collapse
Affiliation(s)
- Carlos F L Gonçalves
- Carlos Frederico Lima Gonçalves, Laboratory of Endocrine Physiology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.
| | - Mariana L de Freitas
- Mariana Lopes de Freitas, Laboratory of Endocrine Physiology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.
| | - Andrea C F Ferreira
- Andrea Claudia Freitas Ferreira, Laboratory of Endocrine Physiology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil.
- NUMPEX, Campus Duque de Caxias, Universidade Federal do Rio de Janeiro, Duque de Caxias, 25245-390 Rio de Janeiro, Brazil.
| |
Collapse
|
36
|
Azouzi N, Cailloux J, Cazarin JM, Knauf JA, Cracchiolo J, Al Ghuzlan A, Hartl D, Polak M, Carré A, El Mzibri M, Filali-Maltouf A, Al Bouzidi A, Schlumberger M, Fagin JA, Ameziane-El-Hassani R, Dupuy C. NADPH Oxidase NOX4 Is a Critical Mediator of BRAF V600E-Induced Downregulation of the Sodium/Iodide Symporter in Papillary Thyroid Carcinomas. Antioxid Redox Signal 2017; 26:864-877. [PMID: 27401113 PMCID: PMC5444494 DOI: 10.1089/ars.2015.6616] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIMS The BRAFV600E oncogene, reported in 40%-60% of papillary thyroid cancer (PTC), has an important role in the pathogenesis of PTC. It is associated with the loss of thyroid iodide-metabolizing genes, such as sodium/iodide symporter (NIS), and therefore with radioiodine refractoriness. Inhibition of mitogen-activated protein kinase (MAPK) pathway, constitutively activated by BRAFV600E, is not always efficient in resistant tumors suggesting that other compensatory mechanisms contribute to a BRAFV600E adaptive resistance. Recent studies pointed to a key role of transforming growth factor β (TGF-β) in BRAFV600E-induced effects. The reactive oxygen species (ROS)-generating NADPH oxidase NOX4, which is increased in PTC, has been identified as a new key effector of TGF-β in cancer, suggestive of a potential role in BRAFV600E-induced thyroid tumors. RESULTS Here, using two human BRAFV600E-mutated thyroid cell lines and a rat thyroid cell line expressing BRAFV600E in a conditional manner, we show that NOX4 upregulation is controlled at the transcriptional level by the oncogene via the TGF-β/Smad3 signaling pathway. Importantly, treatment of cells with NOX4-targeted siRNA downregulates BRAFV600E-induced NIS repression. Innovation and Conclusion: Our results establish a link between BRAFV600E and NOX4, which is confirmed by a comparative analysis of NOX4 expression in human (TCGA) and mouse thyroid cancers. Remarkably, analysis of human and murine BRAFV600E-mutated thyroid tumors highlights that the level of NOX4 expression is inversely correlated to thyroid differentiation suggesting that other genes involved in thyroid differentiation in addition to NIS might be silenced by a mechanism controlled by NOX4-derived ROS. This study opens a new opportunity to optimize thyroid cancer therapy. Antioxid. Redox Signal. 26, 864-877.
