1
|
Anthofer L, Gmach P, Uretmen Kagiali ZC, Kleinau G, Rotter J, Opitz R, Scheerer P, Beck-Sickinger AG, Wolf P, Biebermann H, Bechmann I, Kühnen P, Krude H, Paisdzior S. Melanocortin-4 Receptor PLC Activation Is Modulated by an Interaction with the Monocarboxylate Transporter 8. Int J Mol Sci 2024; 25:7565. [PMID: 39062808 PMCID: PMC11277258 DOI: 10.3390/ijms25147565] [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: 06/14/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
The melanocortin-4 receptor (MC4R) is a key player in the hypothalamic leptin-melanocortin pathway that regulates satiety and hunger. MC4R belongs to the G protein-coupled receptors (GPCRs), which are known to form heterodimers with other membrane proteins, potentially modulating receptor function or characteristics. Like MC4R, thyroid hormones (TH) are also essential for energy homeostasis control. TH transport across membranes is facilitated by the monocarboxylate transporter 8 (MCT8), which is also known to form heterodimers with GPCRs. Based on the finding in single-cell RNA-sequencing data that both proteins are simultaneously expressed in hypothalamic neurons, we investigated a putative interplay between MC4R and MCT8. We developed a novel staining protocol utilizing a fluorophore-labeled MC4R ligand and demonstrated a co-localization of MC4R and MCT8 in human brain tissue. Using in vitro assays such as BRET, IP1, and cAMP determination, we found that MCT8 modulates MC4R-mediated phospholipase C activation but not cAMP formation via a direct interaction, an effect that does not require a functional MCT8 as it was not altered by a specific MCT8 inhibitor. This suggests an extended functional spectrum of MCT8 as a GPCR signaling modulator and argues for the investigation of further GPCR-protein interactions with hitherto underrepresented physiological functions.
Collapse
Affiliation(s)
- Larissa Anthofer
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- Institute of Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Philipp Gmach
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Zeynep Cansu Uretmen Kagiali
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Gunnar Kleinau
- Group Structural Biology of Cellular Signaling, Institute of Medical Physics and Biophysics, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Jonas Rotter
- Institute of Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Robert Opitz
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Patrick Scheerer
- Group Structural Biology of Cellular Signaling, Institute of Medical Physics and Biophysics, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | | | - Philipp Wolf
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, D-04103 Leipzig, Germany
| | - Heike Biebermann
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Peter Kühnen
- Department for Pediatric Endocrinology and Diabetology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Heiko Krude
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Sarah Paisdzior
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| |
Collapse
|
2
|
Groeneweg S, Zevenbergen C, Lima de Souza EC, van Geest FS, Kloeckener-Gruissem B, Laczko E, Camargo SMR, Meima ME, Peeters RP, Visser WE. Identification of Iodotyrosines as Novel Substrates for the Thyroid Hormone Transporter MCT8. Thyroid 2024; 34:931-941. [PMID: 38661522 DOI: 10.1089/thy.2023.0551] [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: 04/26/2024]
Abstract
Background: Monocarboxylate transporter 8 (MCT8) is the most specific thyroid hormone transporter identified to date, deficiency of which has been associated with severe intellectual and motor disability and abnormal serum thyroid function tests. However, it is presently unknown if MCT8, similar to other thyroid hormone transporters, also accepts additional substrates, and if disruption of their transport may contribute to the observed phenotype. Methods: In this study, we aimed to identify such substrates by applying liquid chromatography-mass spectrometry-based metabolome analysis in lysates of control and MCT8-overexpressing Xenopus oocytes. A subset of identified candidate substrates were validated by direct transport studies in transiently transfected COS-1 cells and human fibroblasts, which endogenously express MCT8. Moreover, transport characteristics were determined, including transport saturation and cis-inhibition potency of thyroid hormone transport. Results: Metabolome analysis identified 21 m/z ratios, corresponding to 87 candidate metabolites, with a 2.0-times differential abundance in MCT8-injected oocytes compared with controls. These metabolites included 3,5-diiodotyrosine (DIT) and several amino acids, including glutamate and glutamine. In accordance, MCT8-expressing COS-1 cells had 2.2-times lower intracellular accumulation of [125I]-DIT compared with control cells. This effect was largely blocked in the presence of 3,3',5-triiodothyronine (T3) (IC50: 2.5 ± 1.5 µM) or thyroxine (T4) (IC50: 5.8 ± 1.3 µM). Conversely, increasing concentrations of DIT enhanced the accumulation of T3 and T4. The MCT8-specific inhibitor silychristin increased the intracellular accumulation of DIT in human fibroblasts. COS-1 cells expressing MCT8 also exhibited a 50% reduction in intracellular accumulation of [125I]-3-monoiodotyrosine (MIT). In contrast, COS-1 cells expressing MCT8 did not alter the intracellular accumulation of [3H]-glutamate or [3H]-glutamine. However, studies in human fibroblasts showed a 1.5-1.9 times higher glutamate uptake in control fibroblasts compared with fibroblasts derived from patients with MCT8 deficiency, which was not affected in the presence of silychristin. Conclusions: Taken together, our results suggest that the iodotyrosines DIT and MIT can be exported by MCT8. MIT and DIT interfere with MCT8-mediated transport of thyroid hormone in vitro and vice versa. Future studies should elucidate if MCT8, being highly expressed in thyroidal follicular cells, also transports iodotyrosines in vivo.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Chantal Zevenbergen
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Elaine C Lima de Souza
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ferdy S van Geest
