1
|
Tan J, Xiao Y, Kong F, Qian J, Zhu A, Yan C. Structural insights into thyroid hormone transporter MCT8. Nat Commun 2025; 16:2958. [PMID: 40140416 PMCID: PMC11947448 DOI: 10.1038/s41467-025-58131-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Thyroid hormones (THs), including T4 (3,5,3',5'-tetraiodo-L-thyronine) and T3 (3,5,3'-triiodo-L-thyronine), play critical roles in regulating tissue development and basal metabolism. Monocarboxylate transporter 8 (MCT8) is a key player in TH transport, known for its high specificity and affinity for THs and its direct association with Allan-Herndon-Dudley syndrome (AHDS) caused by pathogenic mutations. In this study, we present the cryo-EM structures of human MCT8 bound to the substrate T3 or the inhibitor silychristin, both in an outward-open conformation at resolutions of 3.0-3.2 Å. MCT8 forms a homodimer with a lipid molecule positioned at the dimerization interface. The carboxyl group of T3 is recognized by Arg371, while its three iodine atoms interact with distinct hydrophobic cavities. Silychristin is also recognized by Arg371, competing with T3 for binding. Complemented by structure-guided biochemical analyses, our research elucidates the mechanisms of substrate recognition and transport, as well as the mode of action of the inhibitor silychristin. These findings may offer insights for developing targeted therapies for TH-related disorders.
Collapse
Affiliation(s)
- Jiaxin Tan
- Beijing Frontier Research Center for Biological Structure, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Yuan Xiao
- Beijing Frontier Research Center for Biological Structure, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Fang Kong
- Beijing Frontier Research Center for Biological Structure, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Jiawei Qian
- Beijing Frontier Research Center for Biological Structure, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Angqi Zhu
- Beijing Frontier Research Center for Biological Structure, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Chuangye Yan
- Beijing Frontier Research Center for Biological Structure, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
2
|
Groeneweg S, van Geest FS, Martín M, Dias M, Frazer J, Medina-Gomez C, Sterenborg RBTM, Wang H, Dolcetta-Capuzzo A, de Rooij LJ, Teumer A, Abaci A, van den Akker ELT, Ambegaonkar GP, Armour CM, Bacos I, Bakhtiani P, Barca D, Bauer AJ, van den Berg SAA, van den Berge A, Bertini E, van Beynum IM, Brunetti-Pierri N, Brunner D, Cappa M, Cappuccio G, Castellotti B, Castiglioni C, Chatterjee K, Chesover A, Christian P, Coenen-van der Spek J, de Coo IFM, Coutant R, Craiu D, Crock P, DeGoede C, Demir K, Dewey C, Dica A, Dimitri P, Dremmen MHG, Dubey R, Enderli A, Fairchild J, Gallichan J, Garibaldi L, George B, Gevers EF, Greenup E, Hackenberg A, Halász Z, Heinrich B, Hurst AC, Huynh T, Isaza AR, Klosowska A, van der Knoop MM, Konrad D, Koolen DA, Krude H, Kulkarni A, Laemmle A, LaFranchi SH, Lawson-Yuen A, Lebl J, Leeuwenburgh S, Linder-Lucht M, López Martí A, Lorea CF, Lourenço CM, Lunsing RJ, Lyons G, Malikova JK, Mancilla EE, McCormick KL, McGowan A, Mericq V, Lora FM, Moran C, Muller KE, Nicol LE, Oliver-Petit I, Paone L, Paul PG, Polak M, Porta F, Poswar FO, Reinauer C, Rozenkova K, Seckold R, Seven Menevse T, Simm P, Simon A, Singh Y, Spada M, Stals MAM, Stegenga MT, Stoupa A, et alGroeneweg S, van Geest FS, Martín M, Dias M, Frazer J, Medina-Gomez C, Sterenborg RBTM, Wang H, Dolcetta-Capuzzo A, de Rooij LJ, Teumer A, Abaci A, van den Akker ELT, Ambegaonkar GP, Armour CM, Bacos I, Bakhtiani P, Barca D, Bauer AJ, van den Berg SAA, van den Berge A, Bertini E, van Beynum IM, Brunetti-Pierri N, Brunner D, Cappa M, Cappuccio G, Castellotti B, Castiglioni C, Chatterjee K, Chesover A, Christian P, Coenen-van der Spek J, de Coo IFM, Coutant R, Craiu D, Crock P, DeGoede C, Demir K, Dewey C, Dica A, Dimitri P, Dremmen MHG, Dubey R, Enderli A, Fairchild J, Gallichan J, Garibaldi L, George B, Gevers EF, Greenup E, Hackenberg A, Halász Z, Heinrich B, Hurst AC, Huynh T, Isaza AR, Klosowska A, van der Knoop MM, Konrad D, Koolen DA, Krude H, Kulkarni A, Laemmle A, LaFranchi SH, Lawson-Yuen A, Lebl J, Leeuwenburgh S, Linder-Lucht M, López Martí A, Lorea CF, Lourenço CM, Lunsing RJ, Lyons G, Malikova JK, Mancilla EE, McCormick KL, McGowan A, Mericq V, Lora FM, Moran C, Muller KE, Nicol LE, Oliver-Petit I, Paone L, Paul PG, Polak M, Porta F, Poswar FO, Reinauer C, Rozenkova K, Seckold R, Seven Menevse T, Simm P, Simon A, Singh Y, Spada M, Stals MAM, Stegenga MT, Stoupa A, Subramanian GM, Szeifert L, Tonduti D, Turan S, Vanderniet J, van der Walt A, Wémeau JL, van Wermeskerken AM, Wierzba J, de Wit MCY, Wolf NI, Wurm M, Zibordi F, Zung A, Zwaveling-Soonawala N, Rivadeneira F, Meima ME, Marks DS, Nicola JP, Chen CH, Medici M, Visser WE. Mapping variants in thyroid hormone transporter MCT8 to disease severity by genomic, phenotypic, functional, structural and deep learning integration. Nat Commun 2025; 16:2479. [PMID: 40075072 PMCID: PMC11904026 DOI: 10.1038/s41467-025-56628-w] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 01/23/2025] [Indexed: 03/14/2025] Open
Abstract
Predicting and quantifying phenotypic consequences of genetic variants in rare disorders is a major challenge, particularly pertinent for 'actionable' genes such as thyroid hormone transporter MCT8 (encoded by the X-linked SLC16A2 gene), where loss-of-function (LoF) variants cause a rare neurodevelopmental and (treatable) metabolic disorder in males. The combination of deep phenotyping data with functional and computational tests and with outcomes in population cohorts, enabled us to: (i) identify the genetic aetiology of divergent clinical phenotypes of MCT8 deficiency with genotype-phenotype relationships present across survival and 24 out of 32 disease features; (ii) demonstrate a mild phenocopy in ~400,000 individuals with common genetic variants in MCT8; (iii) assess therapeutic effectiveness, which did not differ among LoF-categories; (iv) advance structural insights in normal and mutated MCT8 by delineating seven critical functional domains; (v) create a pathogenicity-severity MCT8 variant classifier that accurately predicted pathogenicity (AUC:0.91) and severity (AUC:0.86) for 8151 variants. Our information-dense mapping provides a generalizable approach to advance multiple dimensions of rare genetic disorders.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Ferdy S van Geest
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Mariano Martín
- Department of Clinical Biochemistry (CIBICI-CONICET), Faculty of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mafalda Dias
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jonathan Frazer
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rosalie B T M Sterenborg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hao Wang
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Anna Dolcetta-Capuzzo
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Linda J de Rooij
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Alexander Teumer
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Ayhan Abaci
- Division of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Erica L T van den Akker
- Department of Paediatrics, Division of Endocrinology, Erasmus Medical Centre -Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Gautam P Ambegaonkar
- Department of Paediatric Neurology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Christine M Armour
- Regional Genetics Program, Children's Hospital of Eastern Ontario and Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Iiuliu Bacos
- Centrul Medical Dr. Bacos Cosma, Timisoara, Romania
| | - Priyanka Bakhtiani
- University of Louisville, Louisville, KY, USA
- Childrens Hospital Los Angeles, Los Angeles, CA, USA
| | - Diana Barca
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest, Romania
| | - Andrew J Bauer
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sjoerd A A van den Berg
- Diagnostic Laboratory for Endocrinology, 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 Centre, Rotterdam, The Netherlands
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesu' Children's Research Hospital IRCCS, Rome, Italy
| | - Ingrid M van Beynum
- Department of Pediatrics, Division of Pediatric Cardiology, Erasmus Medical Centre - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, 80131, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy
| | - Doris Brunner
- Gottfried Preyer's Children Hospital, Vienna, Austria
| | - Marco Cappa
- Research Area for Innovative Therapies in Endocrinopathies, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gerarda Cappuccio
- Department of Translational Medicine, Federico II University, 80131, Naples, Italy
- Neurological Research Institute and Baylor College of Medicine, Houston, TX, USA
| | - Barbara Castellotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudia Castiglioni
- Department of Neurology, Clinica Meds, School of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Krishna Chatterjee
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Alexander Chesover
- Division of Endocrinology, The Hospital for Sick Children and Department of Paediatrics, University of Toronto, Toronto, M5G 1×8, Canada
- Department of Endocrinology, Great Ormond Street Hospital for Children, London, UK
| | - Peter Christian
- East Kent Hospitals University NHS Foundation Trust, Ashford, UK
| | - Jet Coenen-van der Spek
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Irenaeus F M de Coo
- Department of Toxicogenomics, Unit Clinical Genomics, Maastricht University, MHeNs School for Mental Health and Neuroscience, Maastricht, The Netherlands
| | - Regis Coutant
- Department of Pediatric Endocrinology and Diabetology, University Hospital, Angers, France
| | - Dana Craiu
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest, Romania
| | - Patricia Crock
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Christian DeGoede
- Department of Paediatric Neurology, Clinical Research Facility, Lancashire Teaching Hospitals NHS Trust, Lancashire, UK
| | - Korcan Demir
- Division of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Cheyenne Dewey
- Genomics Institute Mary Bridge Children's Hospital, MultiCare Health System, Tacoma, WA, USA
| | - Alice Dica
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest, Romania
| | - Paul Dimitri
- The Department of Oncology and Metabolism, The University of Sheffield, Western Bank, Sheffield, S10, 2TH, UK
| | - Marjolein H G Dremmen
- Division of Paediatric Radiology, Erasmus Medical Centre - Sophia's Children Hospital, Rotterdam, The Netherlands
| | | | - Anina Enderli
- Department of Neuropediatrics, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Jan Fairchild
- Department of Diabetes and Endocrinology, Women's and Children's Hospital, North Adelaide, 5066, South Australia, Australia
| | | | | | - Belinda George
- Department of Endocrinology, St. John's Medical College Hospital, Bengaluru, India
| | - Evelien F Gevers
- Centre for Endocrinology, William Harvey Research institute, Queen Mary University of London, London, UK
| | - Erin Greenup
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Pediatric Endocrinology, Department of Pediatrics, Orlando Health Arnold Palmer Hospital for Children, Orlando, FL, USA
| | - Annette Hackenberg
- Department of Neuropediatrics, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Zita Halász
- Pediatric Center, Semmelweis University Budapest, Budapest, Hungary
| | - Bianka Heinrich
- Department of Neuropediatrics, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Anna C Hurst
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tony Huynh
- Department of Endocrinology & Diabetes, Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - Amber R Isaza
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anna Klosowska
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - David A Koolen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Heiko Krude
- Institute of Experimental Paediatric Endocrinology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Abhishek Kulkarni
- Department of Paediatric Endocrinology, SRCC Children's Hospital, Mumbai, India
| | - Alexander Laemmle
- Institute of Clinical Chemistry and Department of Pediatrics, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Stephen H LaFranchi
- Department of Pediatrics, Doernbecher Children's Hospital, Oregon Health & Sciences University, Portland, OR, USA
| | - Amy Lawson-Yuen
- Genomics Institute Mary Bridge Children's Hospital, MultiCare Health System, Tacoma, WA, USA
- Department of Genetics, Kaiser Permanente Washington, Seattle, WA, USA
| | - Jan Lebl
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Selmar Leeuwenburgh
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Michaela Linder-Lucht
- Division of Neuropediatrics and Muscular Disorders, Department of Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Anna López Martí
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Cláudia F Lorea
- Teaching Hospital of Universidade Federal de Pelotas, Pelotas, Brazil
- Federal University of Rio Grande do Sul, Porto Alegre-RS, Brazil
| | - Charles M Lourenço
- National Reference Center for Rare Diseases, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
- Personalized Medicine area -Special Education Sector at DLE/Grupo Pardini, Rio de Janeiro, Brazil
| | - Roelineke J Lunsing
- Department of Child Neurology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Greta Lyons
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jana Krenek Malikova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Edna E Mancilla
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kenneth L McCormick
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anne McGowan
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Veronica Mericq
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile, Department of Pediatrics, Clinica Las Condes, Santiago, Chile
| | - Felipe Monti Lora
- Pediatric Endocrinology Group, Sabara Children's Hospital, São Paulo, Brazil
| | - Carla Moran
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | | | - Lindsey E Nicol
- Department of Pediatrics, Doernbecher Children's Hospital, Oregon Health & Sciences University, Portland, OR, USA
| | - Isabelle Oliver-Petit
- Department of Paediatric Endocrinology and Genetics, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Laura Paone
- Endocrinology and Diabetology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Praveen G Paul
- Department of Paediatrics, Christian Medical College, Vellore, India
| | - Michel Polak
- Paediatric Endocrinology, Diabetology and Gynaecology, Department, Necker Children's University Hospital, Imagine Institute Affiliate, Université de Paris Cité, Paris, France
| | - Francesco Porta
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Turin, Italy
| | - Fabiano O Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Christina Reinauer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Medical Faculty, Dusseldorf, Germany
| | - Klara Rozenkova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Rowen Seckold
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Tuba Seven Menevse
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Peter Simm
- Royal Children's Hospital/University of Melbourne, Parkville, Australia
| | - Anna Simon
- Department of Paediatrics, Christian Medical College, Vellore, India
| | - Yogen Singh
- Department of Paediatric Cardiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pediatrics, University of California - UC Davis Children's Hospital, Sacramento, CA, USA
| | - Marco Spada
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Turin, Italy
| | - Milou A M Stals
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Merel T Stegenga
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Athanasia Stoupa
- Paediatric Endocrinology, Diabetology and Gynaecology, Department, Necker Children's University Hospital, Imagine Institute Affiliate, Université de Paris Cité, Paris, France
| | - Gopinath M Subramanian
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Lilla Szeifert
- Pediatric Center, Semmelweis University Budapest, Budapest, Hungary
| | - Davide Tonduti
- Child Neurology Unit - C.O.A.L.A. (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Milano, Italy
- Department of Clinical and Biomedical Science, Università degli Studi di Milano, Milano, Italy
| | - Serap Turan
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Joel Vanderniet
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Adri van der Walt
- Private paediatric Neurology practice Dr A van der Walt, Durbanville, South Africa
| | | | | | - Jolanta Wierzba
- Department of Internal and Pediatric Nursing, Institute of Nursing and Midwifery, Medical University of Gdańsk, Gdańsk, Poland
| | - Marie-Claire Y de Wit
- Department of Paediatric Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Michael Wurm
- University Children's Hospital Regensburg (KUNO), University of Regensburg, Campus St. Hedwig, Regensburg, Germany
| | - Federica Zibordi
- Child Neurology Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Amnon Zung
- Pediatric Endocrinology Unit, Kaplan Medical center, Rehovot and the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nitash Zwaveling-Soonawala
- Emma Children's Hospital, Department of Paediatric Endocrinology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marcel E Meima
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Debora S Marks
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Juan P Nicola
- Department of Clinical Biochemistry (CIBICI-CONICET), Faculty of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Chi-Hua Chen
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Marco Medici
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - W Edward Visser
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Giri D, Govindaraj V, Kumar S, Ungati H, Mugesh G. A Highly Selective Fluorescent Probe for Monitoring the Thyroid Hormone Transporter Activity in Mammalian Cells. Chemistry 2024; 30:e202401719. [PMID: 38995511 DOI: 10.1002/chem.202401719] [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: 05/01/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/13/2024]
Abstract
Monocarboxylate transporter 8 (MCT8) is a trans-membrane transporter, which mediates the cellular delivery of thyroid hormones, L-thyroxine (T4) and 3,5,3'-triiodo-L-thyronine (T3). In humans, the MCT8 protein is encoded by the SLC16A2 gene and mutations in the transporter cause a genetic neurological disorder known as Allan-Herndon-Dudley Syndrome (AHDS). MCT8 deficiency leads to impaired transport of thyroid hormones in the brain. Radiolabelled T4 and T3 or LC/MS-MS methods have been used to monitor the thyroid hormone uptake through MCT8. Herein, we developed a fluorescent based assay to monitor the thyroid hormone uptake through MCT8. A dansyl-based fluorescent probe having L-thyroxine moiety is found to be highly selective towards MCT8 in living cells. The high selectivity of the probe towards MCT8 can be attributed to the halogen bond-mediated recognition by the transporter protein. The presence of a free carboxylic acid group is essential for the specificity of the probe towards MCT8. Additionally, the selectivity of the probe for MCT8 is abolished upon esterification of the carboxylic group. Similarly, MCT8 does not recognize the probe when it contains a free amine group.