Collapse
Affiliation(s)
- Naïma Azouzi
- 1 UMR 8200 CNRS , Villejuif, France .,2 Institut Gustave Roussy , Villejuif, France .,3 Université Paris-Saclay , Orsay, France .,4 Unité de Biologie et Recherche Médicale, Centre National de l'Energie , des Sciences et des Techniques Nucléaires, Rabat, Morocco
| | - Jérémy Cailloux
- 1 UMR 8200 CNRS , Villejuif, France .,2 Institut Gustave Roussy , Villejuif, France .,3 Université Paris-Saclay , Orsay, France
| | - Juliana M Cazarin
- 1 UMR 8200 CNRS , Villejuif, France .,2 Institut Gustave Roussy , Villejuif, France .,3 Université Paris-Saclay , Orsay, France .,5 Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro , Rio de Janeiro, Brazil
| | - Jeffrey A Knauf
- 6 Department of Medicine and Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center , New York, New York
| | - Jennifer Cracchiolo
- 6 Department of Medicine and Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center , New York, New York
| | - Abir Al Ghuzlan
- 1 UMR 8200 CNRS , Villejuif, France .,2 Institut Gustave Roussy , Villejuif, France .,3 Université Paris-Saclay , Orsay, France
| | - Dana Hartl
- 2 Institut Gustave Roussy , Villejuif, France
| | - Michel Polak
- 7 INSERM U1016 , Paris, France .,8 Imagine Institute , Paris, France .,9 Pediatric Endocrinology, Gynaecology and Diabetology Unit, Hôpital Universitaire Necker-Enfants Malades , AP-HP, Paris, France .,10 Université Paris Descartes-Sorbonne Paris Cité , Paris, France
| | - Aurore Carré
- 7 INSERM U1016 , Paris, France .,8 Imagine Institute , Paris, France
| | - Mohammed El Mzibri
- 4 Unité de Biologie et Recherche Médicale, Centre National de l'Energie , des Sciences et des Techniques Nucléaires, Rabat, Morocco
| | - Abdelkarim Filali-Maltouf
- 11 Laboratoire de Microbiologie et Biologie Moléculaire, Faculté des Sciences, Université Mohammed V , Rabat, Morocco
| | - Abderrahmane Al Bouzidi
- 12 Equipe de recherche en pathologie tumorale, Faculté de Médecine et de Pharmacie, Université Mohammed V , Rabat, Morocco
| | - Martin Schlumberger
- 1 UMR 8200 CNRS , Villejuif, France .,2 Institut Gustave Roussy , Villejuif, France .,3 Université Paris-Saclay , Orsay, France
| | - James A Fagin
- 6 Department of Medicine and Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center , New York, New York
| | - Rabii Ameziane-El-Hassani
- 1 UMR 8200 CNRS , Villejuif, France .,2 Institut Gustave Roussy , Villejuif, France .,4 Unité de Biologie et Recherche Médicale, Centre National de l'Energie , des Sciences et des Techniques Nucléaires, Rabat, Morocco
| | - Corinne Dupuy
- 1 UMR 8200 CNRS , Villejuif, France .,2 Institut Gustave Roussy , Villejuif, France .,3 Université Paris-Saclay , Orsay, France
| |
Collapse
|
37
|
Samadi R, Shafiei B, Azizi F, Ghasemi A. Radioactive Iodine Therapy and Glucose Tolerance. CELL JOURNAL 2017; 19:184-193. [PMID: 28670511 PMCID: PMC5413587 DOI: 10.22074/cellj.2016.4251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 07/19/2016] [Indexed: 11/09/2022]
Abstract
Radioactive iodine therapy is commonly used as an adjuvant therapy in follicular and
papillary thyroid carcinoma (PTC) and in the treatment of Graves’ disease (GD). The
basis of this therapy is the accumulation of radioactive iodine by the sodium-iodide
symporter (NIS) in the thyroid gland. Expression of NIS by extrathyroidal tissues such
as islets of pancreas has been reported. Radioactive iodine uptake by pancreatic
beta-cells can potentially damage these cells. In this study, we discuss the possible
associations between radioactive iodine and glucose intolerance. Overall, radioactive
iodine uptake by the pancreas may damage beta-cells and predispose patients to
glucose intolerance or type 2 diabetes, particularly in patients exposed to radioactive
iodine therapy following total thyroidectomy. Further studies are needed to clarify and
confirm this association.