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Barbara Kloeckener-Gruissem
- Institute of Medical Molecular Genetics, University of Zurich, Schlieren, Switzerland
- Department of Biology, ETHZ, Zurich, Switzerland
| | - Endre Laczko
- Functional Genomics Center, University and ETH Zurich, Zurich, Switzerland
| | - Simone M R Camargo
- Institute of Physiology, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Marcel E Meima
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Mégier C, Dumery G, Luton D. Iodine and Thyroid Maternal and Fetal Metabolism during Pregnancy. Metabolites 2023; 13:metabo13050633. [PMID: 37233673 DOI: 10.3390/metabo13050633] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Thyroid hormones and iodine are required to increase basal metabolic rate and to regulate protein synthesis, long bone growth and neuronal maturation. They are also essential for protein, fat and carbohydrate metabolism regulation. Imbalances in thyroid and iodine metabolism can negatively affect these vital functions. Pregnant women are at risk of hypo or hyperthyroidism, in relation to or regardless of their medical history, with potential dramatic outcomes. Fetal development highly relies on thyroid and iodine metabolism and can be compromised if they malfunction. As the interface between the fetus and the mother, the placenta plays a crucial role in thyroid and iodine metabolism during pregnancy. This narrative review aims to provide an update on current knowledge of thyroid and iodine metabolism in normal and pathological pregnancies. After a brief description of general thyroid and iodine metabolism, their main modifications during normal pregnancies and the placental molecular actors are described. We then discuss the most frequent pathologies to illustrate the upmost importance of iodine and thyroid for both the mother and the fetus.
Collapse
Affiliation(s)
- Charles Mégier
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| | - Grégoire Dumery
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| | - Dominique Luton
- Assistance Publique-Hôpitaux de Paris, Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicetre, France
| |
Collapse
|
4
|
Shybeka I, Maynard JRJ, Saidjalolov S, Moreau D, Sakai N, Matile S. Dynamic Covalent Michael Acceptors to Penetrate Cells: Thiol-Mediated Uptake with Tetrel-Centered Exchange Cascades, Assisted by Halogen-Bonding Switches. Angew Chem Int Ed Engl 2022; 61:e202213433. [PMID: 36272154 PMCID: PMC10098706 DOI: 10.1002/anie.202213433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Indexed: 11/18/2022]
Abstract
Chalcogen-centered cascade exchange chemistry is increasingly understood to account for thiol-mediated uptake, that is, the ability of reversibly thiol-reactive agents to penetrate cells. Here, reversible Michael acceptors are shown to enable and inhibit thiol-mediated uptake, including the cytosolic delivery of proteins. Dynamic cyano-cinnamate dimers rival the best chalcogen-centered inhibitors. Patterns generated in inhibition heatmaps reveal contributions from halogen-bonding switches that occur independent from the thyroid transporter MCT8. The uniqueness of these patterns supports that the entry of tetrel-centered exchangers into cells differs from chalcogen-centered systems. These results expand the chemical space of thiol-mediated uptake and support the existence of a universal exchange network to bring matter into cells, abiding to be decoded for drug delivery and drug discovery in the broadest sense.
Collapse
Affiliation(s)
- Inga Shybeka
- School of Chemistry and BiochemistryNational Centre of Competence in Research (NCCR) Chemical BiologyUniversity of GenevaGenevaSwitzerland
| | - John R. J. Maynard
- School of Chemistry and BiochemistryNational Centre of Competence in Research (NCCR) Chemical BiologyUniversity of GenevaGenevaSwitzerland
| | - Saidbakhrom Saidjalolov
- School of Chemistry and BiochemistryNational Centre of Competence in Research (NCCR) Chemical BiologyUniversity of GenevaGenevaSwitzerland
| | - Dimitri Moreau
- School of Chemistry and BiochemistryNational Centre of Competence in Research (NCCR) Chemical BiologyUniversity of GenevaGenevaSwitzerland
| | - Naomi Sakai
- School of Chemistry and BiochemistryNational Centre of Competence in Research (NCCR) Chemical BiologyUniversity of GenevaGenevaSwitzerland
| | - Stefan Matile
- School of Chemistry and BiochemistryNational Centre of Competence in Research (NCCR) Chemical BiologyUniversity of GenevaGenevaSwitzerland
| |
Collapse
|
5
|
Huttunen J, Adla SK, Markowicz-Piasecka M, Huttunen KM. Increased/Targeted Brain (Pro)Drug Delivery via Utilization of Solute Carriers (SLCs). Pharmaceutics 2022; 14:pharmaceutics14061234. [PMID: 35745806 PMCID: PMC9228667 DOI: 10.3390/pharmaceutics14061234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Membrane transporters have a crucial role in compounds’ brain drug delivery. They allow not only the penetration of a wide variety of different compounds to cross the endothelial cells of the blood–brain barrier (BBB), but also the accumulation of them into the brain parenchymal cells. Solute carriers (SLCs), with nearly 500 family members, are the largest group of membrane transporters. Unfortunately, not all SLCs are fully characterized and used in rational drug design. However, if the structural features for transporter interactions (binding and translocation) are known, a prodrug approach can be utilized to temporarily change the pharmacokinetics and brain delivery properties of almost any compound. In this review, main transporter subtypes that are participating in brain drug disposition or have been used to improve brain drug delivery across the BBB via the prodrug approach, are introduced. Moreover, the ability of selected transporters to be utilized in intrabrain drug delivery is discussed. Thus, this comprehensive review will give insights into the methods, such as computational drug design, that should be utilized more effectively to understand the detailed transport mechanisms. Moreover, factors, such as transporter expression modulation pathways in diseases that should be taken into account in rational (pro)drug development, are considered to achieve successful clinical applications in the future.