Collapse
Affiliation(s)
- Debasish Giri
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Vijayakumar Govindaraj
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
- Department of Biochemistry, School of Allied Health Sciences, REVA University, Bangalore, India
| | - Sagar Kumar
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Harinarayana Ungati
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
4
|
Ferro-Gallego P, Vila-Sanjurjo A, Valderrama Pereira AK, Porres Pérez G, Domínguez-Gerpe L. Circular PCR as an efficient and precise umbrella of methods for the generation of circular dsDNA with staggered nicks: Mechanism and types. Biol Methods Protoc 2024; 9:bpae051. [PMID: 39139995 PMCID: PMC11319657 DOI: 10.1093/biomethods/bpae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
Here, we introduce the highly versatile circular polymerase chain reaction (CiPCR) technique, propose a mechanism of action, and describe a number of examples demonstrating the versatility of this technique. CiPCR takes place between two fragments of dsDNA with two homologous regions, as long as one of the fragments carries said regions at its 3'- and 5'-ends. Upon hybridization, elongation by a polymerase occurs from all 3'-ends continuously until a 5'-end is reached, leading to stable circular dsDNA with staggered nicks. When both dsDNA fragments carry the homology at their 3'- and 5'-ends (Type I CiPCR), all four 3'-ends effectively prime amplification of the intervening region and CiPCR products can function as template during the reaction. In contrast, when only one of the two dsDNA fragments carries the homologous regions at its 3'- and 5'-ends and the other carries such regions internally (Type II CiPCR), only two 3'-ends can be amplified and CiPCR products possess no template activity. We demonstrate the applicability of both CiPCR types via well-illustrated experimental examples. CiPCR is well adapted to the quick resolution of most of the molecular cloning challenges faced by the biology/biomedicine laboratory, including the generation of insertions, deletions, and mutations.
Collapse
Affiliation(s)
- Pedro Ferro-Gallego
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Present Address: IBIMA, Instituto de Investigación Biomédica y Plataforma en Nanomedicina, BIONAND, Málaga, 29590, Spain
| | - Antón Vila-Sanjurjo
- Grupo GIBE, Biology Department of the School of Sciences & Interdisciplinary Center for Chemistry and Biology (CICA), Universidade da Coruña (UDC), A Coruña, Spain
| | - Andrea Katherine Valderrama Pereira
- Chemistry Department of the School of Sciences & Interdisciplinary Center for Chemistry and Biology (CICA), Universidade da Coruña (UDC), A Coruña, Spain
| | - Gonzalo Porres Pérez
- Grupo GIBE, Biology Department of the School of Sciences & Interdisciplinary Center for Chemistry and Biology (CICA), Universidade da Coruña (UDC), A Coruña, Spain
| | - Lourdes Domínguez-Gerpe
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| |
Collapse
|
5
|
Wagenaars F, Cenijn P, Scholze M, Frädrich C, Renko K, Köhrle J, Hamers T. Screening for endocrine disrupting chemicals inhibiting monocarboxylate 8 (MCT8) transporter facilitated thyroid hormone transport using a modified nonradioactive assay. Toxicol In Vitro 2024; 96:105770. [PMID: 38151217 DOI: 10.1016/j.tiv.2023.105770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/14/2023] [Accepted: 12/22/2023] [Indexed: 12/29/2023]
Abstract
Early neurodevelopmental processes are strictly dependent on spatial and temporally modulated of thyroid hormone (TH) availability and action. Thyroid hormone transmembrane transporters (THTMT) are critical for regulating the local concentrations of TH, namely thyroxine (T4) and 3,5,3'-tri-iodothyronine (T3), in the brain. Monocarboxylate transporter 8 (MCT8) is one of the most prominent THTMT. Genetically induced deficiencies in expression, function or localization of MCT8 are associated with irreversible and severe neurodevelopmental adversities. Due to the importance of MCT8 in brain development, studies addressing chemical interferences of MCT8 facilitated T3 uptake are a crucial step to identify TH system disrupting chemicals with this specific mode of action. Recently a non-radioactive in vitro assay has been developed to rapidly screen for endocrine disrupting chemicals (EDCs) acting upon MCT8 mediated transport. This study explored the use of an UV-light digestion step as an alternative for the original ammonium persulfate (APS) digestion step. The non-radioactive TH uptake assay, with the incorporated UV-light digestion step of TH, was then used to screen a set of 31 reference chemicals and environmentally relevant substances to detect inhibition of MCT8-depending T3 uptake. This alternative assay identified three novel MCT8 inhibitors: methylmercury, bisphenol-AF and bisphenol-Z and confirmed previously known MCT8 inhibitors.
Collapse
Affiliation(s)
- Fabian Wagenaars
- Amsterdam Institute for Life and Environment (A-Life), Vrije Universiteit Amsterdam (VU), De Boelelaan 1085, 1081, HV, Amsterdam, the Netherlands
| | - Peter Cenijn
- Amsterdam Institute for Life and Environment (A-Life), Vrije Universiteit Amsterdam (VU), De Boelelaan 1085, 1081, HV, Amsterdam, the Netherlands
| | - Martin Scholze
- Brunel University London, Centre for Pollution Research and Policy, College of Health, Medicine and Life Sciences, Kingston Lane, Uxbridge UB8 3PH, UK
| | - Caroline Frädrich
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin Institut für Experimentelle Endokrinologie, Hessische Strasse 3-4, 10115 Berlin, Germany
| | - Kostja Renko
- German Centre for the Protection of Laboratory Animals (Bf3R), Bundesinstitut für Risikobewertung (BfR), Berlin, Germany
| | - Josef Köhrle
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin Institut für Experimentelle Endokrinologie, Hessische Strasse 3-4, 10115 Berlin, Germany
| | - Timo Hamers
- Amsterdam Institute for Life and Environment (A-Life), Vrije Universiteit Amsterdam (VU), De Boelelaan 1085, 1081, HV, Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Hoen E, Goossens FM, Falize K, Mayerl S, van der Spek AH, Boelen A. The Differential Effect of a Shortage of Thyroid Hormone Compared with Knockout of Thyroid Hormone Transporters Mct8 and Mct10 on Murine Macrophage Polarization. Int J Mol Sci 2024; 25:2111. [PMID: 38396788 PMCID: PMC10889717 DOI: 10.3390/ijms25042111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Innate immune cells, including macrophages, are functionally affected by thyroid hormone (TH). Macrophages can undergo phenotypical alterations, shifting between proinflammatory (M1) and immunomodulatory (M2) profiles. Cellular TH concentrations are, among others, determined by TH transporters. To study the effect of TH and TH transporters on macrophage polarization, specific proinflammatory and immunomodulatory markers were analyzed in bone marrow-derived macrophages (BMDMs) depleted of triiodothyronine (T3) and BMDMs with a knockout (KO) of Mct8 and Mct10 and a double KO (dKO) of Mct10/Mct8. Our findings show that T3 is important for M1 polarization, while a lack of T3 stimulates M2 polarization. Mct8 KO BMDMs are unaffected in their T3 responsiveness, but exhibit slight alterations in M2 polarization, while Mct10 KO BMDMs show reduced T3 responsiveness, but unaltered polarization markers. KO of both the Mct8 and Mct10 transporters decreased T3 availability and, contrary to the T3-depleted BMDMs, showed partially increased M1 markers and unaltered M2 markers. These data suggest a role for TH transporters besides transport of TH in BMDMs. This study highlights the complex role of TH transporters in macrophages and provides a new angle on the interaction between the endocrine and immune systems.
Collapse
Affiliation(s)
- Esmée Hoen
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam Gastroenterology Endocrinology & Metabolism (AGEM), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (E.H.); (K.F.)
| | - Franka M. Goossens
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam Gastroenterology Endocrinology & Metabolism (AGEM), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (E.H.); (K.F.)
| | - Kim Falize
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam Gastroenterology Endocrinology & Metabolism (AGEM), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (E.H.); (K.F.)
| | - Steffen Mayerl
- Department of Endocrinology, Diabetes & Metabolism, University Duisburg-Essen, 47057 Essen, Germany
| | - Anne H. van der Spek
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology & Metabolism (AGEM), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Anita Boelen
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam Gastroenterology Endocrinology & Metabolism (AGEM), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (E.H.); (K.F.)
| |
Collapse
|
7
|
Sabatino L, Lapi D, Del Seppia C. Factors and Mechanisms of Thyroid Hormone Activity in the Brain: Possible Role in Recovery and Protection. Biomolecules 2024; 14:198. [PMID: 38397435 PMCID: PMC10886502 DOI: 10.3390/biom14020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Thyroid hormones (THs) are essential in normal brain development, and cognitive and emotional functions. THs act through a cascade of events including uptake by the target cells by specific cell membrane transporters, activation or inactivation by deiodinase enzymes, and interaction with nuclear thyroid hormone receptors. Several thyroid responsive genes have been described in the developing and in the adult brain and many studies have demonstrated a systemic or local reduction in TH availability in neurologic disease and after brain injury. In this review, the main factors and mechanisms associated with the THs in the normal and damaged brain will be evaluated in different regions and cellular contexts. Furthermore, the most common animal models used to study the role of THs in brain damage and cognitive impairment will be described and the use of THs as a potential recovery strategy from neuropathological conditions will be evaluated. Finally, particular attention will be given to the link observed between TH alterations and increased risk of Alzheimer's Disease (AD), the most prevalent neurodegenerative and dementing condition worldwide.
