1
|
Beltrão FEDL, Beltrão DCDA, Carvalhal G, Beltrão FLDL, Oliveira JDB, Silva HDS, Teixeira HMP, Rodrigues JL, de Figueiredo CAV, Costa RDS, Hecht F, Vieira GC, Gonçalves MDCR, Bianco AC, Ramos HE. Thr92Ala-DIO2 heterozygosity is associated with skeletal muscle mass and myosteatosis in patients with COVID-19. Eur Thyroid J 2024; 13:e240068. [PMID: 38869458 PMCID: PMC11301567 DOI: 10.1530/etj-24-0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/13/2024] [Indexed: 06/14/2024] Open
Abstract
Introduction The type 2 deiodinase and its Thr92Ala-DIO2 polymorphism have been linked to clinical outcomes in acute lung injury and coronavirus disease 2019 (COVID-19). Objective The objective was to identify a potential association between Thr92Ala-DIO2 polymorphism and body composition (appendicular muscle mass, myosteatosis, and fat distribution) and to determine whether they reflect the severity or mortality associated with the disease. Methods In this prospective cohort study (June-August 2020), 181 patients hospitalized with moderate-to-severe COVID-19 underwent a non-contrast-enhanced computed tomography (CT) of the thorax to assess body composition, laboratory tests, and genotyping for the Thr92Ala-DIO2 polymorphism. Results In total, 181 consecutive patients were stratified into three subgroups according to the genotype: Thr/Thr (n = 64), Thr/Ala (n = 96), and Ala/Ala (n = 21). The prevalence of low muscle area (MA) (< 92 cm²) was 52.5%. Low MA was less frequent in Ala/Thr patients (44.8%) than in Thr/Thr (60.9%) or Ala/Ala patients (61.9%) (P = 0.027). Multivariate logistic regression analysis confirmed that the Thr/Ala allele was associated with a reduced risk of low MA (41% to 69%) and myosteatosis (62% to 72%) compared with Thr/Thr + Ala/Ala (overdominant model). Kaplan-Meier curves showed that patients with low muscle mass and homozygosity had lower survival rates than the other groups. Notably, the heterozygotes with MA ≥92 cm² exhibited the best survival rate. Conclusion Thr92Ala-DIO2 heterozygosity is associated with increased skeletal MA and less myosteatosis in patients with COVID-19. The protective effect of Thr92Ala-DIO2 heterozygosity on COVID-19 mortality is restricted to patients with reduced MA.
Collapse
Affiliation(s)
- Fabyan Esberard de Lima Beltrão
- Lauro Wanderley University Hospital, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
- University Center of João Pessoa – UNIPE, João Pessoa, PB, Brazil
| | - Daniele Carvalhal de Almeida Beltrão
- University Center of João Pessoa – UNIPE, João Pessoa, PB, Brazil
- Post-Graduation Program in Cognitive Neuroscience and Behavior, Psychology Department of the Center of Human Sciences, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Giulia Carvalhal
- Center for Biological and Health Sciences, Federal University of Campina Grande, Campina Grande, Paraíba, Brazil
| | | | - Jocyel de Brito Oliveira
- Bioregulation Department, Health and Science Institut, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Hatilla dos Santos Silva
- Bioregulation Department, Health and Science Institut, Federal University of Bahia, Salvador, Bahia, Brazil
| | | | - Juliana Lopes Rodrigues
- Laboratory of Immunopharmacology and Molecular Biology, Health Sciences Institute, Federal University of Bahia, Brazil
| | | | - Ryan dos Santos Costa
- Laboratory of Immunopharmacology and Molecular Biology, Health Sciences Institute, Federal University of Bahia, Brazil
| | - Fabio Hecht
- The Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giciane Carvalho Vieira
- Post-Graduation Program in Cognitive Neuroscience and Behavior, Psychology Department of the Center of Human Sciences, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | | | - Antonio C. Bianco
- Section of Endocrinology and Metabolism, Division of the Biological Sciences, University of Chicago, Chicago, Illinois, USA
| | - Helton Estrela Ramos
- Post-Graduate Program in Medicine and Health, Medical School of Medicine, Federal University of Bahia, Salvador, Brazil
- Postgraduate Program in Interactive Processes of Organs and Systems, Health & Science Institute, Federal University of Bahia, Salvador, BA, Brazil
| |
Collapse
|
2
|
Huttunen J, Adla SK, Markowicz-Piasecka M, Huttunen KM. Increased/Targeted Brain (Pro)Drug Delivery via Utilization of Solute Carriers (SLCs). Pharmaceutics 2022; 14:pharmaceutics14061234. [PMID: 35745806 PMCID: PMC9228667 DOI: 10.3390/pharmaceutics14061234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Membrane transporters have a crucial role in compounds’ brain drug delivery. They allow not only the penetration of a wide variety of different compounds to cross the endothelial cells of the blood–brain barrier (BBB), but also the accumulation of them into the brain parenchymal cells. Solute carriers (SLCs), with nearly 500 family members, are the largest group of membrane transporters. Unfortunately, not all SLCs are fully characterized and used in rational drug design. However, if the structural features for transporter interactions (binding and translocation) are known, a prodrug approach can be utilized to temporarily change the pharmacokinetics and brain delivery properties of almost any compound. In this review, main transporter subtypes that are participating in brain drug disposition or have been used to improve brain drug delivery across the BBB via the prodrug approach, are introduced. Moreover, the ability of selected transporters to be utilized in intrabrain drug delivery is discussed. Thus, this comprehensive review will give insights into the methods, such as computational drug design, that should be utilized more effectively to understand the detailed transport mechanisms. Moreover, factors, such as transporter expression modulation pathways in diseases that should be taken into account in rational (pro)drug development, are considered to achieve successful clinical applications in the future.