Collapse
Affiliation(s)
- Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
| | - Santosh Kumar Adla
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
- Institute of Organic Chemistry and Biochemistry (IOCB), Czech Academy of Sciences, Flemingovo Namesti 542/2, 160 00 Prague, Czech Republic
| | - Magdalena Markowicz-Piasecka
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
- Correspondence:
| |
Collapse
|
6
|
Groeneweg S, van Geest FS, Chen Z, Farina S, van Heerebeek REA, Meima ME, Peeters RP, Heuer H, Medici M, Visser WE. Functional Characterization of the Novel and Specific Thyroid Hormone Transporter SLC17A4. Thyroid 2022; 32:326-335. [PMID: 34937426 DOI: 10.1089/thy.2021.0257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background: A recent genome-wide association study identified the SLC17A4 locus associated with circulating free thyroxine (T4) concentrations. Human SLC17A4, being widely expressed in the gastrointestinal tract, was characterized as a novel triiodothyronine (T3) and T4 transporter. However, apart from the cellular uptake of T3 and T4, transporter characteristics are currently unknown. In this study, we delineated basic transporter characteristics of this novel thyroid hormone (TH) transporter. Methods: We performed a broad range of well-established TH transport studies in COS-1 cells transiently overexpressing SLC17A4. We studied cellular TH uptake in various incubation buffers, TH efflux, and the inhibitory effects of different TH metabolites and known inhibitors of other TH transporters on SLC17A4-mediated TH transport. Finally, we determined the effect of tunicamycin, a pharmacological inhibitor of N-linked glycosylation, and targeted mutations in Asn residues on SLC17A4 function. Results: SLC17A4 induced the cellular uptake of T3 and T4 by ∼4 times, and of reverse (r)T3 by 1.5 times over control cells. The uptake of T4 by SLC17A4 was Na+ and Cl- independent, stimulated by low extracellular pH, and reduced by various iodothyronines and metabolites thereof, particularly those that contain at least three iodine moieties irrespective of the presence of modification at the alanine side chain. None of the classical TH transporter inhibitors studied attenuated SLC17A4-mediated TH transport. SLC17A4 also facilitates the efflux of T3 and T4, and to a lesser extent of 3,3'-diiodothyronine (T2). Immunoblot studies on lysates of transfected cells cultured in absence or presence of tunicamycin indicated that SLC17A4 is subject to N-linked glycosylation. Complementary mutational studies identified Asn66, Asn75, and Asn90, which are located in extracellular loop 1, as primary targets. Conclusions: Our studies show that SLC17A4 facilitates the transport of T3 and T4, and less efficiently rT3 and 3,3'-T2. Further studies should reveal the physiological role of SLC17A4 in TH regulation.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ferdy S van Geest
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Zhongli Chen
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Stefania Farina
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ramona E A van Heerebeek
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marcel E Meima
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robin P Peeters
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Duisburg-Essen, Essen, Germany
| | - Marco Medici
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W Edward Visser
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
7
|
van Geest FS, Meima ME, Stuurman KE, Wolf NI, van der Knaap MS, Lorea CF, Poswar FO, Vairo F, Brunetti-Pierri N, Cappuccio G, Bakhtiani P, de Munnik SA, Peeters RP, Visser WE, Groeneweg S. Clinical and Functional Consequences of C-Terminal Variants in MCT8: A Case Series. J Clin Endocrinol Metab 2021; 106:539-553. [PMID: 33141165 PMCID: PMC7823235 DOI: 10.1210/clinem/dgaa795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Indexed: 12/17/2022]
Abstract
CONTEXT Genetic variants in SLC16A2, encoding the thyroid hormone transporter MCT8, can cause intellectual and motor disability and abnormal serum thyroid function tests, known as MCT8 deficiency. The C-terminal domain of MCT8 is poorly conserved, which complicates prediction of the deleteriousness of variants in this region. We studied the functional consequences of 5 novel variants within this domain and their relation to the clinical phenotypes. METHODS We enrolled male subjects with intellectual disability in whom genetic variants were identified in exon 6 of SLC16A2. The impact of identified variants was evaluated in transiently transfected cell lines and patient-derived fibroblasts. RESULTS Seven individuals from 5 families harbored potentially deleterious variants affecting the C-terminal domain of MCT8. Two boys with clinical features considered atypical for MCT8 deficiency had a missense variant [c.1724A>G;p.(His575Arg) or c.1796A>G;p.(Asn599Ser)] that did not affect MCT8 function in transfected cells or patient-derived fibroblasts, challenging a causal relationship. Two brothers with classical MCT8 deficiency had a truncating c.1695delT;p.(Val566*) variant that completely inactivated MCT8 in vitro. The 3 other boys had relatively less-severe clinical features and harbored frameshift variants that elongate the MCT8 protein [c.1805delT;p.(Leu602HisfsTer680) and c.del1826-1835;p.(Pro609GlnfsTer676)] and retained ~50% residual activity. Additional truncating variants within transmembrane domain 12 were fully inactivating, whereas those within the intracellular C-terminal tail were tolerated. CONCLUSIONS Variants affecting the intracellular C-terminal tail of MCT8 are likely benign unless they cause frameshifts that elongate the MCT8 protein. These findings provide clinical guidance in the assessment of the pathogenicity of variants within the C-terminal domain of MCT8.