Collapse
Affiliation(s)
- Laura Sabatino
- Institute of Clinical Physiology, National Council of Research, Via Moruzzi 1, 56124 Pisa, Italy;
| | - Dominga Lapi
- Department of Biology, University of Pisa, 56127 Pisa, Italy;
| | - Cristina Del Seppia
- Institute of Clinical Physiology, National Council of Research, Via Moruzzi 1, 56124 Pisa, Italy;
| |
Collapse
|
8
|
Salas-Lucia F, Fekete C, Sinkó R, Egri P, Rada K, Ruska Y, Gereben B, Bianco AC. Axonal T3 uptake and transport can trigger thyroid hormone signaling in the brain. eLife 2023; 12:e82683. [PMID: 37204837 PMCID: PMC10241515 DOI: 10.7554/elife.82683] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 05/18/2023] [Indexed: 05/20/2023] Open
Abstract
The development of the brain, as well as mood and cognitive functions, are affected by thyroid hormone (TH) signaling. Neurons are the critical cellular target for TH action, with T3 regulating the expression of important neuronal gene sets. However, the steps involved in T3 signaling remain poorly known given that neurons express high levels of type 3 deiodinase (D3), which inactivates both T4 and T3. To investigate this mechanism, we used a compartmentalized microfluid device and identified a novel neuronal pathway of T3 transport and action that involves axonal T3 uptake into clathrin-dependent, endosomal/non-degradative lysosomes (NDLs). NDLs-containing T3 are retrogradely transported via microtubules, delivering T3 to the cell nucleus, and doubling the expression of a T3-responsive reporter gene. The NDLs also contain the monocarboxylate transporter 8 (Mct8) and D3, which transport and inactivate T3, respectively. Notwithstanding, T3 gets away from degradation because D3's active center is in the cytosol. Moreover, we used a unique mouse system to show that T3 implanted in specific brain areas can trigger selective signaling in distant locations, as far as the contralateral hemisphere. These findings provide a pathway for L-T3 to reach neurons and resolve the paradox of T3 signaling in the brain amid high D3 activity.
Collapse
Affiliation(s)
- Federico Salas-Lucia
- Section of Adult and Pediatric Endocrinology and Metabolism, University of ChicagoChicagoUnited States
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental MedicineBudapestHungary
| | - Richárd Sinkó
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
- János Szentágothai PhD School of Neurosciences, Semmelweis UniversityBudapestHungary
| | - Péter Egri
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
| | - Kristóf Rada
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
| | - Yvette Ruska
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental MedicineBudapestHungary
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
| | - Antonio C Bianco
- Section of Adult and Pediatric Endocrinology and Metabolism, University of ChicagoChicagoUnited States
| |
Collapse
|
9
|
Chen Z, van der Sman AE, Groeneweg S, de Rooij LJ, Visser WE, Peeters RP, Meima ME. Thyroid Hormone Transporters in a Human Placental Cell Model. Thyroid 2022; 32:1129-1137. [PMID: 35699060 PMCID: PMC9526468 DOI: 10.1089/thy.2021.0503] [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: 11/28/2022]
Abstract
Background: Fetal brain development in the first half of pregnancy is dependent on maternal thyroid hormone (TH), highlighting the importance of trans-placental TH transport. It is yet unclear which transporters are involved in this process. We aimed to identify the major TH transporters in a human placental cell model (BeWo cells). Methods: Messenger RNA expression of the known TH transporters (the monocarboxylate transporter [MCT]8, MCT10, the L-type amino acid transporter [LAT]1, LAT2, the organic anion transporting peptide [OATP]1A2 and OATP4A1) in BeWo cells and human placenta were determined by quantitative PCR. To determine the specificity and efficacy of transporter inhibitors, we first determined TH uptake at different inhibitor concentrations in African green monkey kidney fibroblast-like cells (COS1 cells) overexpressing TH transporters. We then tested TH uptake in BeWo cells in the presence or absence of the optimal inhibitor concentrations. Results: All tested TH transporters were expressed in human term placentas, whereas MCT8 was absent in BeWo cells. Both 2-amino-2-norbornanecarboxylic acid (BCH) and L-tryptophan at 1 mM inhibited LATs, whereas at the highest concentration (10 mM) L-tryptophan also inhibited MCT10. Verapamil inhibited OATP1A2 and less efficiently both MCTs, but not LATs. Both rifampicin and naringin reduced OATP1A2 activity. Finally, silychristin inhibited MCT8 at submicromolar concentrations and OATP1A2 partially only at the highest concentration tested (10 μM). In BeWo cells, verapamil reduced triiodothyronine (T3) uptake by 24%, BCH by 31%, and 1 mM L-tryptophan by 41%. The combination of BCH and verapamil additively decreased T3 uptake by 53% and the combination of BCH and 10 mM L-tryptophan by 60%, suggesting a major role for MCT10 and LATs in placental T3 uptake. Indeed, transfection of BeWo cells with MCT10-specific small interfering RNA significantly reduced T3 uptake. Only the combination of BCH and verapamil significantly reduced thyroxine (T4) uptake in BeWo cells, by 32%. Conclusions: Using pharmacological inhibitors, we show that MCT10 and LATs play a major role in T3 uptake in BeWo cells. T4 uptake appears independent of known TH transporters, suggesting the presence of, currently unknown, alternative transporter(s).
Collapse
Affiliation(s)
- Zhongli Chen
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A.S. Elise van der Sman
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Stefan Groeneweg
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Linda Johanna de Rooij
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - W. Edward Visser
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robin P. Peeters
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marcel E. Meima
- Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Address correspondence to: Marcel E. Meima, PhD, Erasmus MC Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam 3015 CN, The Netherlands
| |
Collapse
|
10
|
Brûlé E, Silander TL, Wang Y, Zhou X, Bak B, Groeneweg S, Bernard DJ. IGSF1 Deficiency Leads to Reduced TSH Production Independent of Alterations in Thyroid Hormone Action in Male Mice. Endocrinology 2022; 163:6609251. [PMID: 35708735 PMCID: PMC9258739 DOI: 10.1210/endocr/bqac092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Indexed: 11/19/2022]
Abstract
Loss of function mutations in IGSF1/Igsf1 cause central hypothyroidism. Igsf1 knockout mice have reduced pituitary thyrotropin-releasing hormone receptor, Trhr, expression, perhaps contributing to the phenotype. Because thyroid hormones negatively regulate Trhr, we hypothesized that IGSF1 might affect thyroid hormone availability in pituitary thyrotropes. Consistent with this idea, IGSF1 coimmunoprecipitated with the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) in transfected cells. This association was impaired with IGSF1 bearing patient-derived mutations. Wild-type IGSF1 did not, however, alter MCT8-mediated thyroid hormone import into heterologous cells. IGSF1 and MCT8 are both expressed in the apical membrane of the choroid plexus. However, MCT8 protein levels and localization in the choroid plexus were unaltered in Igsf1 knockout mice, ruling out a necessary chaperone function for IGSF1. MCT8 expression was low in the pituitary and was similarly unaffected in Igsf1 knockouts. We next assessed whether IGSF1 affects thyroid hormone transport or action, by MCT8 or otherwise, in vivo. To this end, we treated hypothyroid wild-type and Igsf1 knockout mice with exogenous thyroid hormones. T4 and T3 inhibited TSH release and regulated pituitary and forebrain gene expression similarly in both genotypes. Interestingly, pituitary TSH beta subunit (Tshb) expression was consistently reduced in Igsf1 knockouts relative to wild-type regardless of experimental condition, whereas Trhr was more variably affected. Although IGSF1 and MCT8 can interact in heterologous cells, the physiological relevance of their association is not clear. Nevertheless, the results suggest that IGSF1 loss can impair TSH production independently of alterations in TRHR levels or thyroid hormone action.
Collapse
Affiliation(s)
- Emilie Brûlé
- Department of Anatomy and Cell Biology, McGill University, Montreal H3G 1Y6, Canada
| | - Tanya L Silander
- Integrated Program in Neuroscience, McGill University, Montreal H3G 1Y6, Canada
| | - Ying Wang
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Xiang Zhou
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Beata Bak
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - Daniel J Bernard
- Correspondence: Daniel J. Bernard, PhD, Department of Pharmacology and Therapeutics, McGill University, McIntyre Medical Building, 3655 Prom. Sir William Osler, Room 1320, Montreal, Quebec H3G 1Y6, Canada.
| |
Collapse
|
11
|
Neuropsychological Alterations in Patients with Congenital Hypothyroidism Treated with Levothyroxine: Linked Factors and Thyroid Hormone Hyposensitivity. J Clin Med 2022; 11:jcm11123427. [PMID: 35743497 PMCID: PMC9224966 DOI: 10.3390/jcm11123427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/04/2022] Open
Abstract
Eighty-five percent of the studies of patients with congenital hypothyroidism (CH) treated with Levothyroxine (L-T4) report neuropsychological sequelae throughout life. In neonates and infants, there is a deficit in sensorimotor skills (impaired balance). In preschool and elementary school children and adolescents, there are alterations in intellectual quotient (low scores), language (delayed phonological acquisition), memory (visual, verbal, visuospatial, visuoconstructive, autobiographical, and semantic), sensorimotor skills (impaired fine and gross motor control), and visuoconstructive–visuospatial domain (low scores in spatial location, block design, and object assembly). These neuropsychological domains are also affected in young adults, except for language (adequate verbal fluency) and visuoconstructive–visuospatial domain (no data). The onset and severity of neuropsychological sequelae in patients with treated CH depend on several factors: extrinsic, related to L-T4 treatment and social aspects, and intrinsic, such as severity and etiology of CH, as well as structural and physiological changes in the brain. In this review, we hypothesized that thyroid hormone hyposensitivity (THH) could also contribute to neuropsychological alterations by reducing the effectiveness of L-T4 treatment in the brain. Thus, further research could approach the THH hypothesis at basic and clinical levels to implement new endocrinological and neuropsychological therapies for CH patients.
Collapse
|
12
|
Di Cosmo C, De Marco G, Agretti P, Ferrarini E, Dimida A, Falcetta P, Benvenga S, Vitti P, Tonacchera M. Screening for drugs potentially interfering with MCT8-mediated T 3 transport in vitro identifies dexamethasone and some commonly used drugs as inhibitors of MCT8 activity. J Endocrinol Invest 2022; 45:803-814. [PMID: 34850364 DOI: 10.1007/s40618-021-01711-4] [Citation(s) in RCA: 3] [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] [Received: 07/08/2021] [Accepted: 11/19/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Monocarboxylate transporter 8 (MCT8) is the first thyroid hormone transporter that has been linked to a human disease. Besides genetic alterations other factors might impair MCT8 activity. AIM This study aimed at investigating whether some common drugs having a structural similarity with TH and/or whose treatment is associated with thyroid function test abnormalities, or which behave as antagonists of TH action can inhibit MCT8-mediated T3 transport. METHODS [125I]T3 uptake and efflux were measured in COS-7 cells transiently transfected with hMCT8 before and after exposure to increasing concentrations of hydrocortisone, dexamethasone, prednisone, prednisolone, amiodarone, desethylamiodarone, dronedarone, buspirone, carbamazepine, valproic acid, and L-carnitine. The mode of inhibition was also determined. RESULTS Dexamethasone significantly inhibited T3 uptake at 10 μM; hydrocortisone reduced T3 uptake only at high concentrations, i.e. at 500 and 1000 μM; prednisone and prednisolone were devoid of inhibitory potential. Amiodarone caused a reduction of T3 uptake by MCT8 only at the highest concentrations used (44% at 50 μM and 68% at 100 μM), and this effect was weaker than that produced by desethylamiodarone and dronedarone; buspirone resulted a potent inhibitor, reducing T3 uptake at 0.1-10 μM. L-Carnitine inhibited T3 uptake only at 500 mM and 1 M. Kinetic experiments revealed a noncompetitive mode of inhibition for all compounds. All drugs inhibiting T3 uptake did not affect T3 release. CONCLUSION This study shows a novel effect of some common drugs, which is inhibition of T3 transport mediated by MCT8. Specifically, dexamethasone, buspirone, desethylamiodarone, and dronedarone behave as potent inhibitors of MCT8.
Collapse
Affiliation(s)
- C Di Cosmo
- Department of Clinical and Experimental Medicine, Endocrinology Unit, University of Pisa, via Paradisa 2, 56124, Pisa, Italy.
| | - G De Marco
- Department of Clinical and Experimental Medicine, Endocrinology Unit, University of Pisa, via Paradisa 2, 56124, Pisa, Italy
| | - P Agretti
- Laboratory of Chemistry and Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - E Ferrarini
- Department of Clinical and Experimental Medicine, Endocrinology Unit, University of Pisa, via Paradisa 2, 56124, Pisa, Italy
| | - A Dimida
- Department of Clinical and Experimental Medicine, Endocrinology Unit, University of Pisa, via Paradisa 2, 56124, Pisa, Italy
| | - P Falcetta
- Department of Clinical and Experimental Medicine, Endocrinology Unit, University of Pisa, via Paradisa 2, 56124, Pisa, Italy
| | - S Benvenga
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - P Vitti
- Department of Clinical and Experimental Medicine, Endocrinology Unit, University of Pisa, via Paradisa 2, 56124, Pisa, Italy
| | - M Tonacchera
- Department of Clinical and Experimental Medicine, Endocrinology Unit, University of Pisa, via Paradisa 2, 56124, Pisa, Italy
| |
Collapse
|
13
|
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: 7] [Impact Index Per Article: 2.3] [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
|
14
|
Lademann F, Mayerl S, Tsourdi E, Verrey F, Leitch VD, Williams GR, Bassett JHD, Hofbauer LC, Heuer H, Rauner M. The Thyroid Hormone Transporter MCT10 Is a Novel Regulator of Trabecular Bone Mass and Bone Turnover in Male Mice. Endocrinology 2022; 163:bqab218. [PMID: 34669927 PMCID: PMC8598386 DOI: 10.1210/endocr/bqab218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Indexed: 11/19/2022]
Abstract
Thyroid hormones (TH) are essential for skeletal development and adult bone homeostasis. Their bioavailability is determined by specific transporter proteins at the cell surface. The TH-specific transporter monocarboxylate transporter 8 (MCT8) was recently reported as a regulator of bone mass in mice. Given that high systemic triiodothyronine (T3) levels in Mct8 knockout (KO) mice are still able to cause trabecular bone loss, alternative TH transporters must substitute for MCT8 function in bone. In this study, we analyzed the skeletal phenotypes of male Oatp1c1 KO and Mct10 KO mice, which are euthyroid, and male Mct8/Oatp1c1 and Mct8/Mct10 double KO mice, which have elevated circulating T3 levels, to unravel the role of TH transport in bone. MicroCT analysis showed no significant trabecular bone changes in Oatp1c1 KO mice at 4 weeks and 16 weeks of age compared with wild-type littermate controls, whereas 16-week-old Mct8/Oatp1c1 double KO animals displayed trabecular bone loss. At 12 weeks, Mct10 KO mice, but not Mct8/Mct10 double KO mice, had decreased trabecular femoral bone volume with reduced osteoblast numbers. By contrast, lack of Mct10 in 24-week-old mice led to trabecular bone gain at the femur with increased osteoblast numbers and decreased osteoclast numbers whereas Mct8/Mct10 double KO did not alter bone mass. Neither Mct10 nor Mct8/Mct10 deletion affected vertebral bone structures at both ages. In vitro, osteoblast differentiation and activity were impaired by Mct10 and Mct8/Mct10-deficiency. These data demonstrate that MCT10, but not OATP1C1, is a site- and age-dependent regulator of bone mass and turnover in male mice.