Collapse
Affiliation(s)
- Johanna Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
| | - Santosh Kumar Adla
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
- Institute of Organic Chemistry and Biochemistry (IOCB), Czech Academy of Sciences, Flemingovo Namesti 542/2, 160 00 Prague, Czech Republic
| | - Magdalena Markowicz-Piasecka
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland;
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland; (J.H.); (S.K.A.)
- Correspondence:
| |
Collapse
|
3
|
Yellajoshyula D, Pappas SS, Dauer WT. Oligodendrocyte and Extracellular Matrix Contributions to Central Nervous System Motor Function: Implications for Dystonia. Mov Disord 2022; 37:456-463. [PMID: 34989453 PMCID: PMC11152458 DOI: 10.1002/mds.28892] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
The quest to elucidate nervous system function and dysfunction in disease has focused largely on neurons and neural circuits. However, fundamental aspects of nervous system development, function, and plasticity are regulated by nonneuronal elements, including glial cells and the extracellular matrix (ECM). The rapid rise of genomics and neuroimaging techniques in recent decades has highlighted neuronal-glial interactions and ECM as a key component of nervous system development, plasticity, and function. Abnormalities of neuronal-glial interactions have been understudied but are increasingly recognized to play a key role in many neurodevelopmental disorders. In this report, we consider the role of myelination and the ECM in the development and function of central nervous system motor circuits and the neurodevelopmental disease dystonia. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Samuel S Pappas
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - William T Dauer
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
4
|
Wolff TM, Veil C, Dietrich JW, Müller MA. Mathematical modeling and simulation of thyroid homeostasis: Implications for the Allan-Herndon-Dudley syndrome. Front Endocrinol (Lausanne) 2022; 13:882788. [PMID: 36568087 PMCID: PMC9772020 DOI: 10.3389/fendo.2022.882788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 10/31/2022] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION A mathematical model of the pituitary-thyroid feedback loop is extended to deepen the understanding of the Allan-Herndon-Dudley syndrome (AHDS). The AHDS is characterized by unusual thyroid hormone concentrations and a mutation in the SLC16A2 gene encoding for the monocarboxylate transporter 8 (MCT8). This mutation leads to a loss of thyroid hormone transport activity. One hypothesis to explain the unusual hormone concentrations of AHDS patients is that due to the loss of thyroid hormone transport activity, thyroxine (T 4) is partially retained in thyroid cells. METHODS This hypothesis is investigated by extending a mathematical model of the pituitary-thyroid feedback loop to include a model of the net effects of membrane transporters such that the thyroid hormone transport activity can be considered. A nonlinear modeling approach based on the Michaelis-Menten kinetics and its linear approximation are employed to consider the membrane transporters. The unknown parameters are estimated through a constrained parameter optimization. RESULTS In dynamic simulations, damaged membrane transporters result in a retention of T 4 in thyroid cells and ultimately in the unusual hormone concentrations of AHDS patients. The Michaelis-Menten modeling approach and its linear approximation lead to similar results. DISCUSSION The results support the hypothesis that a partial retention of T 4 in thyroid cells represents one mechanism responsible for the unusual hormone concentrations of AHDS patients. Moreover, our results suggest that the retention of T 4 in thyroid cells could be the main reason for the unusual hormone concentrations of AHDS patients.
Collapse
Affiliation(s)
- Tobias M. Wolff
- Institute of Automatic Control, Leibniz University Hannover, Hannover, Germany
- *Correspondence: Tobias M. Wolff,
| | - Carina Veil
- Institute for System Dynamics, University of Stuttgart, Stuttgart, Germany
| | - Johannes W. Dietrich
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
- Diabetes Centre Bochum-Hattingen, St. Elisabeth-Hospital Blankenstein, Hattingen, Germany
- Ruhr Center for RareDiseases (CeSER), Ruhr University of Bochum and Witten/Herdecke University, Bochum, Germany
| | - Matthias A. Müller
- Institute of Automatic Control, Leibniz University Hannover, Hannover, Germany
| |
Collapse
|
5
|
Pagnin M, Kondos-Devcic D, Chincarini G, Cumberland A, Richardson SJ, Tolcos M. Role of thyroid hormones in normal and abnormal central nervous system myelination in humans and rodents. Front Neuroendocrinol 2021; 61:100901. [PMID: 33493504 DOI: 10.1016/j.yfrne.2021.100901] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/07/2021] [Accepted: 01/16/2021] [Indexed: 12/13/2022]
Abstract
Thyroid hormones (THs) are instrumental in promoting the molecular mechanisms which underlie the complex nature of neural development and function within the central nervous system (CNS) in vertebrates. The key neurodevelopmental process of myelination is conserved between humans and rodents, of which both experience peak fetal TH concentrations concomitant with onset of myelination. The importance of supplying adequate levels of THs to the myelin producing cells, the oligodendrocytes, for promoting their maturation is crucial for proper neural function. In this review we examine the key TH distributor and transport proteins, including transthyretin (TTR) and monocarboxylate transporter 8 (MCT8), essential for supporting proper oligodendrocyte and myelin health; and discuss disorders with impaired TH signalling in relation to abnormal CNS myelination in humans and rodents. Furthermore, we explore the importance of using novel TH analogues in the treatment of myelination disorders associated with abnormal TH signalling.