Collapse
Affiliation(s)
- Ferdy S van Geest
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - Marcel E Meima
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - Kyra E Stuurman
- Department of Clinical Genetics, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - Nicole I Wolf
- Department of Pediatric Neurology, Emma Children’s Hospital, Amsterdam University Medical Centre, AZ Amsterdam, Netherlands
- Amsterdam Neuroscience, HV Amsterdam, Netherlands
| | - Marjo S van der Knaap
- Department of Pediatric Neurology, Emma Children’s Hospital, Amsterdam University Medical Centre, AZ Amsterdam, Netherlands
- Amsterdam Neuroscience, HV Amsterdam, Netherlands
| | - Cláudia F Lorea
- Teaching Hospital of Universidade Federal de Pelotas, Brazil
| | - Fabiano O Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Filippo Vairo
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Gerarda Cappuccio
- Department of Translational Medicine, Federico II University, Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | | | - Sonja A de Munnik
- Department of Human Genetics, Radboud University Medical Centre Nijmegen, GA Nijmegen, the Netherlands
| | - Robin P Peeters
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - W Edward Visser
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - Stefan Groeneweg
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| |
Collapse
|
8
|
Bosshart PD, Charles RP, Garibsingh RAA, Schlessinger A, Fotiadis D. SLC16 Family: From Atomic Structure to Human Disease. Trends Biochem Sci 2020; 46:28-40. [PMID: 32828650 DOI: 10.1016/j.tibs.2020.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/30/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022]
Abstract
The solute carrier 16 (SLC16) family represents a diverse group of membrane proteins mediating the transport of monocarboxylates across biological membranes. Family members show a variety of functional roles ranging from nutrient transport and intracellular pH regulation to thyroid hormone homeostasis. Changes in the expression levels and transport function of certain SLC16 transporters are manifested in severe health disorders including cancer, diabetes, and neurological disorders. L-Lactate-transporting SLC16 family members play essential roles in the metabolism of certain tumors and became validated drug targets. This review illuminates the SLC16 family under a new light using structural information obtained from a SLC16 homolog. Furthermore, the role of these transporters in cancer metabolism and how their inhibition can contribute to anticancer therapy are discussed.
Collapse
Affiliation(s)
- Patrick D Bosshart
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, CH-3012 Bern, Switzerland
| | - Roch-Philippe Charles
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, CH-3012 Bern, Switzerland
| | - Rachel-Ann A Garibsingh
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dimitrios Fotiadis
- Institute of Biochemistry and Molecular Medicine and Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, CH-3012 Bern, Switzerland.
| |
Collapse
|
9
|
Groeneweg S, van den Berge A, Lima de Souza EC, Meima ME, Peeters RP, Visser WE. Insights Into the Mechanism of MCT8 Oligomerization. J Endocr Soc 2020; 4:bvaa080. [PMID: 32724870 PMCID: PMC7375341 DOI: 10.1210/jendso/bvaa080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/12/2020] [Indexed: 12/03/2022] Open
Abstract
Mutations in the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) result in MCT8 deficiency, characterized by severe intellectual and motor disability. The MCT8 protein is predicted to have 12 transmembrane domains (TMDs) and is expressed as monomers, homodimers, and homo-oligomers. This study aimed to delineate the mechanism of MCT8 oligomerization. Coimmunoprecipitation studies demonstrated that lithium dodecyl sulfate effectively disrupts MCT8 protein complexes, indicating the involvement of non-covalent interactions. Successive C-terminal truncations of the MCT8 protein altered the oligomerization pattern only if introduced in the N-terminal half of the protein (TMD1-6). The truncation at extracellular loop 1 (E206X) still allowed homodimerization, but completely abrogated homo-oligomerization, whereas both were preserved by the C231X mutant (at TMD2), suggesting that the minimally required oligomerization sites are located proximal of Cys231. However, mutant constructs lacking the intracellular N-terminus or TMD1 and 2 were still capable to form homo-oligomers. Therefore, other domains distal of Cys231 are also likely to be involved in the formation of extensive multidomain interactions. This hypothesis was supported by structural modeling. Despite multiple approaches, MCT8 oligomerization could not be fully abrogated unless a substantial part of the protein was removed, precluding detailed studies into its functional role. Together, our findings suggest that MCT8 oligomerization involves extensive noncovalent interactions between the N-terminal halves of MCT8 proteins. Most mutations identified in patients with MCT8 deficiency have only minor effects on MCT8 oligomerization and, thus, impaired oligomerization does not appear to be an important pathogenic mechanism.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Amanda van den Berge
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Elaine C Lima de Souza
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marcel E Meima
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robin P Peeters
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - W Edward Visser
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
10
|