Collapse
Affiliation(s)
- Franziska Lademann
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Steffen Mayerl
- Department of Endocrinology, University of Duisburg-Essen, University Hospital Essen, D-45147 Essen, Germany
| | - Elena Tsourdi
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Francois Verrey
- Institute of Physiology, University of Zurich, CH-8057 Zurich, Switzerland
| | - Victoria D Leitch
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Heike Heuer
- Department of Endocrinology, University of Duisburg-Essen, University Hospital Essen, D-45147 Essen, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, D-01307 Dresden, Germany
| |
Collapse
|
15
|
Nguyen YTK, Ha HTT, Nguyen TH, Nguyen LN. The role of SLC transporters for brain health and disease. Cell Mol Life Sci 2021; 79:20. [PMID: 34971415 PMCID: PMC11071821 DOI: 10.1007/s00018-021-04074-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/05/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022]
Abstract
The brain exchanges nutrients and small molecules with blood via the blood-brain barrier (BBB). Approximately 20% energy intake for the body is consumed by the brain. Glucose is known for its critical roles for energy production and provides substrates for biogenesis in neurons. The brain takes up glucose via glucose transporters GLUT1 and 3, which are expressed in several neural cell types. The brain is also equipped with various transport systems for acquiring amino acids, lactate, ketone bodies, lipids, and cofactors for neuronal functions. Unraveling the mechanisms by which the brain takes up and metabolizes these nutrients will be key in understanding the nutritional requirements in the brain. This could also offer opportunities for therapeutic interventions in several neurological disorders. For instance, emerging evidence suggests a critical role of lactate as an alternative energy source for neurons. Neuronal cells express monocarboxylic transporters to acquire lactate. As such, treatment of GLUT1-deficient patients with ketogenic diets to provide the brain with alternative sources of energy has been shown to improve the health of the patients. Many transporters are present in the brain, but only a small number has been characterized. In this review, we will discuss about the roles of solute carrier (SLC) transporters at the blood brain barrier (BBB) and neural cells, in transport of nutrients and metabolites in the brain.
Collapse
Affiliation(s)
- Yen T K Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Hoa T T Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Tra H Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
- SLING/Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- Immunology Translational and Cardiovascular Disease Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.
| |
Collapse
|
16
|
Low striatal T3 is implicated in inattention and memory impairment in an ADHD mouse model overexpressing thyroid hormone-responsive protein. Commun Biol 2021; 4:1101. [PMID: 34545202 PMCID: PMC8452653 DOI: 10.1038/s42003-021-02633-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder, potentially with a biological basis; however, its exact cause remains unknown. Thyroid hormone (TH) abnormalities are more prevalent in patients with ADHD than in the general population, indicating a shared pathogenetic mechanism for these conditions. Previously, we identified that overexpression of thyroid hormone-responsive protein (THRSP), a gene highly responsive to TH status, induced inattention in male mice. Herein, we sought to explore whether TH function in THRSP-overexpressing (THRSP OE) mice influences ADHD-like (inattention) behavior. We now confirm that THRSP overexpression in male mice reproduces behavioral features of ADHD, including sustained inattention and memory impairment, accompanied by excessive theta waves that were found normal in both the THRSP-knockout and hetero groups. Physiological characterization revealed low striatal T3 levels in the THRSP OE mice due to reduced striatal T3-specific monocarboxylate transporter 8 (MCT8), indicating brain-specific hypothyroidism in this transgenic mouse strain. TH replacement for seven days rescued inattention and memory impairment and the normalization of theta waves. This study further supports the involvement of the upregulated THRSP gene in ADHD pathology and indicates that THRSP OE mice can serve as an animal model for the predominantly inattentive subtype of ADHD.
Collapse
|
17
|
The Association between the rs312457 Genotype of the SLC16a13 Gene and Diabetes Mellitus in a Chinese Population. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:9918055. [PMID: 34257700 PMCID: PMC8257359 DOI: 10.1155/2021/9918055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/02/2021] [Indexed: 12/13/2022]
Abstract
Objective SLC16a genes encode H + -coupled monocarboxylate transporters (MCTs). MCTs are involved in maintaining interstitial fluids' pH and regulating insulin's binding affinity to its receptor, which is a potential mechanism for the onset of diabetes. In this research, we make explorations of the association between the rs312457 genotype of the SLC16a13 gene and diabetes in the Chinese population. Methods It included 384 type 2 diabetes patients and 1,468 healthy control subjects in total. We measured the anthropometric parameters, glycaemic index, homeostasis model assessment-B cell (HOMA-%B), lipid profile, and homeostasis model assessment-insulin resistance (HOMA-IR). The associations between the rs312457 genotype and type 2 diabetes were analyzed. Results The rs312457 genotype was markedly in relation to type 2 diabetes (P = 0.002). The frequency of the rs312457 risk allele (G) was 4.8%, higher than that of the wild-type allele (A) in patients of type 2 diabetes, indicating that allele (G)'s presence seemed to make the risk of type 2 diabetes go up. Compared to the GA and AA genotypes, the GG genotype of rs312457 significantly increased the risk of contracting diabetes mellitus (P ≤ 0.001). Moreover, the rs312457 genotype was associated with HOMA-%B. Subjects harbored the GG genotype of rs312457, whose HOMA-%B level went down in comparison with that in subjects harboring the AA genotype (P = 0.023). Conclusion Our results revealed that the rs312457 genotype of the SLC16a13 gene was correlated with the development of diabetes mellitus in the Chinese population.
Collapse
|
18
|
Fairweather SJ, Shah N, Brӧer S. Heteromeric Solute Carriers: Function, Structure, Pathology and Pharmacology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 21:13-127. [PMID: 33052588 DOI: 10.1007/5584_2020_584] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Solute carriers form one of three major superfamilies of membrane transporters in humans, and include uniporters, exchangers and symporters. Following several decades of molecular characterisation, multiple solute carriers that form obligatory heteromers with unrelated subunits are emerging as a distinctive principle of membrane transporter assembly. Here we comprehensively review experimentally established heteromeric solute carriers: SLC3-SLC7 amino acid exchangers, SLC16 monocarboxylate/H+ symporters and basigin/embigin, SLC4A1 (AE1) and glycophorin A exchanger, SLC51 heteromer Ost α-Ost β uniporter, and SLC6 heteromeric symporters. The review covers the history of the heteromer discovery, transporter physiology, structure, disease associations and pharmacology - all with a focus on the heteromeric assembly. The cellular locations, requirements for complex formation, and the functional role of dimerization are extensively detailed, including analysis of the first complete heteromer structures, the SLC7-SLC3 family transporters LAT1-4F2hc, b0,+AT-rBAT and the SLC6 family heteromer B0AT1-ACE2. We present a systematic analysis of the structural and functional aspects of heteromeric solute carriers and conclude with common principles of their functional roles and structural architecture.
Collapse
Affiliation(s)
- Stephen J Fairweather
- Research School of Biology, Australian National University, Canberra, ACT, Australia. .,Resarch School of Chemistry, Australian National University, Canberra, ACT, Australia.
| | - Nishank Shah
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Stefan Brӧer
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
19
|
Barisón MJ, Pereira IT, Waloski Robert A, Dallagiovanna B. Reorganization of Metabolism during Cardiomyogenesis Implies Time-Specific Signaling Pathway Regulation. Int J Mol Sci 2021; 22:1330. [PMID: 33572750 PMCID: PMC7869011 DOI: 10.3390/ijms22031330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 11/17/2022] Open
Abstract
Understanding the cell differentiation process involves the characterization of signaling and regulatory pathways. The coordinated action involved in multilevel regulation determines the commitment of stem cells and their differentiation into a specific cell lineage. Cellular metabolism plays a relevant role in modulating the expression of genes, which act as sensors of the extra-and intracellular environment. In this work, we analyzed mRNAs associated with polysomes by focusing on the expression profile of metabolism-related genes during the cardiac differentiation of human embryonic stem cells (hESCs). We compared different time points during cardiac differentiation (pluripotency, embryoid body aggregation, cardiac mesoderm, cardiac progenitor and cardiomyocyte) and showed the immature cell profile of energy metabolism. Highly regulated canonical pathways are thoroughly discussed, such as those involved in metabolic signaling and lipid homeostasis. We reveal the critical relevance of retinoic X receptor (RXR) heterodimers in upstream retinoic acid metabolism and their relationship with thyroid hormone signaling. Additionally, we highlight the importance of lipid homeostasis and extracellular matrix component biosynthesis during cardiomyogenesis, providing new insights into how hESCs reorganize their metabolism during in vitro cardiac differentiation.
Collapse
Affiliation(s)
| | | | | | - Bruno Dallagiovanna
- Basic Stem Cell Biology Laboratory, Instituto Carlos Chagas-FIOCRUZ-PR, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR 81350-010, Brazil; (M.J.B.); (I.T.P.); (A.W.R.)