Collapse
Affiliation(s)
- Maurice Pagnin
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Delphi Kondos-Devcic
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Ginevra Chincarini
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Angela Cumberland
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | | | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia.
| |
Collapse
|
6
|
van Geest FS, Meima ME, Stuurman KE, Wolf NI, van der Knaap MS, Lorea CF, Poswar FO, Vairo F, Brunetti-Pierri N, Cappuccio G, Bakhtiani P, de Munnik SA, Peeters RP, Visser WE, Groeneweg S. Clinical and Functional Consequences of C-Terminal Variants in MCT8: A Case Series. J Clin Endocrinol Metab 2021; 106:539-553. [PMID: 33141165 PMCID: PMC7823235 DOI: 10.1210/clinem/dgaa795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Indexed: 12/17/2022]
Abstract
CONTEXT Genetic variants in SLC16A2, encoding the thyroid hormone transporter MCT8, can cause intellectual and motor disability and abnormal serum thyroid function tests, known as MCT8 deficiency. The C-terminal domain of MCT8 is poorly conserved, which complicates prediction of the deleteriousness of variants in this region. We studied the functional consequences of 5 novel variants within this domain and their relation to the clinical phenotypes. METHODS We enrolled male subjects with intellectual disability in whom genetic variants were identified in exon 6 of SLC16A2. The impact of identified variants was evaluated in transiently transfected cell lines and patient-derived fibroblasts. RESULTS Seven individuals from 5 families harbored potentially deleterious variants affecting the C-terminal domain of MCT8. Two boys with clinical features considered atypical for MCT8 deficiency had a missense variant [c.1724A>G;p.(His575Arg) or c.1796A>G;p.(Asn599Ser)] that did not affect MCT8 function in transfected cells or patient-derived fibroblasts, challenging a causal relationship. Two brothers with classical MCT8 deficiency had a truncating c.1695delT;p.(Val566*) variant that completely inactivated MCT8 in vitro. The 3 other boys had relatively less-severe clinical features and harbored frameshift variants that elongate the MCT8 protein [c.1805delT;p.(Leu602HisfsTer680) and c.del1826-1835;p.(Pro609GlnfsTer676)] and retained ~50% residual activity. Additional truncating variants within transmembrane domain 12 were fully inactivating, whereas those within the intracellular C-terminal tail were tolerated. CONCLUSIONS Variants affecting the intracellular C-terminal tail of MCT8 are likely benign unless they cause frameshifts that elongate the MCT8 protein. These findings provide clinical guidance in the assessment of the pathogenicity of variants within the C-terminal domain of MCT8.
Collapse
Affiliation(s)
- Ferdy S van Geest
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - Marcel E Meima
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - Kyra E Stuurman
- Department of Clinical Genetics, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - Nicole I Wolf
- Department of Pediatric Neurology, Emma Children’s Hospital, Amsterdam University Medical Centre, AZ Amsterdam, Netherlands
- Amsterdam Neuroscience, HV Amsterdam, Netherlands
| | - Marjo S van der Knaap
- Department of Pediatric Neurology, Emma Children’s Hospital, Amsterdam University Medical Centre, AZ Amsterdam, Netherlands
- Amsterdam Neuroscience, HV Amsterdam, Netherlands
| | - Cláudia F Lorea
- Teaching Hospital of Universidade Federal de Pelotas, Brazil
| | - Fabiano O Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Filippo Vairo
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | - Gerarda Cappuccio
- Department of Translational Medicine, Federico II University, Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy
| | | | - Sonja A de Munnik
- Department of Human Genetics, Radboud University Medical Centre Nijmegen, GA Nijmegen, the Netherlands
| | - Robin P Peeters
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - W Edward Visser
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| | - Stefan Groeneweg
- Academic Center For Thyroid Disease, Department of Internal Medicine, Erasmus Medical Center, GD Rotterdam, The Netherlands
| |
Collapse
|
7
|
Groeneweg S, van Geest FS, Peeters RP, Heuer H, Visser WE. Thyroid Hormone Transporters. Endocr Rev 2020; 41:5637505. [PMID: 31754699 DOI: 10.1210/endrev/bnz008] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ferdy S van Geest
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - W Edward Visser
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
8
|
Remerand G, Boespflug-Tanguy O, Tonduti D, Touraine R, Rodriguez D, Curie A, Perreton N, Des Portes V, Sarret C. Expanding the phenotypic spectrum of Allan-Herndon-Dudley syndrome in patients with SLC16A2 mutations. Dev Med Child Neurol 2019; 61:1439-1447. [PMID: 31410843 DOI: 10.1111/dmcn.14332] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2019] [Indexed: 01/01/2023]
Abstract
The aim of the study was to redefine the phenotype of Allan-Herndon-Dudley syndrome (AHDS), which is caused by mutations in the SLC16A2 gene that encodes the brain transporter of thyroid hormones. Clinical phenotypes, brain imaging, thyroid hormone profiles, and genetic data were compared to the existing literature. Twenty-four males aged 11 months to 29 years had a mutation in SLC16A2, including 12 novel mutations and five previously described mutations. Sixteen patients presented with profound developmental delay, three had severe intellectual disability with poor language and walking with an aid, four had moderate intellectual disability with language and walking abilities, and one had mild intellectual disability with hypotonia. Overall, eight had learned to walk, all had hypotonia, 17 had spasticity, 18 had dystonia, 12 had choreoathetosis, 19 had hypomyelination, and 10 had brain atrophy. Kyphoscoliosis (n=12), seizures (n=7), and pneumopathies (n=5) were the most severe complications. This study extends the phenotypic spectrum of AHDS to a mild intellectual disability with hypotonia. Developmental delay, hypotonia, hypomyelination, and thyroid hormone profile help to diagnose patients. Clinical course depends on initial severity, with stable acquisition after infancy; this may be adversely affected by neuro-orthopaedic, pulmonary, and epileptic complications. WHAT THIS PAPER ADDS: Mild intellectual disability is associated with SLC16A2 mutations. A thyroid hormone profile with a free T3 /T4 ratio higher than 0.75 can help diagnose patients. Patients with SLC16A2 mutations present a broad spectrum of neurological phenotypes that are also observed in other hypomyelinating disorders. Axial hypotonia is a consistent feature of Allan-Herndon-Dudley syndrome and leads to specific complications.