Collapse
Affiliation(s)
- Roghaieh Samadi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Shafiei
- Department of Nuclear Medicine, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
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
|
39
|
N-linked glycosylation of N48 is required for equilibrative nucleoside transporter 1 (ENT1) function. Biosci Rep 2016; 36:BSR20160063. [PMID: 27480168 PMCID: PMC5006311 DOI: 10.1042/bsr20160063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 08/01/2016] [Indexed: 11/29/2022] Open
Abstract
Our study confirmed that Asn48 of hENT1 is the only N-glycosylated residue when expressed in HEK293 cells, and loss of the N-glycan resulted in less hENT1 at the plasma membrane, as well as a loss of function and protein–protein self-interaction. Human equilibrative nucleoside transporter 1 (hENT1) transports nucleosides and nucleoside analogue drugs across cellular membranes and is necessary for the uptake of many anti-cancer, anti-parasitic and anti-viral drugs. Previous work, and in silico prediction, suggest that hENT1 is glycosylated at Asn48 in the first extracellular loop of the protein and that glycosylation plays a role in correct localization and function of hENT1. Site-directed mutagenesis of wild-type (wt) hENT1 removed potential glycosylation sites. Constructs (wt 3xFLAG-hENT1, N48Q-3xFLAG-hENT1 or N288Q-3xFLAG-hENT2) were transiently transfected into HEK293 cells and cell lysates were treated with or without peptide–N-glycosidase F (PNGase-F), followed by immunoblotting analysis. Substitution of N48 prevents hENT1 glycosylation, confirming a single N-linked glycosylation site. N48Q-hENT1 protein is found at the plasma membrane in HEK293 cells but at lower levels compared with wt hENT1 based on S-(4-nitrobenzyl)-6-thioinosine (NBTI) binding analysis (wt 3xFLAG-ENT1 Bmax, 41.5±2.9 pmol/mg protein; N48Q-3xFLAG-ENT1 Bmax, 13.5±0.45 pmol/mg protein) and immunofluorescence microscopy. Although present at the membrane, chloroadenosine transport assays suggest that N48Q-hENT1 is non-functional (wt 3xFLAG-ENT1, 170.80±44.01 pmol/mg protein; N48Q-3xFLAG-ENT1, 57.91±17.06 pmol/mg protein; mock-transfected 74.31±19.65 pmol/mg protein). Co-immunoprecipitation analyses suggest that N48Q ENT1 is unable to interact with self or with wt hENT1. Based on these data we propose that glycosylation at N48 is critical for the localization, function and oligomerization of hENT1.
Collapse
|
40
|
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
|
41
|
Mondal S, Raja K, Schweizer U, Mugesh G. Chemie und Biologie der Schilddrüsenhormon-Biosynthese und -Wirkung. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601116] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Santanu Mondal
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore Indien
| | - Karuppusamy Raja
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore Indien
| | - Ulrich Schweizer
- Rheinische Friedrich-Wilhelms-Universität Bonn; Institut für Biochemie und Molekularbiologie; Nussallee 11 53115 Bonn Deutschland
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore Indien
| |
Collapse
|
42
|
Mondal S, Raja K, Schweizer U, Mugesh G. Chemistry and Biology in the Biosynthesis and Action of Thyroid Hormones. Angew Chem Int Ed Engl 2016; 55:7606-30. [DOI: 10.1002/anie.201601116] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Santanu Mondal
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore India
| | - Karuppusamy Raja
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore India
| | - Ulrich Schweizer
- Rheinische Friedrich-Wilhelms-Universität Bonn; Institut für Biochemie und Molekularbiologie; Nussallee 11 53115 Bonn Germany
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore India
| |
Collapse
|
43
|
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
|
44
|
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
|
45
|
Abstract
Breast cancer is the second most common cancer worldwide and the leading cause of cancer death in women, with incidence rates that continue to rise. The heterogeneity of the disease makes breast cancer exceptionally difficult to treat, particularly for those patients with triple-negative disease. To address the therapeutic complexity of these tumours, new strategies for diagnosis and treatment are urgently required. The ability of lactating and malignant breast cells to uptake and transport iodide has led to the hypothesis that radioiodide therapy could be a potentially viable treatment for many breast cancer patients. Understanding how iodide is transported, and the factors regulating the expression and function of the proteins responsible for iodide transport, is critical for translating this hypothesis into reality. This review covers the three known iodide transporters - the sodium iodide symporter, pendrin and the sodium-coupled monocarboxylate transporter - and their role in iodide transport in breast cells, along with efforts to manipulate them to increase the potential for radioiodide therapy as a treatment for breast cancer.
Collapse
Affiliation(s)
- Vikki L Poole
- School of Clinical and Experimental MedicineInstitute of Biomedical Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Christopher J McCabe
- School of Clinical and Experimental MedicineInstitute of Biomedical Research, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
46
|
Glycosylation of solute carriers: mechanisms and functional consequences. Pflugers Arch 2015; 468:159-76. [PMID: 26383868 DOI: 10.1007/s00424-015-1730-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/19/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022]
Abstract
Solute carriers (SLCs) are one of the largest groups of multi-spanning membrane proteins in mammals and include ubiquitously expressed proteins as well as proteins with highly restricted tissue expression. A vast number of studies have addressed the function and organization of SLCs as well as their posttranslational regulation, but only relatively little is known about the role of SLC glycosylation. Glycosylation is one of the most abundant posttranslational modifications of animal proteins and through recent advances in our understanding of protein-glycan interactions, the functional roles of SLC glycosylation are slowly emerging. The purpose of this review is to provide a concise overview of the aspects of glycobiology most relevant to SLCs, to discuss the roles of glycosylation in the regulation and function of SLCs, and to outline the major open questions in this field, which can now be addressed given major technical advances in this and related fields of study in recent years.