| |
Collapse
|
20
|
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: 0.8] [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
|
21
|
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: 0.2] [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
|
22
|
Wassner AJ. MCT8 deficiency: collaborative rare disease phenotyping for care and research. Lancet Diabetes Endocrinol 2020; 8:555-557. [PMID: 32559466 DOI: 10.1016/s2213-8587(20)30186-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 11/19/2022]
Affiliation(s)
- Ari J Wassner
- Thyroid Center, Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
23
|
Groeneweg S, van Geest FS, Abacı A, Alcantud A, Ambegaonkar GP, Armour CM, Bakhtiani P, Barca D, Bertini ES, van Beynum IM, Brunetti-Pierri N, Bugiani M, Cappa M, Cappuccio G, Castellotti B, Castiglioni C, Chatterjee K, de Coo IFM, Coutant R, Craiu D, Crock P, DeGoede C, Demir K, Dica A, Dimitri P, Dolcetta-Capuzzo A, Dremmen MHG, Dubey R, Enderli A, Fairchild J, Gallichan J, George B, Gevers EF, Hackenberg A, Halász Z, Heinrich B, Huynh T, Kłosowska A, van der Knaap MS, van der Knoop MM, Konrad D, Koolen DA, Krude H, Lawson-Yuen A, Lebl J, Linder-Lucht M, Lorea CF, Lourenço CM, Lunsing RJ, Lyons G, Malikova J, Mancilla EE, McGowan A, Mericq V, Lora FM, Moran C, Müller KE, Oliver-Petit I, Paone L, Paul PG, Polak M, Porta F, Poswar FO, Reinauer C, Rozenkova K, Menevse TS, Simm P, Simon A, Singh Y, Spada M, van der Spek J, Stals MAM, Stoupa A, Subramanian GM, Tonduti D, Turan S, den Uil CA, Vanderniet J, van der Walt A, Wémeau JL, Wierzba J, de Wit MCY, Wolf NI, Wurm M, Zibordi F, Zung A, Zwaveling-Soonawala N, Visser WE. Disease characteristics of MCT8 deficiency: an international, retrospective, multicentre cohort study. Lancet Diabetes Endocrinol 2020; 8:594-605. [PMID: 32559475 PMCID: PMC7611932 DOI: 10.1016/s2213-8587(20)30153-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/14/2020] [Accepted: 04/19/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Disordered thyroid hormone transport, due to mutations in the SLC16A2 gene encoding monocarboxylate transporter 8 (MCT8), is characterised by intellectual and motor disability resulting from cerebral hypothyroidism and chronic peripheral thyrotoxicosis. We sought to systematically assess the phenotypic characteristics and natural history of patients with MCT8 deficiency. METHODS We did an international, multicentre, cohort study, analysing retrospective data from Jan 1, 2003, to Dec 31, 2019, from patients with MCT8 deficiency followed up in 47 hospitals in 22 countries globally. The key inclusion criterion was genetically confirmed MCT8 deficiency. There were no exclusion criteria. Our primary objective was to analyse the overall survival of patients with MCT8 deficiency and document causes of death. We also compared survival between patients who did or did not attain full head control by age 1·5 years and between patients who were or were not underweight by age 1-3 years (defined as a bodyweight-for-age Z score <-2 SDs or <5th percentile according to WHO definition). Other objectives were to assess neurocognitive function and outcomes, and clinical parameters including anthropometric characteristics, biochemical markers, and neuroimaging findings. FINDINGS Between Oct 14, 2014, and Jan 17, 2020, we enrolled 151 patients with 73 different MCT8 (SLC16A2) mutations. Median age at diagnosis was 24·0 months (IQR 12·0-60·0, range 0·0-744·0). 32 (21%) of 151 patients died; the main causes of mortality in these patients were pulmonary infection (six [19%]) and sudden death (six [19%]). Median overall survival was 35·0 years (95% CI 8·3-61·7). Individuals who did not attain head control by age 1·5 years had an increased risk of death compared with patients who did attain head control (hazard ratio [HR] 3·46, 95% CI 1·76-8·34; log-rank test p=0·0041). Patients who were underweight during age 1-3 years had an increased risk for death compared with patients who were of normal bodyweight at this age (HR 4·71, 95% CI 1·26-17·58, p=0·021). The few motor and cognitive abilities of patients did not improve with age, as evidenced by the absence of significant correlations between biological age and scores on the Gross Motor Function Measure-88 and Bayley Scales of Infant Development III. Tri-iodothyronine concentrations were above the age-specific upper limit in 96 (95%) of 101 patients and free thyroxine concentrations were below the age-specific lower limit in 94 (89%) of 106 patients. 59 (71%) of 83 patients were underweight. 25 (53%) of 47 patients had elevated systolic blood pressure above the 90th percentile, 34 (76%) of 45 patients had premature atrial contractions, and 20 (31%) of 64 had resting tachycardia. The most consistent MRI finding was a global delay in myelination, which occurred in 13 (100%) of 13 patients. INTERPRETATION Our description of characteristics of MCT8 deficiency in a large patient cohort reveals poor survival with a high prevalence of treatable underlying risk factors, and provides knowledge that might inform clinical management and future evaluation of therapies. FUNDING Netherlands Organisation for Health Research and Development, and the Sherman Foundation.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ferdy S van Geest
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ayhan Abacı
- Division of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Alberto Alcantud
- Pediatric Neurology Section, Hospital Francesc de Borja de Gandia, Valencia, Spain
| | - Gautem P Ambegaonkar
- Department of Paediatric Neurology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Christine M Armour
- Regional Genetics Program, Children's Hospital of Eastern Ontario, and Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | | | - Diana Barca
- Paediatric Neurology Clinic, Alexandru Obregia Hospital, Bucharest, Romania; Department of Neurosciences, Paediatric Neurology Discipline II, Carol Davila University of Medicine, Bucharest, Romania
| | - Enrico S Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesu' Children's Research Hospital IRCCS, Rome, Italy
| | - Ingrid M van Beynum
- Sophia Children's Hospital, Division of Paediatric Cardiology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, Naples, Italy; Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Marianna Bugiani
- Department of Child Neurology, Center for Childhood White Matter Diseases, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, and Amsterdam Neuroscience, Amsterdam, Netherlands; Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Marco Cappa
- Division of Endocrinology, Bambino Gesu' Children's Research Hospital IRCCS, Rome, Italy
| | - Gerarda Cappuccio
- Department of Translational Medicine, Federico II University, Naples, Italy; Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Barbara Castellotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Krishna Chatterjee
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Irenaeus F M de Coo
- Department of Paediatric Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Régis Coutant
- Department of Pediatric Endocrinology and Diabetology, University Hospital, Angers, France
| | - Dana Craiu
- Paediatric Neurology Clinic, Alexandru Obregia Hospital, Bucharest, Romania; Department of Neurosciences, Paediatric Neurology Discipline II, Carol Davila University of Medicine, Bucharest, Romania
| | - Patricia Crock
- John Hunter Children's Hospital and University of Newcastle, Newcastle, NSW, Australia
| | | | - Korcan Demir
- Division of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Alice Dica
- Paediatric Neurology Clinic, Alexandru Obregia Hospital, Bucharest, Romania; Department of Neurosciences, Paediatric Neurology Discipline II, Carol Davila University of Medicine, Bucharest, Romania
| | - Paul Dimitri
- Sheffield Children's NHS Foundation Trust, Sheffield Hallam University and University of Sheffield, Sheffield, UK
| | - Anna Dolcetta-Capuzzo
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands; Università Vita-Salute San Raffaele, Milan, Italy
| | | | | | - Anina Enderli
- Department of Neuropediatrics, University Children's Hospital Zurich, Zürich, Switzerland
| | - Jan Fairchild
- Department of Diabetes and Endocrinology, Women's and Children's Hospital, North Adelaide, SA, Australia
| | | | - Belinda George
- Department of Endocrinology, St. John's Medical College Hospital, Bengaluru, Karnataka, India
| | - Evelien F Gevers
- Centre for Endocrinology, William Harvey Research institute, Queen Mary University London, London, UK; Dept of Paediatric Endocrinology, Barts Health NHS Trust, London, UK
| | - Annette Hackenberg
- Department of Neuropediatrics, University Children's Hospital Zurich, Zürich, Switzerland
| | - Zita Halász
- Department of Paediatrics, Semmelweis University, Budapest, Hungary
| | - Bianka Heinrich
- Department of Neuropediatrics, University Children's Hospital Zurich, Zürich, Switzerland
| | - Tony Huynh
- Department of Endocrinology & Diabetes, Queensland Children's Hospital, South Brisbane, QLD, Australia; Department of Chemical Pathology, Mater Pathology, South Brisbane, QLD, Australia; Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Anna Kłosowska
- Medical University of Gdańsk, Department of Paediatrics, Haematology & Oncology, Department of General Nursery, Gdańsk, Poland
| | - Marjo S van der Knaap
- Department of Child Neurology, Center for Childhood White Matter Diseases, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, and Amsterdam Neuroscience, Amsterdam, Netherlands
| | | | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - David A Koolen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center (Radboudumc), Nijmegen, Netherlands
| | - Heiko Krude
- Department of Paediatric Endocrinology and Diabetology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Amy Lawson-Yuen
- Genomics Institute Mary Bridge Children's Hospital, MultiCare Health System Tacoma, WA, USA
| | - Jan Lebl
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Michaela Linder-Lucht
- Division of Neuropediatrics and Muscular Disorders, Department of Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Cláudia F Lorea
- Teaching Hospital of Universidade Federal de Pelotas, Pelotas, Brazil
| | - Charles M Lourenço
- Faculdade de Medicina, Centro Universitario Estácio de Ribeirão Preto, Ribeirão Preto, Brazil
| | - Roelineke J Lunsing
- Department of Child Neurology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Greta Lyons
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jana Malikova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Edna E Mancilla
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Anne McGowan
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Veronica Mericq
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile; Department of Pediatrics, Clinica Las Condes, Santiago, Chile
| | - Felipe M Lora
- Pediatric Endocrinology Group, Santa Catarina Hospital, São Paulo, Brazil
| | - Carla Moran
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | | | - Isabelle Oliver-Petit
- Department of Paediatric Endocrinology and Genetics, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Laura Paone
- Division of Endocrinology, Bambino Gesu' Children's Research Hospital IRCCS, Rome, Italy
| | - Praveen G Paul
- Department of Paediatrics, Christian Medical College, Vellore, India
| | - Michel Polak
- Paediatric Endocrinology, Diabetology and Gynaecology Department, Necker Children's University Hospital, Imagine Institute, Paris, France
| | - Francesco Porta
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Fabiano O Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Christina Reinauer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Medical Faculty, Duesseldorf, Germany
| | - Klara Rozenkova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Tuba S Menevse
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Peter Simm
- Royal Children's Hospital, Parkville, Melbourne, VIC, Australia
| | - Anna Simon
- Department of Paediatrics, Christian Medical College, Vellore, India
| | - Yogen Singh
- Department of Paediatric Cardiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Marco Spada
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Jet van der Spek
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center (Radboudumc), Nijmegen, Netherlands
| | - Milou A M Stals
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Athanasia Stoupa
- Paediatric Endocrinology, Diabetology and Gynaecology Department, Necker Children's University Hospital, Imagine Institute, Paris, France
| | | | - Davide Tonduti
- Child Neurology Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Serap Turan
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Corstiaan A den Uil
- Department of Cardiology and Intensive Care Medicine, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Joel Vanderniet
- John Hunter Children's Hospital and University of Newcastle, Newcastle, NSW, Australia
| | | | | | - Jolante Wierzba
- Medical University of Gdańsk, Department of Paediatrics, Haematology & Oncology, Department of General Nursery, Gdańsk, Poland
| | | | - Nicole I Wolf
- Department of Child Neurology, Center for Childhood White Matter Diseases, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, and Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Michael Wurm
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany; KUNO Children's University Hospital, Campus St. Hedwig, University of Regensburg, Regensburg, Germany
| | - Federica Zibordi
- Child Neurology Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Amnon Zung
- Paediatric Endocrinology Unit, Kaplan Medical Center, Rehovot, Israel; Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nitash Zwaveling-Soonawala
- Emma Children's Hospital, Department of Paediatric Endocrinology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - W Edward Visser
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
24
|
Colella M, Cuomo D, Giacco A, Mallardo M, De Felice M, Ambrosino C. Thyroid Hormones and Functional Ovarian Reserve: Systemic vs. Peripheral Dysfunctions. J Clin Med 2020; 9:E1679. [PMID: 32492950 PMCID: PMC7355968 DOI: 10.3390/jcm9061679] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
Thyroid hormones (THs) exert pleiotropic effects in different mammalian organs, including gonads. Genetic and non-genetic factors, such as ageing and environmental stressors (e.g., low-iodine intake, exposure to endocrine disruptors, etc.), can alter T4/T3 synthesis by the thyroid. In any case, peripheral T3, controlled by tissue-specific enzymes (deiodinases), receptors and transporters, ensures organ homeostasis. Conflicting reports suggest that both hypothyroidism and hyperthyroidism, assessed by mean of circulating T4, T3 and Thyroid-Stimulating Hormone (TSH), could affect the functionality of the ovarian reserve determining infertility. The relationship between ovarian T3 level and functional ovarian reserve (FOR) is poorly understood despite that the modifications of local T3 metabolism and signalling have been associated with dysfunctions of several organs. Here, we will summarize the current knowledge on the role of TH signalling and its crosstalk with other pathways in controlling the physiological and premature ovarian ageing and, finally, in preserving FOR. We will consider separately the reports describing the effects of circulating and local THs on the ovarian health to elucidate their role in ovarian dysfunctions.
Collapse
Affiliation(s)
- Marco Colella
- Department of Science and Technology, University of Sannio, via De Sanctis, 82100 Benevento, Italy; (M.C.); (A.G.)
- IRGS, Biogem-Scarl, Via Camporeale, Ariano Irpino, 83031 Avellino, Italy
| | - Danila Cuomo
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77843, USA;
| | - Antonia Giacco
- Department of Science and Technology, University of Sannio, via De Sanctis, 82100 Benevento, Italy; (M.C.); (A.G.)
| | - Massimo Mallardo
- Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Mario De Felice
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77843, USA;
- Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy;
- IEOS-CNR, Via Pansini 6, 80131 Naples, Italy
| | - Concetta Ambrosino
- Department of Science and Technology, University of Sannio, via De Sanctis, 82100 Benevento, Italy; (M.C.); (A.G.)
- IRGS, Biogem-Scarl, Via Camporeale, Ariano Irpino, 83031 Avellino, Italy
- IEOS-CNR, Via Pansini 6, 80131 Naples, Italy
| |
Collapse
|
25
|
Du ZP, Feng S, Li YL, Li R, Lv J, Ren WQ, Feng QW, Liu P, Wang QN. Di-(2-ethylhexyl) phthalate inhibits expression and internalization of transthyretin in human placental trophoblastic cells. Toxicol Appl Pharmacol 2020; 394:114960. [PMID: 32201330 DOI: 10.1016/j.taap.2020.114960] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/29/2020] [Accepted: 03/18/2020] [Indexed: 11/23/2022]
Abstract
During pregnancy, fetal thyroid hormones (THs) are dependent on maternal placental transport and their physiological level is crucial for normal fetal neurodevelopment. Earlier research has shown that Di-(2-ethylhexyl) phthalate (DEHP) disrupts thyroid function and THs homeostasis in pregnant women and fetuses, and affects placental THs transport. However, the underlying mechanisms are poorly understood. The present study, therefore, aimed to systematically investigate the potential mechanisms of DEHP-induced disruption in the placental THs transport using two human placental trophoblastic cells, HTR-8/SVneo cells and JEG-3 cells. While the exposure of DEHP at the doses of 0-400 μM for 24 h did not affect cell viability, we found reduced consumption of T3 and T4 in the culture medium of HTR-8/Svneo cells treated with DEHP at 400 μM. DEHP treatment did not affect T3 uptake and the expression of monocarboxylate transporters 8 (MCT8) and organic anion transporters 1C1 (OATP1C1). However, DEHP significantly inhibited transthyretin (TTR) internalization, down-regulated TTR, deiodinase 2 (DIO2), and thyroid hormone receptors mRNA expression and protein levels, and up-regulated deiodinase 3 (DIO3) protein levels in a dose-dependent manner. These results indicate that DEHP acts on placental trophoblast cells, inhibits its TTR internalization, down-regulates TTR expression and affects the expression of DIO2, DIO3, and thyroid hormone receptor. These may be the mechanisms by which PAEs affects THs transport through placental.
Collapse
Affiliation(s)
- Zhi-Ping Du
- Department of Toxicology, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Shun Feng
- Department of Toxicology, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Yan-Ling Li
- Department of Toxicology, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Rong Li
- Department of Toxicology, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Jia Lv
- Department of Toxicology, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Wen-Qiang Ren
- Department of Toxicology, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Qiang-Wei Feng
- Department of Toxicology, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
| | - Ping Liu
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin, New Zealand.
| | - Qu-Nan Wang
- Department of Toxicology, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes/Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China.
| |
Collapse
|
26
|
Gorini F, Bustaffa E, Coi A, Iervasi G, Bianchi F. Bisphenols as Environmental Triggers of Thyroid Dysfunction: Clues and Evidence. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E2654. [PMID: 32294918 PMCID: PMC7216215 DOI: 10.3390/ijerph17082654] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/25/2022]
Abstract
Bisphenols (BPs), and especially bisphenol A (BPA), are known endocrine disruptors (EDCs), capable of interfering with estrogen and androgen activities, as well as being suspected of other health outcomes. Given the crucial role of thyroid hormones and the increasing incidence of thyroid carcinoma in the last few decades, this review analyzes the effects of BPS on the thyroid, considering original research in vitro, in vivo, and in humans published from January 2000 to October 2019. Both in vitro and in vivo studies reported the ability of BPs to disrupt thyroid function through multiple mechanisms. The antagonism with thyroid receptors (TRs), which affects TR-mediated transcriptional activity, the direct action of BPs on gene expression at the thyroid and the pituitary level, the competitive binding with thyroid transport proteins, and the induction of toxicity in several cell lines are likely the main mechanisms leading to thyroid dysfunction. In humans, results are more contradictory, though some evidence suggests the potential of BPs in increasing the risk of thyroid nodules. A standardized methodology in toxicological studies and prospective epidemiological studies with individual exposure assessments are warranted to evaluate the pathophysiology resulting in the damage and to establish the temporal relationship between markers of exposure and long-term effects.