Collapse
Affiliation(s)
- Ganaelle Remerand
- Centre de Compétence des Leucodystrophies et Leucoencéphalopathies de Cause Rare, Pôle Femme et Enfant, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Odile Boespflug-Tanguy
- Centre de Référence des Leucodystrophies et Leucoencéphalopathies de Cause Rare, Service de Neurologie Pédiatrique, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France.,NeuroDiderot, INSERM UMR1141, Université Paris Diderot, Paris, France
| | - Davide Tonduti
- Unit of Child Neurology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Unit of Child Neurology, V. Buzzi Children's Hospital, Milan, Italy
| | - Renaud Touraine
- Service de Génétique, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
| | - Diana Rodriguez
- Sorbonne Université, GRC no. 19, Pathologies Congénitales du Cervelet-LeucoDystrophies, Assistance Publique-Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France.,Centre de Référence Neurogénétique, Service de Neurologie Pédiatrique, Assistance Publique-Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Aurore Curie
- Centre de Référence des Déficiences Intellectuelles de Cause Rare, Service de Neurologie Pédiatrique, Centre Hospitalier Universitaire de Lyon, Hôpital Femme-Mère-Enfant, Lyon, France
| | - Nathalie Perreton
- CIC 1407Inserm, Centre Hospitalo-Universitaire de Lyon, Lyon, France
| | - Vincent Des Portes
- Centre de Référence des Déficiences Intellectuelles de Cause Rare, Service de Neurologie Pédiatrique, Centre Hospitalier Universitaire de Lyon, Hôpital Femme-Mère-Enfant, Lyon, France
| | - Catherine Sarret
- Centre de Compétence des Leucodystrophies et Leucoencéphalopathies de Cause Rare, Pôle Femme et Enfant, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France.,IGCNC, Université Clermont Auvergne, CNRS, SIGMA Clermont, Institut Pascal, Clermont-Ferrand, France
| | | |
Collapse
|
9
|
Kim MJ, Petratos S. Oligodendroglial Lineage Cells in Thyroid Hormone-Deprived Conditions. Stem Cells Int 2019; 2019:5496891. [PMID: 31182964 PMCID: PMC6515029 DOI: 10.1155/2019/5496891] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/20/2019] [Indexed: 01/06/2023] Open
Abstract
Oligodendrocytes are supporting glial cells that ensure the metabolism and homeostasis of neurons with specific synaptic axoglial interactions in the central nervous system. These require key myelinating glial trophic signals important for growth and metabolism. Thyroid hormone (TH) is one such trophic signal that regulates oligodendrocyte maturation, myelination, and oligodendroglial synaptic dynamics via either genomic or nongenomic pathways. The intracellular and extracellular transport of TH is facilitated by a specific transmembrane transporter known as the monocarboxylate transporter 8 (MCT8). Dysfunction of the MCT8 due to mutation, inhibition, or downregulation during brain development leads to inherited hypomyelination, which manifests as psychomotor retardation in the X-linked inherited Allan-Herndon-Dudley syndrome (AHDS). In particular, oligodendroglial-specific MCT8 deficiency may restrict the intracellular T3 availability, culminating in deficient metabolic communication between the oligodendrocytes and the neurons they ensheath, potentially promulgating neurodegenerative adult diseases such as multiple sclerosis (MS). Based on the therapeutic effects exhibited by TH in various preclinical studies, particularly related to its remyelinating potential, TH has now entered the initial stages of a clinical trial to test the therapeutic efficacy in relapsing-remitting MS patients (NCT02506751). However, TH analogs, such as DITPA or Triac, may well serve as future therapeutic options to rescue mature oligodendrocytes and/or promote oligodendrocyte precursor cell differentiation in an environment of MCT8 deficiency within the CNS. This review outlines the therapeutic strategies to overcome the differentiation blockade of oligodendrocyte precursors and maintain mature axoglial interactions in TH-deprived conditions.