Groeneweg S, van Geest FS, Peeters RP, Heuer H, Visser WE. Thyroid Hormone Transporters. Endocr Rev 2020; 41:5637505. [PMID: 31754699 DOI: 10.1210/endrev/bnz008] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ferdy S van Geest
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - W Edward Visser
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
11
|
Masnada S, Groenweg S, Saletti V, Chiapparini L, Castellotti B, Salsano E, Visser WE, Tonduti D. Novel mutations in SLC16A2 associated with a less severe phenotype of MCT8 deficiency. Metab Brain Dis 2019; 34:1565-1575. [PMID: 31332729 DOI: 10.1007/s11011-019-00464-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/08/2019] [Indexed: 02/08/2023]
Abstract
Mutations in the thyroid hormone transporter MCT8 cause severe intellectual and motor disability and abnormal serum thyroid function tests, a syndrome known as MCT8 deficiency (or: Allan-Herndon-Dudley syndrome, AHDS). Although the majority of patients are unable to sit or walk independently and do not develop any speech, some are able to walk and talk in simple sentences. Here, we report on two cases with such a less severe clinical phenotype and consequent gross delay in diagnosis. Genetic analyses revealed two novel hemizygous mutations in the SLC16A2 gene resulting in a p.Thr239Pro and a p.Leu543Pro substitution in the MCT8 protein. In vitro studies in transiently transfected COS-1 and JEG-3 cells, and ex vivo studies in patient-derived fibroblasts revealed substantial residual uptake capacity of both mutant proteins (Leu543Pro > Thr239Pro), providing an explanation for the less severe clinical phenotype. Both mutations impair MCT8 protein stability and interfere with proper subcellular trafficking. In one of the patients calcifications were observed in the basal ganglia at the age of 29 years; an abnormal neuroradiological feature at this age that has been linked to untreated (congenital) hypothyroidism and neural cretinism. Our studies extend on previous work by identifying two novel pathogenic mutations in SLC16A2 gene resulting in a mild clinical phenotype.
Collapse
Affiliation(s)
- Silvia Masnada
- Pediatric Neurology Unit, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Stefan Groenweg
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus MC, University Medical Center, CN, Rotterdam, The Netherlands
| | - Veronica Saletti
- Child Neurology Department, IRCCS Foundation C. Besta Neurological Institute, Milan, Italy
| | - Luisa Chiapparini
- Neuroradiology Unit, IRCCS Foundation C. Besta Neurological Institute, Milan, Italy
| | - Barbara Castellotti
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, IRCCS Foundation C. Besta Neurological Institute, Milan, Italy
| | - Ettore Salsano
- Unit of Neurodegenerative and Neurometabolic Rare Diseases, IRCCS Foundation C. Besta Neurological Institute, Milan, Italy
| | - W Edward Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus MC, University Medical Center, CN, Rotterdam, The Netherlands
| | - Davide Tonduti
- Pediatric Neurology Unit, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy.
- Child Neurology Department, IRCCS Foundation C. Besta Neurological Institute, Milan, Italy.
| |
Collapse
|
12
|
Groeneweg S, Kersseboom S, van den Berge A, Dolcetta-Capuzzo A, van Geest FS, van Heerebeek REA, Arjona FJ, Meima ME, Peeters RP, Visser WE, Visser TJ. In Vitro Characterization of Human, Mouse, and Zebrafish MCT8 Orthologues. Thyroid 2019; 29:1499-1510. [PMID: 31436139 DOI: 10.1089/thy.2019.0009] [Citation(s) in RCA: 6] [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] [Indexed: 12/12/2022]
Abstract
Background: Mutations in the thyroid hormone (TH) transporter monocarboxylate transporter 8 (MCT8) cause MCT8 deficiency, characterized by severe intellectual and motor disability and abnormal serum thyroid function tests. Various Mct8 knock-out mouse models as well as mct8 knock-out and knockdown zebrafish models are used as a disease model for MCT8 deficiency. Although important for model eligibility, little is known about the functional characteristics of the MCT8 orthologues in these species. Therefore, we here compared the functional characteristics of mouse (mm) MCT8 and zebrafish (dr) Mct8 to human (hs) MCT8. Methods: We performed extensive transport studies in COS-1 and JEG-3 cells transiently transfected with hsMCT8, drMct8, and mmMCT8. Protein expression levels and subcellular localization were assessed by immunoblotting, surface biotinylation, and immunocytochemistry. Sequence alignment and structural modeling were used to interpret functional differences between the orthologues. Results: hsMCT8, drMct8, and mmMCT8 all facilitated the uptake and efflux of 3,3'-diiodothyronine (3,3'-T2), rT3, triiodothyronine (T3), and thyroxine (T4), although the initial uptake rates of drMct8 were 1.5-4.0-fold higher than for hsMCT8 and mmMCT8. drMct8 exhibited 3-50-fold lower apparent IC50 values than hsMCT8 and mmMCT8 for all tested substrates, and substrate preference of drMct8 (3,3'-T2, T3 > T4 > rT3) differed from hsMCT8 and mmMCT8 (T3 > T4 > rT3, 3,3'-T2). Compared with hsMCT8 and mmMCT8, cis-inhibition studies showed that T3 uptake by drMct8 was inhibited at a lower concentration and by a broader spectrum of TH metabolites. Total and cell surface expression levels of drMct8 and hsMCT8 were equal and both significantly exceeded those of mmMCT8. Structural modeling located most non-conserved residues outside the substrate pore, except for H192 in hsMCT8, which is replaced by a glutamine in drMct8. However, a H192Q substituent of hsMCT8 did not alter its transporter characteristics. Conclusion: Our studies substantiate the eligibility of mice and zebrafish models for human MCT8 deficiency. However, differences in the intrinsic transporter properties of MCT8 orthologues may exist, which should be realized when comparing MCT8 deficiency in different in vivo models. Moreover, our findings may indicate that the protein domains outside the substrate channel may play a role in substrate selection and protein stability.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - Simone Kersseboom