Collapse
|
27
|
Felmlee MA, Jones RS, Rodriguez-Cruz V, Follman KE, Morris ME. Monocarboxylate Transporters (SLC16): Function, Regulation, and Role in Health and Disease. Pharmacol Rev 2020; 72:466-485. [PMID: 32144120 PMCID: PMC7062045 DOI: 10.1124/pr.119.018762] [Citation(s) in RCA: 240] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The solute carrier family 16 (SLC16) is comprised of 14 members of the monocarboxylate transporter (MCT) family that play an essential role in the transport of important cell nutrients and for cellular metabolism and pH regulation. MCTs 1-4 have been extensively studied and are involved in the proton-dependent transport of L-lactate, pyruvate, short-chain fatty acids, and monocarboxylate drugs in a wide variety of tissues. MCTs 1 and 4 are overexpressed in a number of cancers, and current investigations have focused on transporter inhibition as a novel therapeutic strategy in cancers. MCT1 has also been used in strategies aimed at enhancing drug absorption due to its high expression in the intestine. Other MCT isoforms are less well characterized, but ongoing studies indicate that MCT6 transports xenobiotics such as bumetanide, nateglinide, and probenecid, whereas MCT7 has been characterized as a transporter of ketone bodies. MCT8 and MCT10 transport thyroid hormones, and recently, MCT9 has been characterized as a carnitine efflux transporter and MCT12 as a creatine transporter. Expressed at the blood brain barrier, MCT8 mutations have been associated with an X-linked intellectual disability, known as Allan-Herndon-Dudley syndrome. Many MCT isoforms are associated with hormone, lipid, and glucose homeostasis, and recent research has focused on their potential roles in disease, with MCTs representing promising novel therapeutic targets. This review will provide a summary of the current literature focusing on the characterization, function, and regulation of the MCT family isoforms and on their roles in drug disposition and in health and disease. SIGNIFICANCE STATEMENT: The 14-member solute carrier family 16 of monocarboxylate transporters (MCTs) plays a fundamental role in maintaining intracellular concentrations of a broad range of important endogenous molecules in health and disease. MCTs 1, 2, and 4 (L-lactate transporters) are overexpressed in cancers and represent a novel therapeutic target in cancer. Recent studies have highlighted the importance of MCTs in glucose, lipid, and hormone homeostasis, including MCT8 in thyroid hormone brain uptake, MCT12 in carnitine transport, and MCT11 in type 2 diabetes.
Collapse
Affiliation(s)
- Melanie A Felmlee
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| | - Robert S Jones
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| | - Vivian Rodriguez-Cruz
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| | - Kristin E Follman
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| | - Marilyn E Morris
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| |
Collapse
|
28
|
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: 114] [Impact Index Per Article: 22.8] [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
|
29
|
Lademann F, Tsourdi E, Rijntjes E, Köhrle J, Hofbauer LC, Heuer H, Rauner M. Lack of the Thyroid Hormone Transporter Mct8 in Osteoblast and Osteoclast Progenitors Increases Trabecular Bone in Male Mice. Thyroid 2020; 30:329-342. [PMID: 31910109 DOI: 10.1089/thy.2019.0271] [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: 11/12/2022]
Abstract
Background: Bone is an important target of thyroid hormones (THs), which require transport into target cells to exert their actions. Recently, the TH-specific monocarboxylate transporter 8 (Mct8) was reported as a regulator of bone mass in male mice. However, its global deletion leads to high 3,3',5-L-triiodothyronine (T3) serum concentrations that may mask direct effects of Mct8-deficiency on bone. In this study, we assessed the bone cell intrinsic function of Mct8 ex vivo and in vivo using conditional Mct8-knockout lines specifically targeting osteoclast and osteoblast progenitors, as well as mature osteoblasts and osteocytes. Materials and Methods: Twelve-week-old male mice with a global Mct8-deficiency or a conditional Mct8-knockout in osteoclast precursors, osteoprogenitors, or mature osteoblasts/osteocytes were analyzed regarding their bone microarchitecture, turnover, and strength. Furthermore, ex vivo studies were conducted to investigate the role of Mct8 in bone cell differentiation and functionality, as well as TH uptake. Results: Global Mct8-knockout mice demonstrated 1.7-fold higher T3 serum concentrations and trabecular bone loss (-28%) likely due to an increased bone turnover as shown by increased osteoblast (+45%) and osteoclast numbers (+41%). However, cortical bone mineral density was increased. Ex vivo cultures of bone marrow-derived osteoblasts and osteoclasts revealed highest expression of Mct8 in mature bone cells. In addition, Mct8-deficiency resulted in a lower mRNA expression of osteoblast and osteoclast differentiation markers, as well as a reduced mineralization capacity and osteoclast numbers, respectively, indicating a bone cell intrinsic role of Mct8. In fact, conditional Mct8-knockout and inhibition of Mct8 in osteoblasts led to an attenuated T3 uptake ex vivo. In vivo, osteoprogenitor-specific Mct8-knockout enhanced trabecular bone volume (+16%) with osteoblast numbers being increased 3.7 fold. Interestingly, Mct8-deficiency in osteoprogenitors and late osteoblasts/osteocytes both resulted in cortical bone loss. Finally, Mct8-deletion in osteoclast progenitors increased trabecular bone volume (+20%) due to reduced osteoclast numbers (-32%), whereas osteoblast numbers were enhanced (+25%). Conclusions: This study confirms that high systemic T3 in global Mct8-knockout mice masks the direct effect of Mct8. Moreover, it identifies Mct8 as a critical regulator of trabecular vs. cortical bone by regulating T3 uptake and highlights its cell intrinsic role in osteoclast and osteoblast progenitors.
Collapse
Affiliation(s)
- Franziska Lademann
- Department of Medicine III, Universitätsklinikum Dresden, Dresden, Germany
- Center for Healthy Aging, Universitätsklinikum Dresden, Dresden, Germany
| | - Elena Tsourdi
- Department of Medicine III, Universitätsklinikum Dresden, Dresden, Germany
- Center for Healthy Aging, Universitätsklinikum Dresden, Dresden, Germany
| | - Eddy Rijntjes
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Universitätsklinikum Dresden, Dresden, Germany
- Center for Healthy Aging, Universitätsklinikum Dresden, Dresden, Germany
| | - Heike Heuer
- Klinik für Endokrinologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Martina Rauner
- Department of Medicine III, Universitätsklinikum Dresden, Dresden, Germany
- Center for Healthy Aging, Universitätsklinikum Dresden, Dresden, Germany
| |
Collapse
|
30
|
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: 1.5] [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
|
31
|
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: 1.0] [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
|
32
|
Govindaraj V, Ungati H, Jakka SR, Bose S, Mugesh G. Directing Traffic: Halogen‐Bond‐Mediated Membrane Transport. Chemistry 2019; 25:11180-11192. [DOI: 10.1002/chem.201902243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/15/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Vijayakumar Govindaraj
- Department of Inorganic & Physical ChemistryIndian Institute of Science Bangalore 560012 India
| | - Harinarayana Ungati
- Department of Inorganic & Physical ChemistryIndian Institute of Science Bangalore 560012 India
| | - Surendar R. Jakka
- Department of Inorganic & Physical ChemistryIndian Institute of Science Bangalore 560012 India
| | - Sritama Bose
- Department of Inorganic & Physical ChemistryIndian Institute of Science Bangalore 560012 India
| | - Govindasamy Mugesh
- Department of Inorganic & Physical ChemistryIndian Institute of Science Bangalore 560012 India
| |
Collapse
|
33
|
Šošić-Jurjević B, Lütjohann D, Renko K, Filipović B, Radulović N, Ajdžanović V, Trifunović S, Nestorović N, Živanović J, Manojlović Stojanoski M, Kӧhrle J, Milošević V. The isoflavones genistein and daidzein increase hepatic concentration of thyroid hormones and affect cholesterol metabolism in middle-aged male rats. J Steroid Biochem Mol Biol 2019; 190:1-10. [PMID: 30885834 DOI: 10.1016/j.jsbmb.2019.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 01/05/2023]
Abstract
We examined whether isoflavones interfere with thyroid homeostasis, increase hepatic thyroid hormone concentrations and affect cholesterol metabolism in middle-aged (MA) male rats. Thirteen-month-old Wistar rats were injected subcutaneously with 35 mg/kg b.w./day of genistein, daidzein or vehicle (controls) for four weeks. Hepatic Dio1 gene expression was up-regulated by 70% (p < 0.001 for both) and Dio1 enzyme activity increased by 64% after genistein (p < 0.001) and 73% after daidzein treatment (p < 0.0001). Hepatic T3 was 75% higher (p < 0.05 for both), while T4 increased only after genistein treatment. Serum T4 concentrations were 31% lower in genistein- and 49% lower in dadzein-treated rats (p < 0.001 for both) compared with controls. Hepatic Cyp7a1 gene expression was up-regulated by 40% after genistein and 32% after daidzein treatment (p < 0.05 for both), in agreement with a 7α-hydroxycholesterol increase of 50% (p < 0.01) and 88% (p < 0.001), respectively. Serum 24- and 27-hydroxycholesterol were 30% lower (p < 0.05 for both), while only 24-hydroxycholesterol was decreased in the liver by 45% after genistein (p < 0.05) and 39% (p < 0.01) after dadzein treatment. Serum concentration of the cholesterol precursor desmosterol was 32% (p < 0.05) lower only after dadzein treatment alone, while both isoflavones elevated this parameter in the liver by 45% (p < 0.01). In conclusion, isoflavones increased T3 availability in the liver of MA males, despite decreasing serum T4. Hepatic increase of T3 possibly contributes to activation of the neutral pathway of cholesterol degradation into bile acids in the liver. While isoflavones obviously have the potential to trigger multiple mechanisms involved in cholesterol metabolism and oxysterol production, they failed to induce any hypocholesterolemic effect.
Collapse
Affiliation(s)
- B Šošić-Jurjević
- Institute for Biological Research, "Siniša Stanković", University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia.
| | - D Lütjohann
- Institut für Klinische Chemie und Klinische Pharmakologie, Universitätsklinikum Bonn, Sigmund-Freud-Str. 25, D-53127 Bonn, Germany
| | - K Renko
- Institut für Experimentelle Endokrinologie, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - B Filipović
- Institute for Biological Research, "Siniša Stanković", University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| | - N Radulović
- Department of Chemistry, Faculty of Science and Mathematics, University of Niš, Višegradska 33, 18000 Niš, Serbia
| | - V Ajdžanović
- Institute for Biological Research, "Siniša Stanković", University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| | - S Trifunović
- Institut für Klinische Chemie und Klinische Pharmakologie, Universitätsklinikum Bonn, Sigmund-Freud-Str. 25, D-53127 Bonn, Germany
| | - N Nestorović
- Institute for Biological Research, "Siniša Stanković", University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| | - J Živanović
- Institute for Biological Research, "Siniša Stanković", University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| | - M Manojlović Stojanoski
- Institute for Biological Research, "Siniša Stanković", University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| | - J Kӧhrle
- Institut für Experimentelle Endokrinologie, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - V Milošević
- Institute for Biological Research, "Siniša Stanković", University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| |
Collapse
|
34
|
Zevenbergen C, Groeneweg S, Swagemakers SMA, de Jong A, Medici-Van den Herik E, Rispens M, Klootwijk W, Medici M, de Rijke YB, Meima ME, Larsen PR, Chavatte L, Venter D, Peeters RP, Van der Spek PJ, Visser WE. Functional Analysis of Genetic Variation in the SECIS Element of Thyroid Hormone Activating Type 2 Deiodinase. J Clin Endocrinol Metab 2019; 104:1369-1377. [PMID: 30423129 DOI: 10.1210/jc.2018-01605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/08/2018] [Indexed: 01/05/2023]
Abstract
CONTEXT Thyroid hormone is important for normal brain development. The type 2 deiodinase (D2) controls thyroid hormone action in the brain by activating T4 to T3. The enzymatic activity of D2 depends on the incorporation of selenocysteine for which the selenocysteine-insertion sequence (SECIS) element located in the 3' untranslated region is indispensable. We hypothesized that mutations in the SECIS element could affect D2 function, resulting in a neurocognitive phenotype. OBJECTIVE To identify mutations in the SECIS element of DIO2 in patients with intellectual disability and to test their functional consequences. DESIGN, SETTING, AND PATIENTS The SECIS element of DIO2 was sequenced in 387 patients with unexplained intellectual disability using a predefined pattern of thyroid function tests. SECIS element read-through in wild-type or mutant D2 was quantified by a luciferase reporter system in transfected cells. Functional consequences were assessed by quantifying D2 activity in cell lysate or intact cell metabolism studies. RESULTS Sequence analysis revealed 2 heterozygous mutations: c.5703C>T and c.5730A>T, which were also present in the unaffected family members. The functional evaluation showed that both mutations did not affect D2 enzyme activity in cell lysates or intact cells, although the 5730A>T mutation decreased SECIS element read-through by 75%. In the patient harboring the c.5730A>T variant, whole genome sequencing revealed a pathogenic deletion of the STXBP1 gene. CONCLUSIONS We report on two families with mutations in the SECIS element of D2. Although functional analysis showed that nucleotide 5730 is important for normal SECIS element read-through, the two variants did not segregate with a distinct phenotype.