Collapse
Affiliation(s)
- Min Joung Kim
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia
| |
Collapse
|
10
|
Vancamp P, Bourgeois NMA, Houbrechts AM, Darras VM. Knockdown of the thyroid hormone transporter MCT8 in chicken retinal precursor cells hampers early retinal development and results in a shift towards more UV/blue cones at the expense of green/red cones. Exp Eye Res 2018; 178:135-147. [PMID: 30273578 DOI: 10.1016/j.exer.2018.09.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 09/21/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022]
Abstract
Thyroid hormones (THs) play a crucial role in coordinating brain development in vertebrates. They fine-tune processes like cell proliferation, migration, and differentiation mainly by regulating the transcriptional activity of many essential genes. Regulators of TH availability thereby define the cellular concentration of the bioactive 3,5,3'-triiodothyronine, which binds to nuclear TH receptors. One important regulator, the monocarboxylate transporter 8 (MCT8), facilitates cellular TH uptake and is known to be necessary for correct brain development, but data on its potential role during retinal development is lacking. The retinal cyto-architecture has been conserved throughout vertebrate evolution, and we used the chicken embryo to study the need for MCT8 during retinal development. Its external development allows easy manipulation, and MCT8 is abundantly expressed in the retina from early stages onwards. We induced MCT8 knockdown by electroporating a pRFP-MCT8-RNAi vector into the retinal precursor cells (RPCs) at embryonic day 4 (E4), and studied the consequences for early (E6) and late (E18) retinal development. The empty pRFP-RNAi vector was used as a control. RPC proliferation was reduced at E6. This resulted in cellular hypoplasia and a thinner retina at E18 where mainly photoreceptors and horizontal cells were lost, the two predominant cell types that are born around the stage of electroporation. At E6, differentiation into retinal ganglion cells and amacrine cells was delayed. However, since the proportion of a given cell type within the transfected cell population at E18 was similar in knockdown and controls, the partial loss of some cell types was most-likely due to reduced RPC proliferation and not impaired cell differentiation. Photoreceptors displayed delayed migration at first, but had successfully reached the outer nuclear layer at E18. However, they increasingly differentiated into short wavelength-sensitive cones at the expense of medium/long wavelength-sensitive cones, while the proportion of rods was unaltered. Improperly formed sublaminae in the inner plexiform layer additionally suggested defects in synaptogenesis. Altogether, our data echoes effects of hypothyroidism and the loss of some other regulators of TH availability in the developing zebrafish and rodent retina. Therefore, the expression of MCT8 in RPCs is crucial for adequate TH uptake during cell type-specific events in retinal development.
Collapse
Affiliation(s)
- Pieter Vancamp
- KU Leuven, Laboratory of Comparative Endocrinology, Department of Biology, B-3000, Leuven, Belgium
| | - Nele M A Bourgeois
- KU Leuven, Laboratory of Comparative Endocrinology, Department of Biology, B-3000, Leuven, Belgium
| | - Anne M Houbrechts
- 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
|
11
|
Bloise FF, Oliveira TS, Cordeiro A, Ortiga-Carvalho TM. Thyroid Hormones Play Role in Sarcopenia and Myopathies. Front Physiol 2018; 9:560. [PMID: 29910736 PMCID: PMC5992417 DOI: 10.3389/fphys.2018.00560] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/30/2018] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle maintains posture and enables movement by converting chemical energy into mechanical energy, further contributing to basal energy metabolism. Thyroid hormones (thyroxine, or T4, and triiodothyronine, or T3) participate in contractile function, metabolic processes, myogenesis and regeneration of skeletal muscle. T3 classically modulates gene expression after binding to thyroid hormone nuclear receptors. Thyroid hormone effects depend on nuclear receptor occupancy, which is directly related to intracellular T3 levels. Sarcolemmal thyroid hormone levels are regulated by their transport across the plasma membrane by specific transporters, as well as by the action of deiodinases types 2 and 3, which can activate or inactivate T4 and T3. Thyroid hormone level oscillations have been associated with the worsening of many myopathies such as myasthenia gravis, Duchenne muscular dystrophy (DMD) and rhabdomyolysis. During aging skeletal muscle show a decrease in mass and quality, known as sarcopenia. There is increasing evidence that thyroid hormones could have a role in the sarcopenic process. Therefore, in this review, we aim to discuss the main effects of thyroid hormones in skeletal muscular aging processes and myopathy-related pathologies.
Collapse
Affiliation(s)
- Flavia F Bloise
- Laboratory of Translational Endocrinology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thamires S Oliveira
- Laboratory of Translational Endocrinology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aline Cordeiro
- Laboratory of Translational Endocrinology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tania M Ortiga-Carvalho
- Laboratory of Translational Endocrinology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Gouveia CHA, Miranda-Rodrigues M, Martins GM, Neofiti-Papi B. Thyroid Hormone and Skeletal Development. VITAMINS AND HORMONES 2018; 106:383-472. [PMID: 29407443 DOI: 10.1016/bs.vh.2017.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thyroid hormone (TH) is essential for skeletal development from the late fetal life to the onset of puberty. During this large window of actions, TH has key roles in endochondral and intramembranous ossifications and in the longitudinal bone growth. There is evidence that TH acts directly in skeletal cells but also indirectly, specially via the growth hormone/insulin-like growth factor-1 axis, to control the linear skeletal growth and maturation. The presence of receptors, plasma membrane transporters, and activating and inactivating enzymes of TH in skeletal cells suggests that direct actions of TH in these cells are crucial for skeletal development, which has been confirmed by several in vitro and in vivo studies, including mouse genetic studies, and clinical studies in patients with resistance to thyroid hormone due to dominant-negative mutations in TH receptors. This review examines progress made on understanding the mechanisms by which TH regulates the skeletal development.