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - Amanda van den Berge
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - Anna Dolcetta-Capuzzo
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
- Department of Endocrinology and Internal Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Ferdy S van Geest
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - Ramona E A van Heerebeek
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - Francisco J Arjona
- Department of Animal Ecology and Physiology, Institute for Water and Wetland Research, Faculty of Science, Radboud University Nijmegen, Nijmegen, The Netherlands
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel E Meima
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - Theo J Visser
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| |
Collapse
|
13
|
Braun D, Reuter U, Schweizer U. Modeling the Biochemical Phenotype of MCT8 Mutations In Vitro: Resolving a Troubling Inconsistency. Endocrinology 2019; 160:1536-1546. [PMID: 31127274 DOI: 10.1210/en.2019-00069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/19/2019] [Indexed: 02/07/2023]
Abstract
Allan-Herndon-Dudley syndrome (AHDS) is a severe genetic disease caused by mutations in the monocarboxylate transporter 8 (MCT8) gene. MCT8 mediates transport of thyroid hormones in and out of cells, which is thought to play a pivotal role for embryonic and postnatal development of the human brain. Disconcertingly, MCT8R271H leads to a severe form of AHDS but shows residual transport activity when expressed in several types of cultured cells. Here we try to determine the mechanism behind the transport function of MCT8R271H found in overexpressing cell systems. Mutations of Arg271 were introduced into human MCT8 and stably transfected into Madin-Darby canine kidney cells and the human-derived cell line JEG1. Radioactive thyroid hormone-uptake experiments were performed to analyze the pH-dependent effect of the mutation on transport activity. Arg271His transports thyroid hormones in and out of cells in a pH-dependent manner. Its transport activity increases below pH 7.3 and is clearly diminished at physiological pH. The Michaelis constant of the mutant is unaltered, whereas the maximum velocity is reduced. The expression of Arg271His in JEG1 cells leads to an almost nonfunctional transporter at physiological pH replicating the human phenotype for this mutant in vitro and demonstrates, again, that mutant MCT8 activity depends on cellular background. The protonation of His271 at acidic pH restores activity of the mutant protein, which is not active in its deprotonated form at physiological pH. Thus, experimental parameters must be controlled carefully when modeling MCT8 deficiency in cells.
Collapse
Affiliation(s)
- Doreen Braun
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Uschi Reuter
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| |
Collapse
|
14
|
Islam MS, Namba N, Ohata Y, Fujiwara M, Nakano C, Takeyari S, Miyata K, Nakano Y, Yamamoto K, Nakayama H, Kitaoka T, Kubota T, Ozono K. Functional analysis of monocarboxylate transporter 8 mutations in Japanese Allan-Herndon-Dudley syndrome patients. Endocr J 2019; 66:19-29. [PMID: 30369548 DOI: 10.1507/endocrj.ej18-0251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Monocarboxylate transporter 8 (MCT8) facilitates T3 uptake into cells. Mutations in MCT8 lead to Allan-Herndon-Dudley syndrome (AHDS), which is characterized by severe psychomotor retardation and abnormal thyroid hormone profile. Nine uncharacterized MCT8 mutations in Japanese patients with severe neurocognitive impairment and elevated serum T3 levels were studied regarding the transport of T3. Human MCT8 (hMCT8) function was studied in wild-type (WT) or mutant hMCT8-transfected human placental choriocarcinoma cells (JEG3) by visualizing the locations of the proteins in the cells, detecting specific proteins, and measuring T3 uptake. We identified 6 missense (p.Arg445Ser, p.Asp498Asn, p.Gly276Arg, p.Gly196Glu, p.Gly401Arg, and p.Gly312Arg), 2 frameshift (p.Arg355Profs*64 and p.Tyr550Serfs*17), and 1 deletion (p.Pro561del) mutation(s) in the hMCT8 gene. All patients exhibited clinical characteristics of AHDS with high free T3, low-normal free T4, and normal-elevated TSH levels. All tested mutants were expressed at the protein level, except p.Arg355Profs*64 and p.Tyr550Serfs*17, which were truncated, and were inactive in T3 uptake, excluding p.Arg445Ser and p.Pro561del mutants, compared with WT-hMCT8. Immunocytochemistry revealed plasma membrane localization of p.Arg445Ser and p.Pro561del mutants similar with WT-hMCT8. The other mutants failed to localize in significant amount(s) in the plasma membrane and instead localized in the cytoplasm. These data indicate that p.Arg445Ser and p.Pro561del mutants preserve residual function, whereas p.Asp498Asn, p.Gly276Arg, p.Gly196Glu, p.Gly401Arg, p.Gly312Arg, p.Arg355Profs*64, and p.Tyr550Serfs*17 mutants lack function. These findings suggest that the mutations in MCT8 cause loss of function by reducing protein expression, impairing trafficking of protein to plasma membrane, and disrupting substrate channel.