Collapse
Affiliation(s)
- Chantal Zevenbergen
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Stefan Groeneweg
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Sigrid M A Swagemakers
- Department of Bioinformatics, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Pathology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | | | - Evita Medici-Van den Herik
- Department of Child Neurology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | | | - Wim Klootwijk
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Marco Medici
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Marcel E Meima
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - P Reed Larsen
- Department of Internal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Laurent Chavatte
- Centre International de Recherche en Infectiologie, CIRI, INSERM U1111, CNRS/ENS/UCBL1 UMR5308, Lyon, France
| | - Deon Venter
- Department of Pathology, Mater Health Services, South Brisbane, Queensland, Australia
| | - Robin P Peeters
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Peter J Van der Spek
- Department of Bioinformatics, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Pathology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
35
|
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: 0.8] [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
|
36
|
Goemann IM, Marczyk VR, Romitti M, Wajner SM, Maia AL. Current concepts and challenges to unravel the role of iodothyronine deiodinases in human neoplasias. Endocr Relat Cancer 2018; 25:R625-R645. [PMID: 30400023 DOI: 10.1530/erc-18-0097] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Thyroid hormones (THs) are essential for the regulation of several metabolic processes and the energy consumption of the organism. Their action is exerted primarily through interaction with nuclear receptors controlling the transcription of thyroid hormone-responsive genes. Proper regulation of TH levels in different tissues is extremely important for the equilibrium between normal cellular proliferation and differentiation. The iodothyronine deiodinases types 1, 2 and 3 are key enzymes that perform activation and inactivation of THs, thus controlling TH homeostasis in a cell-specific manner. As THs seem to exert their effects in all hallmarks of the neoplastic process, dysregulation of deiodinases in the tumoral context can be critical to the neoplastic development. Here, we aim at reviewing the deiodinases expression in different neoplasias and exploit the mechanisms by which they play an essential role in human carcinogenesis. TH modulation by deiodinases and other classical pathways may represent important targets with the potential to oppose the neoplastic process.
Collapse
Affiliation(s)
- Iuri Martin Goemann
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Vicente Rodrigues Marczyk
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Mirian Romitti
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Brussels, Belgium
| | - Simone Magagnin Wajner
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Luiza Maia
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
37
|
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: 194] [Impact Index Per Article: 27.7] [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
|
38
|
Vancamp P, Darras VM. From zebrafish to human: A comparative approach to elucidate the role of the thyroid hormone transporter MCT8 during brain development. Gen Comp Endocrinol 2018; 265:219-229. [PMID: 29183795 DOI: 10.1016/j.ygcen.2017.11.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
Abstract
Monocarboxylate transporter 8 (MCT8) facilitates transmembrane transport of thyroid hormones (THs) ensuring their action on gene expression during vertebrate neurodevelopment. A loss of MCT8 in humans results in severe psychomotor deficits associated with the Allan-Herndon-Dudley Syndrome (AHDS). However, where and when exactly a lack of MCT8 causes the neurological manifestations remains unclear because of the varying expression pattern of MCT8 between specific brain regions and cells. Here, we elaborate on the animal models that have been generated to elucidate the mechanisms underlying MCT8-deficient brain development. The absence of a clear neurological phenotype in Mct8 knockout mice made it clear that a single species would not suffice. The evolutionary conservation of TH action on neurodevelopment as well as the components regulating TH signalling however offers the opportunity to answer different aspects of MCT8 function in brain development using different vertebrate species. Moreover, the plethora of tools for genome editing available today facilitates gene silencing in these animals as well. Studies in the recently generated mct8-deficient zebrafish and Mct8/Oatp1c1 double knockout mice have put forward the current paradigm of impaired TH uptake at the level of the blood-brain barrier during peri- and postnatal development as being the main pathophysiological mechanism of AHDS. RNAi vector-based, cell-specific induction of MCT8 knockdown in the chicken embryo points to an additional function of MCT8 at the level of the neural progenitors during early brain development. Future studies including also additional in vivo models like Xenopus or in vitro approaches such as induced pluripotent stem cells will continue to help unravelling the exact role of MCT8 in developmental events. In the end, this multispecies approach will lead to a unifying thesis regarding the cellular and molecular mechanisms responsible for the neurological phenotype in AHDS patients.
Collapse
Affiliation(s)
- Pieter Vancamp
- KU Leuven, Laboratory of Comparative Endocrinology, Department of Biology, B-3000 Leuven, Belgium
| | - Veerle M Darras
- KU Leuven, Laboratory of Comparative Endocrinology, Department of Biology, B-3000 Leuven, Belgium.
| |
Collapse
|
39
|
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: 1.7] [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
|
40
|
Abstract
Thyroid hormone signaling is customized in a time and cell-specific manner by the deiodinases, homodimeric thioredoxin fold containing selenoproteins. This ensures adequate T3 action in developing tissues, healthy adults and many disease states. D2 activates thyroid hormone by converting the pro-hormone T4 to T3, the biologically active thyroid hormone. D2 expression is tightly regulated by transcriptional mechanisms triggered by endogenous as well as environmental cues. There is also an on/off switch mechanism that controls D2 activity that is triggered by catalysis and functions via D2 ubiquitination/deubiquitination. D3 terminates thyroid hormone action by inactivation of both T4 and T3 molecules. Deiodinases play a role in thyroid hormone homeostasis, development, growth and metabolic control by affecting the intracellular levels of T3 and thus gene expression on a cell-specific basis. In many cases, tight control of these pathways by T3 is achieved with coordinated reciprocal changes in D2-mediated thyroid hormone activation D3-mediated thyroid hormone inactivation.
Collapse
|
41
|
Lee JY, Kim MJ, Deliyanti D, Azari MF, Rossello F, Costin A, Ramm G, Stanley EG, Elefanty AG, Wilkinson-Berka JL, Petratos S. Overcoming Monocarboxylate Transporter 8 (MCT8)-Deficiency to Promote Human Oligodendrocyte Differentiation and Myelination. EBioMedicine 2017; 25:122-135. [PMID: 29111262 PMCID: PMC5704066 DOI: 10.1016/j.ebiom.2017.10.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/05/2017] [Accepted: 10/16/2017] [Indexed: 01/09/2023] Open
Abstract
Cell membrane thyroid hormone (TH) transport can be facilitated by the monocarboxylate transporter 8 (MCT8), encoded by the solute carrier family 16 member 2 (SLC16A2) gene. Human mutations of the gene, SLC16A2, result in the X-linked-inherited psychomotor retardation and hypomyelination disorder, Allan-Herndon-Dudley syndrome (AHDS). We posited that abrogating MCT8-dependent TH transport limits oligodendrogenesis and myelination. We show that human oligodendrocytes (OL), derived from the NKX2.1-GFP human embryonic stem cell (hESC) reporter line, express MCT8. Moreover, treatment of these cultures with DITPA (an MCT8-independent TH analog), up-regulates OL differentiation transcription factors and myelin gene expression. DITPA promotes hESC-derived OL myelination of retinal ganglion axons in co-culture. Pharmacological and genetic blockade of MCT8 induces significant OL apoptosis, impairing myelination. DITPA treatment limits OL apoptosis mediated by SLC16A2 down-regulation primarily signaling through AKT phosphorylation, driving myelination. Our results highlight the potential role of MCT8 in TH transport for human OL development and may implicate DITPA as a promising treatment for developmentally-regulated myelination in AHDS. NKX2.1-based sorting enhances OL derivation from hESC MCT8 is required for the survival of OL precursor cells DITPA promotes OL differentiation and myelination DITPA overrides SLC16A2 (MCT8) down-regulation to potentiate myelination
Thyroid hormone is vital for oligodendrocyte differentiation and myelination. Lee and colleagues show that MCT8 is an integral thyroid hormone transporter for oligodendrocytes derived from human embryonic stem cells. Knockdown of this transporter induces apoptosis of OLs, which could be prevented by the provision of DITPA.
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia
| | - Min Joung Kim
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia
| | - Devy Deliyanti
- Department of Diabetes, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia
| | - Michael F Azari
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia
| | - Fernando Rossello
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Adam Costin
- The Clive & Vera Ramaciotti Centre for Cryo Electron Microscopy, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Georg Ramm
- The Clive & Vera Ramaciotti Centre for Cryo Electron Microscopy, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Edouard G Stanley
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Rd, Parkville, Victoria 3052, Australia
| | - Andrew G Elefanty
- Murdoch Children's Research Institute, The Royal Children's Hospital, Flemington Rd, Parkville, Victoria 3052, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | | | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia.
| |
Collapse
|
42
|
Kersseboom S, van Gucht ALM, van Mullem A, Brigante G, Farina S, Carlsson B, Donkers JM, van de Graaf SFJ, Peeters RP, Visser TJ. Role of the Bile Acid Transporter SLC10A1 in Liver Targeting of the Lipid-Lowering Thyroid Hormone Analog Eprotirome. Endocrinology 2017; 158:3307-3318. [PMID: 28938430 DOI: 10.1210/en.2017-00433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/14/2017] [Indexed: 12/20/2022]
Abstract
The thyroid hormone (TH) analog eprotirome (KB2115) was developed to lower cholesterol through selective activation of the TH receptor (TR) β1 in the liver. Interestingly, eprotirome shows low uptake in nonhepatic tissues, explaining its lipid-lowering action without adverse extrahepatic thyromimetic effects. Clinical trials have shown marked decreases in serum cholesterol levels. We explored the transport of eprotirome across the plasma membrane by members of three TH transporter families: monocarboxylate transporters MCT8 and MCT10; Na-independent organic anion transporters 1A2, 1B1, 1B3, 1C1, 2A1, and 2B1; and Na-dependent organic anion transporters SLC10A1 to SLC10A7. Cellular transport was studied in transfected COS1 cells using [14C]eprotirome and [125I]TH analogs. Of the 15 transporters tested initially, the liver-specific bile acid transporter SLC10A1 showed the highest eprotirome uptake (greater than a sevenfold induction after 60 minutes) as well as TRβ1-mediated transcriptional activity. Uptake of eprotirome by SLC10A1 was Na+ dependent and saturable with a Michaelis constant of 8 μM. Eprotirome transport was inhibited by known substrates for SLC10A1 (e.g., cholate and taurocholate), and by TH analogs such as triiodothyropropionic acid and triiodothyroacetic acid. However, no significant SLC10A1-mediated transport was observed of these [125I]TH analogs. We also studied the plasma disappearance and biliary excretion of [14C]eprotirome injected in control and Slc10a1 knockout mice. Although eprotirome is also transported by mouse Slc10a1, the pharmacokinetics of eprotirome were not affected by Slc10a1 deficiency. In conclusion, we have demonstrated that the liver-specific bile acid transporter SLC10A1 effectively transports eprotirome. However, Slc10a1 does not appear to be critical for the liver targeting of this TH analog in mice. Therefore, the importance of SLC10A1 for liver uptake of eprotirome in humans remains to be elucidated.
Collapse
Affiliation(s)
- Simone Kersseboom
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Anja L M van Gucht
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Alies van Mullem
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Giulia Brigante
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Stefania Farina
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Bo Carlsson
- Karo Bio AB, Novum Research Park, Huddinge S-141 57, Sweden
| | - Joanne M Donkers
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Amsterdam Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research and Department of Gastroenterology and Hepatology, Amsterdam Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Robin P Peeters
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Theo J Visser
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| |
Collapse
|
43
|
Groeneweg S, Lima de Souza EC, Meima ME, Peeters RP, Visser WE, Visser TJ. Outward-Open Model of Thyroid Hormone Transporter Monocarboxylate Transporter 8 Provides Novel Structural and Functional Insights. Endocrinology 2017; 158:3292-3306. [PMID: 28977587 DOI: 10.1210/en.2017-00082] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 07/06/2017] [Indexed: 12/28/2022]
Abstract
Monocarboxylate transporter 8 (MCT8) facilitates cellular uptake and efflux of thyroid hormone (TH). Mutations in MCT8 result in severe intellectual and motor disability known as the Allan-Herndon-Dudley syndrome (AHDS). Previous studies have provided valuable insights into the putative mechanism of substrate binding in the inward-open conformation, required for TH efflux. The current study aims to delineate the mechanism of substrate binding in the outward-open conformation, required for TH uptake. Extensive chemical modification and site-directed mutagenesis studies were used to guide protein homology modeling of MCT8 in the outward-open conformation. Arg271 and Arg445 were modified by phenylglyoxal, which was partially prevented in the presence of substrate. Substrate docking in our outward-open model suggested an important role for His192 and Arg445 in substrate binding. Interestingly, mutations affecting these residues have been identified in patients who have AHDS. In addition, our outward-open model predicted the location of Phe189, Met227, Phe279, Gly282, Phe287, and Phe501 at the substrate-binding center, and their Ala substitution differentially affected the apparent Vmax and Km of T3 transport, with F189A, F279A, and F287A showing the highest impact. Thus, here we present an MCT8 homology model in the outward-open conformation, which supports the important role of His192 and Arg445 in substrate docking and identifies critical residues at the putative substrate-binding center. Our findings provide insights into MCT8 structure and function, which add to our understanding of the pathogenic mechanism of mutations found in patients who have AHDS and can be used to screen for novel substrates and inhibitors.