Collapse
Affiliation(s)
- Cecilia H A Gouveia
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil.
| | | | - Gisele M Martins
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil; Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Bianca Neofiti-Papi
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
13
|
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
|
14
|
Hereditary spastic paraplegia: clinico-pathologic features and emerging molecular mechanisms. Acta Neuropathol 2013; 126:307-28. [PMID: 23897027 DOI: 10.1007/s00401-013-1115-8] [Citation(s) in RCA: 342] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 03/25/2013] [Accepted: 04/02/2013] [Indexed: 12/11/2022]
Abstract
Hereditary spastic paraplegia (HSP) is a syndrome designation describing inherited disorders in which lower extremity weakness and spasticity are the predominant symptoms. There are more than 50 genetic types of HSP. HSP affects individuals of diverse ethnic groups with prevalence estimates ranging from 1.2 to 9.6 per 100,000. Symptoms may begin at any age. Gait impairment that begins after childhood usually worsens very slowly over many years. Gait impairment that begins in infancy and early childhood may not worsen significantly. Postmortem studies consistently identify degeneration of corticospinal tract axons (maximal in the thoracic spinal cord) and degeneration of fasciculus gracilis fibers (maximal in the cervico-medullary region). HSP syndromes thus appear to involve motor-sensory axon degeneration affecting predominantly (but not exclusively) the distal ends of long central nervous system (CNS) axons. In general, proteins encoded by HSP genes have diverse functions including (1) axon transport (e.g. SPG30/KIF1A, SPG10/KIF5A and possibly SPG4/Spastin); (2) endoplasmic reticulum morphology (e.g. SPG3A/Atlastin, SPG4/Spastin, SPG12/reticulon 2, and SPG31/REEP1, all of which interact); (3) mitochondrial function (e.g. SPG13/chaperonin 60/heat-shock protein 60, SPG7/paraplegin; and mitochondrial ATP6); (4) myelin formation (e.g. SPG2/Proteolipid protein and SPG42/Connexin 47); (5) protein folding and ER-stress response (SPG6/NIPA1, SPG8/K1AA0196 (Strumpellin), SGP17/BSCL2 (Seipin), "mutilating sensory neuropathy with spastic paraplegia" owing to CcT5 mutation and presumably SPG18/ERLIN2); (6) corticospinal tract and other neurodevelopment (e.g. SPG1/L1 cell adhesion molecule and SPG22/thyroid transporter MCT8); (7) fatty acid and phospholipid metabolism (e.g. SPG28/DDHD1, SPG35/FA2H, SPG39/NTE, SPG54/DDHD2, and SPG56/CYP2U1); and (8) endosome membrane trafficking and vesicle formation (e.g. SPG47/AP4B1, SPG48/KIAA0415, SPG50/AP4M1, SPG51/AP4E, SPG52/AP4S1, and VSPG53/VPS37A). The availability of animal models (including bovine, murine, zebrafish, Drosophila, and C. elegans) for many types of HSP permits exploration of disease mechanisms and potential treatments. This review highlights emerging concepts of this large group of clinically similar disorders.
Collapse
|
15
|
Capri Y, Friesema EC, Kersseboom S, Touraine R, Monnier A, Eymard-Pierre E, Des Portes V, De Michele G, Brady AF, Boespflug-Tanguy O, Visser TJ, Vaurs-Barriere C. Relevance of Different Cellular Models in Determining the Effects of Mutations on SLC16A2/MCT8 Thyroid Hormone Transporter Function and Genotype-Phenotype Correlation. Hum Mutat 2013; 34:1018-25. [DOI: 10.1002/humu.22331] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/25/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Yline Capri
- INSERM; UMR 1103, CNRS 6293, GReD, Medical school; Clermont-Ferrand France
- APHP; Genetic Department; Robert Debré University Hospital; Paris France
- Université Paris Diderot; Sorbonne Paris Cité, Robert Debré University Hospital; Paris France
| | - Edith C.H. Friesema
- Department of Internal Medicine; Erasmus University Medical Center; Rotterdam The Netherlands
| | - Simone Kersseboom
- Department of Internal Medicine; Erasmus University Medical Center; Rotterdam The Netherlands
| | - Renaud Touraine
- Department of Clinical Chromosomal and Molecular Genetics; CHU St Etienne France
| | - Aurélie Monnier
- INSERM; UMR 1103, CNRS 6293, GReD, Medical school; Clermont-Ferrand France
- Medical Cytogenetic; Clermont-Ferrand University Hospital; Clermont-Ferrand France
| | - Eléonore Eymard-Pierre
- INSERM; UMR 1103, CNRS 6293, GReD, Medical school; Clermont-Ferrand France
- Medical Cytogenetic; Clermont-Ferrand University Hospital; Clermont-Ferrand France
| | - Vincent Des Portes
- Reference Center for Rare Intellectual Disabilities; Neuro-Paediatric Department, Debrousse Hospital; Lyon France
| | - Giusseppe De Michele
- Dipartimento di Scienze Neurologiche; Università di Napoli Federico II; Napoli Italy
| | - Angela F. Brady
- North West Thames Regional Genetics Service, Kennedy-Galton Centre; Northwick Park Hospital; Harrow United-Kingdom
| | - Odile Boespflug-Tanguy
- Université Paris Diderot; Sorbonne Paris Cité, Robert Debré University Hospital; Paris France
- APHP; Reference Center for Rare diseases “Leukodystrophies”, Pediatric Neurology and Metabolic Disorders Department, Robert Debré University Hospital; Paris France
- INSERM U676; Hôpital Robert Debré; Paris France
| | - Theo J. Visser
- Department of Internal Medicine; Erasmus University Medical Center; Rotterdam The Netherlands
| | - Catherine Vaurs-Barriere
- INSERM; UMR 1103, CNRS 6293, GReD, Medical school; Clermont-Ferrand France
- Auvergne University; Medical School; Clermont-Ferrand France
| |
Collapse
|
16
|
Dietrich JW, Landgrafe G, Fotiadou EH. TSH and Thyrotropic Agonists: Key Actors in Thyroid Homeostasis. J Thyroid Res 2012; 2012:351864. [PMID: 23365787 PMCID: PMC3544290 DOI: 10.1155/2012/351864] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 11/21/2012] [Indexed: 12/11/2022] Open
Abstract
This paper provides the reader with an overview of our current knowledge of hypothalamic-pituitary-thyroid feedback from a cybernetic standpoint. Over the past decades we have gained a plethora of information from biochemical, clinical, and epidemiological investigation, especially on the role of TSH and other thyrotropic agonists as critical components of this complex relationship. Integrating these data into a systems perspective delivers new insights into static and dynamic behaviour of thyroid homeostasis. Explicit usage of this information with mathematical methods promises to deliver a better understanding of thyrotropic feedback control and new options for personalised diagnosis of thyroid dysfunction and targeted therapy, also by permitting a new perspective on the conundrum of the TSH reference range.