Collapse
Affiliation(s)
- Mohammad Saiful Islam
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Noriyuki Namba
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization, Osaka, Japan
| | - Yasuhisa Ohata
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
- The First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Makoto Fujiwara
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Chiho Nakano
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
- The First Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Shinji Takeyari
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kei Miyata
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yukako Nakano
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kenichi Yamamoto
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Statistical Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hirofumi Nakayama
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
- The Japan Environment and Children's Study, Osaka Unit Center, Suita, Japan
| | - Taichi Kitaoka
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takuo Kubota
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
15
|
Strømme P, Groeneweg S, Lima de Souza EC, Zevenbergen C, Torgersbråten A, Holmgren A, Gurcan E, Meima ME, Peeters RP, Visser WE, Høneren Johansson L, Babovic A, Zetterberg H, Heuer H, Frengen E, Misceo D, Visser TJ. Mutated Thyroid Hormone Transporter OATP1C1 Associates with Severe Brain Hypometabolism and Juvenile Neurodegeneration. Thyroid 2018; 28:1406-1415. [PMID: 30296914 DOI: 10.1089/thy.2018.0595] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Thyroid hormones (TH) are essential for brain development and function. The TH transporters monocarboxylate transporter 8 (MCT8) and organic anion transporter1 C1 (OATP1C1) facilitate the transport of TH across the blood-brain barrier and into glia and neuronal cells in the brain. Loss of MCT8 function causes Allan-Herndon-Dudley syndrome (AHDS, OMIM 300523) characterized by severe intellectual and motor disability due to cerebral hypothyroidism. Here, the first patient with loss of OATP1C1 function is described. The patient is a 15.5-year-old girl with normal development in the first year of life, who gradually developed dementia with spasticity and intolerance to cold. Brain imaging demonstrated gray and white matter degeneration and severe glucose hypometabolism. METHODS Exome sequencing of the patient and parents was performed to identify the disease-causing mutation, and the effect of the mutation was studied through a panel of in vitro experiments, including thyroxine uptake studies, immunoblotting, and immunocytochemistry. Furthermore, the clinical effects of treatment with the triiodothyronine analogue triiodothyroacetic acid (Triac) are described. RESULTS Exome sequencing identified a homozygous missense mutation in OATP1C1, changing the highly conserved aspartic acid 252 to asparagine (D252N). In vitro, the mutated OATP1C1 displays impaired plasma membrane localization and decreased cellular thyroxine uptake. After treatment with Triac, the clinical condition improved in several domains. CONCLUSIONS This is the first report of human OATP1C1 deficiency compatible with brain-specific hypothyroidism and neurodegeneration.