Collapse
Affiliation(s)
- Stefan Groeneweg
- The Rotterdam Thyroid Center & Department of Internal Medicine, Erasmus Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Elaine C Lima de Souza
- The Rotterdam Thyroid Center & Department of Internal Medicine, Erasmus Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Marcel E Meima
- The Rotterdam Thyroid Center & Department of Internal Medicine, Erasmus Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Robin P Peeters
- The Rotterdam Thyroid Center & Department of Internal Medicine, Erasmus Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - W Edward Visser
- The Rotterdam Thyroid Center & Department of Internal Medicine, Erasmus Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Theo J Visser
- The Rotterdam Thyroid Center & Department of Internal Medicine, Erasmus Medical Center, 3015 CN, Rotterdam, The Netherlands
| |
Collapse
|
44
|
Badziong J, Ting S, Synoracki S, Tiedje V, Brix K, Brabant G, Moeller LC, Schmid KW, Fuhrer D, Zwanziger D. Differential regulation of monocarboxylate transporter 8 expression in thyroid cancer and hyperthyroidism. Eur J Endocrinol 2017; 177:243-250. [PMID: 28576880 DOI: 10.1530/eje-17-0279] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/08/2017] [Accepted: 06/02/2017] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Thyroid hormone (TH) transporters are expressed in thyrocytes and most play a role in TH release. We asked whether expression of the monocarboxylate transporter 8 (MCT8) and the L-type amino acid transporters LAT2 and LAT4 is changed with thyrocyte dedifferentiation and in hyperfunctioning thyroid tissues. DESIGN AND METHODS Protein expression and localization of transporters was determined by immunohistochemistry in human thyroid specimen including normal thyroid tissue (NT, n = 19), follicular adenoma (FA, n = 44), follicular thyroid carcinoma (FTC, n = 45), papillary thyroid carcinoma (PTC, n = 40), anaplastic thyroid carcinoma (ATC, n = 40) and Graves' disease (GD, n = 50) by calculating the 'hybrid' (H) score. Regulation of transporter expression was investigated in the rat follicular thyroid cell line PCCL3 under basal and thyroid stimulating hormone (TSH) conditions. RESULTS MCT8 and LAT4 were localized at the plasma membrane, while LAT2 transporter showed cytoplasmic localization. MCT8 expression was downregulated in benign and malignant thyroid tumours as compared to NT. In contrast, significant upregulation of MCT8, LAT2 and LAT4 was found in GD. Furthermore, a stronger expression of MCT8 was demonstrated in PCCL3 cells after TSH stimulation. CONCLUSIONS Downregulation of MCT8 in thyroid cancers qualifies MCT8 as a marker of thyroid differentiation. The more variable expression of LATs in distinct thyroid malignancies may be linked with other transporter properties relevant to altered metabolism in cancer cells, i.e. amino acid transport. Consistent upregulation of MCT8 in GD is in line with increased TH release in hyperthyroidism, an assumption supported by our in vitro results showing TSH-dependent upregulation of MCT8.
Collapse
Affiliation(s)
- Julia Badziong
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| | - Saskia Ting
- University Hospital Essen, Institute of Pathology, Essen, Germany
| | - Sarah Synoracki
- University Hospital Essen, Institute of Pathology, Essen, Germany
| | - Vera Tiedje
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Georg Brabant
- University Hospital Schleswig-Holstein, Experimental and Clinical Endocrinology, Lübeck, Germany
| | - Lars Christian Moeller
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| | | | - Dagmar Fuhrer
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| | - Denise Zwanziger
- Department of Endocrinology and Metabolism and Division of Laboratory Research, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
45
|
Mughal BB, Leemans M, Lima de Souza EC, le Mevel S, Spirhanzlova P, Visser TJ, Fini JB, Demeneix BA. Functional Characterization of Xenopus Thyroid Hormone Transporters mct8 and oatp1c1. Endocrinology 2017; 158:2694-2705. [PMID: 28591769 DOI: 10.1210/en.2017-00108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022]
Abstract
Xenopus is an excellent model for studying thyroid hormone signaling as it undergoes thyroid hormone-dependent metamorphosis. Despite the fact that receptors and deiodinases have been described in Xenopus, membrane transporters for these hormones are yet to be characterized. We cloned Xenopus monocarboxylate transporter 8 (mct8) and organic anion-transporting polypeptide 1C1 (oatpc1c1), focusing on these two transporters given their importance for vertebrate brain development. Protein alignment and bootstrap analysis showed that Xenopus mct8 and oatp1c1 are closer to their mammalian orthologs than their teleost counterparts. We functionally characterized the two transporters using a radiolabeled hormones in vitro uptake assay in COS-1 cells. Xenopus mct8 was found to actively transport both T3 and T4 bidirectionally. As to the thyroid precursor molecules, diiodotyrosine (DIT) and monoiodotyrosine (MIT), both human and Xenopus mct8, showed active efflux, but no influx. Again similar to humans, Xenopus oatp1c1 transported T4 but not T3, MIT, or DIT. We used reverse transcription quantitative polymerase chain reaction and in situ hybridization to characterize the temporal and spatial expression of mct8 and oatp1c1 in Xenopus. Specific expression of the transporter was observed in the brain, with increasingly strong expression as development progressed. In conclusion, these results show that Xenopus thyroid hormone transporters are functional and display marked spatiotemporal expression patterns. These features make them interesting targets to elucidate their roles in determining thyroid hormone availability during embryonic development.
Collapse
Affiliation(s)
- Bilal B Mughal
- Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Sorbonne Universities, 75005 Paris, France
| | - Michelle Leemans
- Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Sorbonne Universities, 75005 Paris, France
| | - Elaine C Lima de Souza
- Department of Internal Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Sébastien le Mevel
- Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Sorbonne Universities, 75005 Paris, France
| | - Petra Spirhanzlova
- Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Sorbonne Universities, 75005 Paris, France
| | - Theo J Visser
- Department of Internal Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Jean-Baptiste Fini
- Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Sorbonne Universities, 75005 Paris, France
| | - Barbara A Demeneix
- Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Sorbonne Universities, 75005 Paris, France
| |
Collapse
|
46
|
Dong H, Wade MG. Application of a nonradioactive assay for high throughput screening for inhibition of thyroid hormone uptake via the transmembrane transporter MCT8. Toxicol In Vitro 2017; 40:234-242. [DOI: 10.1016/j.tiv.2017.01.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 12/11/2022]
|
47
|
Abstract
Transport of thyroid hormone (TH) across the plasma membrane is essential for intracellular TH metabolism and action, and this is mediated by specific transporter proteins. During the last two decades several transporters capable of transporting TH have been identified, including monocarboxylate transporter 8 (MCT8), MCT10 and organic anion transporting polypeptide 1C1 (OATP1C1). In particular MCT8 and OATP1C1 are important for the regulation of local TH activity in the brain and thus for brain development. MCT8 is a protein containing 12 transmembrane domains, and is encoded by the SLC16A2 gene located on the X chromosome. It facilitates both TH uptake and efflux across the cell membrane. Male subjects with hemizygous mutations in MCT8 are afflicted with severe intellectual and motor disability, also known as the Allan-Herndon-Dudley syndrome (AHDS), which goes together with low serum T4 and high T3 levels. This review concerns molecular and clinical aspects of MCT8 function.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - W Edward Visser
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Theo J Visser
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
48
|
Lee D, Martinez B, Crocker DE, Ortiz RM. Fasting increases the phosphorylation of AMPK and expression of sirtuin1 in muscle of adult male northern elephant seals ( Mirounga angustirostris). Physiol Rep 2017; 5:5/4/e13114. [PMID: 28242816 PMCID: PMC5328766 DOI: 10.14814/phy2.13114] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/08/2016] [Accepted: 12/11/2016] [Indexed: 12/12/2022] Open
Abstract
Fasting typically suppresses thyroid hormone (TH)‐mediated cellular events and increases sirtuin 1 (SIRT1) activity. THs may regulate metabolism through nongenomic pathways and directly through activation of adenosine monophosphate‐activated protein kinase (AMPK). Adult male elephant seals (Mirounga angustirostris) are active, hypermetabolic, and normothermic during their annual breeding fast, which is characterized by stable TH levels. However, the contribution of TH to maintenance of their fasting metabolism is unknown. To investigate the fasting effects on cellular TH‐mediated events and its potential association with SIRT1 and AMPK, we quantified plasma TH levels, mRNA expressions of muscle SIRT1 and TH‐associated genes as well as the phosphorylation of AMPK in adult, male northern elephant seals (n = 10/fasting period) over 8 weeks of fasting (early vs. late). Deiodinase type I (DI1) expression increased twofold with fasting duration suggesting that the potential for TH‐mediated cellular signaling is increased. AMPK phosphorylation increased 61 ± 21% with fasting suggesting that cellular metabolism is increased. The mRNA expression of the TH transporter, monocarboxylate transporter 10 (MCT10), increased 2.4‐fold and the TH receptor (THrβ‐1) decreased 30‐fold suggesting that cellular uptake of T4 is increased, but its subsequent cellular effects such as activation of AMPK are likely nongenomic. The up‐regulation of SIRT1 mRNA expression (2.6‐fold) likely contributes to the nongenomic activation of AMPK by TH, which may be necessary to maintain the expression of PGC‐1α. These coordinated changes likely contribute to the up‐regulation of mitochondrial metabolism to support the energetic demands associated with prolonged fasting in adult seals.
Collapse
Affiliation(s)
- Debby Lee
- Department of Cellular and Molecular Biology, University of California, Merced, California
| | - Bridget Martinez
- Department of Cellular and Molecular Biology, University of California, Merced, California
| | - Daniel E Crocker
- Department of Biology, Sonoma State University, Rohnert Park, California
| | - Rudy M Ortiz
- Department of Cellular and Molecular Biology, University of California, Merced, California
| |
Collapse
|
49
|
Hellsten SV, Lekholm E, Ahmad T, Fredriksson R. The gene expression of numerous SLC transporters is altered in the immortalized hypothalamic cell line N25/2 following amino acid starvation. FEBS Open Bio 2017; 7:249-264. [PMID: 28174690 PMCID: PMC5292668 DOI: 10.1002/2211-5463.12181] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 12/20/2022] Open
Abstract
Amino acids are known to play a key role in gene expression regulation, and in mammalian cells, amino acid signaling is mainly mediated via two pathways, the mammalian target of rapamycin complex 1 (mTORC1) pathway and the amino acid responsive (AAR) pathway. It is vital for cells to have a system to sense amino acid levels, in order to control protein and amino acid synthesis and catabolism. Amino acid transporters are crucial in these pathways, due to both their sensing and transport functions. In this large-scale study, an immortalized mouse hypothalamic cell line (N25/2) was used to study the gene expression changes following 1, 2, 3, 5 or 16 h of amino acid starvation. We focused on genes encoding solute carriers (SLCs) and putative SLCs, more specifically on amino acid transporters. The microarray contained 28 270 genes and 86.2% of the genes were expressed in the cell line. At 5 h of starvation, 1001 genes were upregulated and 848 genes were downregulated, and among these, 47 genes from the SLC superfamily or atypical SLCs were found. Of these, 15 were genes encoding amino acid transporters and 32 were genes encoding other SLCs or atypical SLCs. Increased expression was detected for genes encoding amino acid transporters from system A, ASC, L, N, T, xc-, and y+. Using GO annotations, genes involved in amino acid transport and amino acid transmembrane transporter activity were found to be most upregulated at 3 h and 5 h of starvation.
Collapse
Affiliation(s)
- Sofie V Hellsten
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology Uppsala University Sweden; Department of Neuroscience, Functional Pharmacology Uppsala University Sweden
| | - Emilia Lekholm
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology Uppsala University Sweden
| | - Tauseef Ahmad
- Department of Neuroscience, Functional Pharmacology Uppsala University Sweden
| | - Robert Fredriksson
- Department of Pharmaceutical Bioscience, Molecular Neuropharmacology Uppsala University Sweden
| |
Collapse
|
50
|
van Mullem AA, van Gucht ALM, Visser WE, Meima ME, Peeters RP, Visser TJ. Effects of thyroid hormone transporters MCT8 and MCT10 on nuclear activity of T3. Mol Cell Endocrinol 2016; 437:252-260. [PMID: 27492966 DOI: 10.1016/j.mce.2016.07.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/10/2016] [Accepted: 07/27/2016] [Indexed: 11/22/2022]
Abstract
Transport of thyroid hormone (TH) across the plasma membrane is necessary for the genomic action of T3 mediated by its nuclear T3 receptor. MCT8 and MCT10 have been identified as important TH transporters. Mutations in MCT8 result in severe psychomotor retardation. In addition to TH transport into the cell, MCT8 and MCT10 also facilitate TH efflux from cells. Therefore, the aim of this study was to examine if MCT8 and MCT10 increase the availability of T3 for its nuclear receptor rather than generate a rapid equilibrium between cellular and serum T3. T3 action was investigated in JEG3 cells co-transfected with TRβ1 and a T3 response element-driven luciferase construct, and T3 metabolism was analyzed in cells transfected with type 3 deiodinase (D3). In addition, cells were transfected with MCT8 or MCT10 and/or the cytoplasmic T3-binding protein mu-crystallin (CRYM). Luciferase signal was markedly stimulated by incubating cells for 24 h with 1 nM T3, but this response was not augmented by MCT8 or MCT10 expression. Limiting the time of T3 exposure to 1-6 h and co-transfection with CRYM allowed for a modest increase in luciferase response to T3. In contrast, T3 metabolism by D3 was potently stimulated by MCT8 or MCT10 expression, but it was not affected by expression of CRYM. These results suggest that MCT8 and MCT10 by virtue of their bidirectional T3 transport have less effect on steady-state nuclear T3 levels than on T3 levels at the cell periphery where D3 is located. CRYM alters the dynamics of cellular TH transport but its exact function in the cellular distribution of TH remains to be determined.
Collapse
Affiliation(s)
- Alies A van Mullem
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Anja L M van Gucht
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - W Edward Visser
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marcel E Meima
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robin P Peeters
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Theo J Visser
- Department of Internal Medicine and Rotterdam Thyroid Center, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|