Collapse
Affiliation(s)
- Johannes W. Dietrich
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
| | - Gabi Landgrafe
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
- Klinik für Allgemein- und Visceralchirurgie, Agaplesion Bethesda Krankenhaus Wuppertal gGmbH, Hainstraße 35, 42109 Wuppertal, NRW, Germany
| | - Elisavet H. Fotiadou
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
| |
Collapse
|
17
|
Zung A, Visser TJ, Uitterlinden AG, Rivadeneira F, Friesema ECH. A child with a deletion in the monocarboxylate transporter 8 gene: 7-year follow-up and effects of thyroid hormone treatment. Eur J Endocrinol 2011; 165:823-30. [PMID: 21896621 DOI: 10.1530/eje-11-0358] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The monocarboxylate transporter 8 (MCT8; SLC16A2) has a pivotal role in neuronal triiodothyronine (T(3)) uptake. Mutations of this transporter determine a distinct X-linked psychomotor retardation syndrome (Allan-Herndon-Dudley syndrome (AHDS)) that is attributed to disturbed thyroid hormone levels, especially elevated T(3) levels. We describe the genetic analysis of the MCT8 gene in a patient suspected for AHDS and the clinical and endocrine effects of L-thyroxine (LT(4)) or liothyronine (LT(3)) treatment intending to overcome the T(3) uptake resistance through alternative transporters. METHODS The six exons of the MCT8 gene were amplified individually by PCR. As multiple exons were missing, the length of the X-chromosomal deletion was determined by a dense SNP array, followed by PCR-based fine mapping to define the exact borders of the deleted segment. The clinical and endocrine data of the patient during 6.5 years of LT(4) treatment and two periods (3 months each) of low- and high-dose LT(3) were evaluated. RESULTS A partial deletion of the MCT8 gene (comprising five of six exons) was detected, confirming the suspected AHDS. MCT8 dysfunction was associated with partial resistance to T(3) at the hypothalamus and pituitary level, with normal responsiveness at the peripheral organs (liver and cardiovascular system). Thyroid hormone administration had no beneficial effect on the neurological status of the patient. CONCLUSION We identified a 70 kb deletion encompassing exons 2-6 of the MCT8 gene in our AHDS patient. Both LT(4) and LT(3) administration had no therapeutic effect. Alternatively, treatment of AHDS patients with thyroid hormone analogs should be considered.
Collapse
Affiliation(s)
- Amnon Zung
- Pediatric Endocrinology Unit, Kaplan Medical Center, Affiliated with the Hebrew University of Jerusalem, Rehovot 76100, Israel Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
18
|
Abstract
Thyroid hormones (TH) are essential for the development of the human brain, growth and cellular metabolism. Investigation of TH transporters became one of the emerging fields in thyroid research after the discovery of inactivating mutations in the Monocarboxylate transporter 8 (MCT8), which was found to be highly specific for TH transport. However, additional transmembrane transporters are also very important for TH uptake and efflux in different cell types. They transport TH as secondary substrates and include the aromatic amino acid transporting MCT10, the organic anion transporting polypeptides (e.g. OATP1C1, OATP1A2, OPTP1A4) and the large neutral amino acid transporters (LAT1 and LAT2). These TH transporters characteristically possess 12 transmembrane spanners but due to the strong differing sequences between the three transporter families we assume an identical conformation is not very likely. In contrast to the others, the LAT family members form a heterodimer with the escort protein 4F2hc/CD98. A comparison of sequence proportions, locations and types of functional sensitive features for TH transport discovered by mutations, revealed that transport sensitive charged residues occur as conserved amino acids only within each family of the transporter types but not in all putative TH transporters. Based on the lack of highly conserved sensitive charged residues throughout the three transporter families as a common counterpart for the amino acid moiety of the substrates, we conclude that the molecular transport mechanism is likely organized either a) by different molecular determinants in the divergent transporter types or b) the counterparts for the substrates` amino acid moiety at the transporter are not any charged side chains but other proton acceptors or donators. However, positions of transport sensitive residues coincide at transmembrane helix 8 in the TH transporter MCT8, OATP1C1 and another amino acid transporter, the L-cystine and L-glutamate exchanger xCT, which is highly homologous to LAT1 and LAT2. Here we review the data available and compare similarities and differences between these primary and secondary TH transporters regarding sequences, topology, potential structures, trafficking to the plasma membrane, molecular features and locations of transport sensitive functionalities. Thereby, we focus on TH transporters occurring in the blood-brain barrier.