Collapse
|
47
|
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
|
48
|
|
49
|
Quick M, Shi L. The sodium/multivitamin transporter: a multipotent system with therapeutic implications. VITAMINS AND HORMONES 2015; 98:63-100. [PMID: 25817866 PMCID: PMC5530880 DOI: 10.1016/bs.vh.2014.12.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The Na(+)/multivitamin transporter (SMVT) is a member of the solute:sodium symporter family that catalyzes the Na(+)-dependent uptake of the structurally diverse water-soluble vitamins pantothenic acid (vitamin B5) and biotin (vitamin H), α-lipoic acid-a vitamin-like substance with strong antioxidant properties-and iodide. The organic substrates of SMVT play central roles in the cellular metabolism and are, therefore, essential for normal human health and development. For example, biotin deficiency leads to growth retardation, dermatological disorders, and neurological disorders. Animal studies have shown that biotin deficiency during pregnancy is directly correlated to embryonic growth retardation, congenital malformation, and death of the embryo. This chapter focuses on the structural and functional features of the human isoform of SMVT (hSMVT); the discovery of which was greatly facilitated by the cloning and expression of hSMVT in tractable expression systems. Special emphasis will be given to mechanistic implications of the transport process of hSMVT that will inform our understanding of the molecular determinants of hSMVT-mediated transport in dynamic context to alleviate the development and optimization of hSMVT as a multipotent platform for drug delivery.
Collapse
Affiliation(s)
- Matthias Quick
- Department of Psychiatry, Division of Molecular Therapeutics, Columbia University College of Physicians and Surgeons, New York State Psychiatric Institute, New York, USA.
| | - Lei Shi
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, USA
| |
Collapse
|
50
|
Fu C, Chen S, Chen R, Fan X, Luo J, Li C, Qian J. Mutation screening of the sodium iodide symporter gene in a cohort of 105 China patients with congenital hypothyroidism. ACTA ACUST UNITED AC 2014; 58:828-32. [PMID: 25465605 DOI: 10.1590/0004-2730000003436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/04/2014] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Dyshormonogenetic congenital hypothyroidism (CH) was reported to be associated with a mutation in the sodium iodide symporter (NIS) gene. The present study was undertaken in the Guangxi Zhuang Autonomous Region of China, to determine the nature and frequency of NIS gene mutations among patients with CH due to dyshormonogenesis. SUBJECTS AND METHODS Blood samples were collected from 105 dyshormonogenetic CH patients in Guangxi Zhuang Autonomous Region, China, and genomic DNA was extracted from peripheral blood leukocytes. All exons of the NIS gene together with their exon-intron boundaries were screened by next-generation sequencing. RESULTS Two silent variations (T221T and T557T) and one missense variation (M435L), as well as two polymorphisms (rs200587561 and rs117626343) were found. CONCLUSIONS Our results indicate that the NIS mutation rate is very low in the Guangxi Zhuang Autonomous Region, China, and it is necessary to study mutations of other genes that have major effects on thyroid dyshormonogenesis and have not as yet been studied in this population.
Collapse
Affiliation(s)
- Chunyun Fu
- Department of Genetic Metabolism, Children's Hospital, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region
| | - Shaoke Chen
- Department of Genetic Metabolism, Children's Hospital, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region
| | - Rongyu Chen
- Department of Genetic Metabolism, Children's Hospital, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region
| | - Xin Fan
- Department of Genetic Metabolism, Children's Hospital, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region
| | - Jingsi Luo
- Department of Genetic Metabolism, Children's Hospital, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region
| | - Chuan Li
- Department of Genetic Metabolism, Children's Hospital, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region
| | - Jiale Qian
- Department of Genetic Metabolism, Children's Hospital, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region
| |
Collapse
|