Collapse
Affiliation(s)
- Petter Strømme
- 1 Division of Pediatric and Adolescent Medicine; Oslo University Hospital , Oslo, Norway
- 2 Faculty of Medicine, University of Oslo , Oslo, Norway
| | - Stefan Groeneweg
- 3 Erasmus Medical Center, Department of Internal Medicine, Academic Center for Thyroid Diseases , Rotterdam, The Netherlands
| | - Elaine C Lima de Souza
- 3 Erasmus Medical Center, Department of Internal Medicine, Academic Center for Thyroid Diseases , Rotterdam, The Netherlands
| | - Chantal Zevenbergen
- 3 Erasmus Medical Center, Department of Internal Medicine, Academic Center for Thyroid Diseases , Rotterdam, The Netherlands
| | - Anette Torgersbråten
- 4 Department of Medical Genetics, Oslo University Hospital and University of Oslo , Oslo, Norway
| | - Asbjørn Holmgren
- 4 Department of Medical Genetics, Oslo University Hospital and University of Oslo , Oslo, Norway
| | - Ebrar Gurcan
- 3 Erasmus Medical Center, Department of Internal Medicine, Academic Center for Thyroid Diseases , Rotterdam, The Netherlands
| | - Marcel E Meima
- 3 Erasmus Medical Center, Department of Internal Medicine, Academic Center for Thyroid Diseases , Rotterdam, The Netherlands
| | - Robin P Peeters
- 3 Erasmus Medical Center, Department of Internal Medicine, Academic Center for Thyroid Diseases , Rotterdam, The Netherlands
| | - W Edward Visser
- 3 Erasmus Medical Center, Department of Internal Medicine, Academic Center for Thyroid Diseases , Rotterdam, The Netherlands
| | | | - Almira Babovic
- 5 Department of Nuclear Medicine; Oslo University Hospital , Oslo, Norway
| | - Henrik Zetterberg
- 6 Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital , Mölndal, Sweden
- 7 Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg , Mölndal, Sweden
- 8 Department of Molecular Neuroscience, UCL Institute of Neurology , Queen Square, London, United Kingdom
- 9 UK Dementia Research Institute at UCL , London, United Kingdom
| | - Heike Heuer
- 10 Department of Endocrinology, University of Duisburg-Essen , Essen, Germany
| | - Eirik Frengen
- 4 Department of Medical Genetics, Oslo University Hospital and University of Oslo , Oslo, Norway
| | - Doriana Misceo
- 4 Department of Medical Genetics, Oslo University Hospital and University of Oslo , Oslo, Norway
| | - Theo J Visser
- 3 Erasmus Medical Center, Department of Internal Medicine, Academic Center for Thyroid Diseases , Rotterdam, The Netherlands
| |
Collapse
|
16
|
Genome-wide analyses identify a role for SLC17A4 and AADAT in thyroid hormone regulation. Nat Commun 2018; 9:4455. [PMID: 30367059 PMCID: PMC6203810 DOI: 10.1038/s41467-018-06356-1] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 08/31/2018] [Indexed: 12/20/2022] Open
Abstract
Thyroid dysfunction is an important public health problem, which affects 10% of the general population and increases the risk of cardiovascular morbidity and mortality. Many aspects of thyroid hormone regulation have only partly been elucidated, including its transport, metabolism, and genetic determinants. Here we report a large meta-analysis of genome-wide association studies for thyroid function and dysfunction, testing 8 million genetic variants in up to 72,167 individuals. One-hundred-and-nine independent genetic variants are associated with these traits. A genetic risk score, calculated to assess their combined effects on clinical end points, shows significant associations with increased risk of both overt (Graves’ disease) and subclinical thyroid disease, as well as clinical complications. By functional follow-up on selected signals, we identify a novel thyroid hormone transporter (SLC17A4) and a metabolizing enzyme (AADAT). Together, these results provide new knowledge about thyroid hormone physiology and disease, opening new possibilities for therapeutic targets. Thyroid dysfunction is a common public health problem and associated with cardiovascular co-morbidities. Here, the authors carry out genome-wide meta-analysis for thyroid hormone (TH) levels, hyper- and hypothyroidism and identify SLC17A4 as a TH transporter and AADAT as a TH metabolizing enzyme.
Collapse
|
17
|
Groeneweg S, van den Berge A, Meima ME, Peeters RP, Visser TJ, Visser WE. Effects of Chemical Chaperones on Thyroid Hormone Transport by MCT8 Mutants in Patient-Derived Fibroblasts. Endocrinology 2018; 159:1290-1302. [PMID: 29309566 DOI: 10.1210/en.2017-00846] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/29/2017] [Indexed: 12/26/2022]
Abstract
Mutations in the thyroid hormone (TH) transporter monocarboxylate transporter 8 (MCT8) result in severe intellectual and motor disability. At present, no effective therapy is available to restore TH signaling in MCT8-dependent tissues. Recent in vitro studies in stable overexpression cell models suggested that the function of certain mutant MCT8 proteins, specifically those that affect protein stability and intracellular trafficking (e.g., p.F501del), could be partially recovered by chemical chaperones. However, the effects of chaperones have not been demonstrated in other commonly used models for MCT8 deficiency, including transient overexpression models and patient-derived fibroblasts. Here, we demonstrate that the chemical chaperone 4-phenylbutyric acid (PBA) similarly potentiates the T3 transport function of wild-type and p.F501del mutant MCT8 in transiently transfected COS-1 cells by increasing MCT8 messenger RNA, total protein, and cell surface expression levels. Although PBA also increased the cell surface expression levels of the p.R445L mutant, no functional improvement was observed, which is in line with the proposed important role of Arg445 in substrate translocation. In contrast, PBA showed only minimal effects in ex vivo studies using control or p.F501del patient-derived fibroblasts. Moreover, the MCT8-specific inhibitor silychristin did not change these minimal effects, suggesting that the underlying mechanism is unrelated to the rescue of functional MCT8. Together, these findings indicate that the potency of chaperones to rescue mutant MCT8 function strongly depends on the cellular model and stress the need for further preclinical studies before clinically available chaperones should be considered as a treatment option in patients with MCT8 deficiency.
Collapse
Affiliation(s)
- Stefan Groeneweg
- The Rotterdam Thyroid Center and Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Amanda van den Berge
- The Rotterdam Thyroid Center and Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Marcel E Meima
- The Rotterdam Thyroid Center and Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Robin P Peeters
- The Rotterdam Thyroid Center and Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Theo J Visser
- The Rotterdam Thyroid Center and Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - W Edward Visser
- The Rotterdam Thyroid Center and Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|