Collapse
Affiliation(s)
- Anita Kinne
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Roessle-Str, 10, 13125 Berlin, Germany.
| | | | | |
Collapse
|
19
|
Menezes Filho HCD, Marui S, Manna TD, Brust ES, Radonsky V, Kuperman H, Dichtchekenian V, Setian N, Damiani D. Novel mutation in MCT8 gene in a Brazilian boy with thyroid hormone resistance and severe neurologic abnormalities. ACTA ACUST UNITED AC 2011; 55:60-6. [DOI: 10.1590/s0004-27302011000100008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 12/14/2010] [Indexed: 11/22/2022]
Abstract
MCT8 is a cellular transporter of thyroid hormones important in their action and metabolization. We report a male patient with the novel inactivating mutation 630insG in the coding region in exon 1 of MCT8. He was characterized clinically by severe neurologic impairment (initially with global hypotonia, later evolving with generalized hypertonia), normal growth during infancy, reduced weight gain, and absence of typical signs and symptoms of hypothyroidism, while the laboratory evaluation disclosed elevated T3, low total and free T4, and mildly elevated TSH serum levels. Treatment with levothyroxine improved thyroid hormone profile but was not able to alter the clinical picture of the patient. These data reinforce the concept that the role of MCT8 is tissue-dependent: while neurons are highly dependent on MCT8, bone tissue, adipose tissue, muscle, and liver are less dependent on MCT8 and, therefore, may suffer the consequences of the exposition to high serum T3 levels.
Collapse
|
20
|
Boccone L, Mariotti S, Dessì V, Pruna D, Meloni A, Loudianos G. Allan–Herndon–Dudley syndrome (AHDS) caused by a novel SLC16A2 gene mutation showing severe neurologic features and unexpectedly low TRH-stimulated serum TSH. Eur J Med Genet 2010; 53:392-5. [DOI: 10.1016/j.ejmg.2010.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 08/05/2010] [Indexed: 11/27/2022]
|
21
|
Sharlin DS, Gilbert ME, Taylor MA, Ferguson DC, Zoeller RT. The nature of the compensatory response to low thyroid hormone in the developing brain. J Neuroendocrinol 2010; 22:153-65. [PMID: 20041985 DOI: 10.1111/j.1365-2826.2009.01947.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thyroid hormone is essential for normal brain development, although the degree to which the developing brain is sensitive to small perturbations in serum thyroxin is not clear. An important concept related to this is that the developing brain possesses potent mechanisms to compensate for low serum thyroid hormone, and this concept is routinely employed in discussions concerning clinical treatments or public health. However, experimental studies have not directly tested whether (or the degree to which) putative compensatory mechanisms can ameliorate the consequences of small reductions in serum thyroxin (T(4)). To formally test this concept, we employed a model of graded T(4) reductions using doses of propylthiouracil (PTU) that were 200- to 67-fold lower than the dose traditionally used to produce hypothyroidism in rats. PTU produced a stepwise decrease in serum total T(4), and a stepwise increase in serum thyroid-stimulating hormone (TSH), in type 2 deiodinase mRNA expression and enzyme activity in the brain, and in the expression of the mRNA encoding the tri-iodothyronine (T(3)) transporter MCT8 in the postnatal day (P) 15 cortex. However, the mRNA encoding RC3/neurogranin, a direct target of T(3) action, exhibited a strong negative linear correlation with serum total T(4) despite these adaptive responses. In addition, single-cell analysis of RC3 mRNA levels in cortical neurones demonstrated that the co-expression of MCT8 did not alter the relationship between RC3 mRNA and serum T(4). These findings do not support the currently envisioned concept of the developing brain being capable of compensating for low T(4).
Collapse
Affiliation(s)
- D S Sharlin
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | | | | | | | | |
Collapse
|
22
|
James SR, Franklyn JA, Reaves BJ, Smith VE, Chan SY, Barrett TG, Kilby MD, McCabe CJ. Monocarboxylate transporter 8 in neuronal cell growth. Endocrinology 2009; 150:1961-9. [PMID: 19022891 DOI: 10.1210/en.2008-1031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Thyroid hormones are essential for the normal growth and development of the fetus, and even small alterations in maternal thyroid hormone status during early pregnancy may be associated with neurodevelopmental abnormalities in childhood. Mutations in the novel and specific thyroid hormone transporter monocarboxylate transporter 8 (MCT8) have been associated with severe neurodevelopmental impairment. However, the mechanism by which MCT8 influences neural development remains poorly defined. We have therefore investigated the effect of wild-type (WT) MCT8, and the previously reported L471P mutant, on the growth and function of human neuronal precursor NT2 cells as well as MCT8-null JEG-3 cells. HA-tagged WT MCT8 correctly localized to the plasma membrane in NT2 cells and increased T(3) uptake in both cell types. In contrast, L471P MCT8 was largely retained in the endoplasmic reticulum and displayed no T(3) transport activity. Transient overexpression of WT and mutant MCT8 proteins failed to induce endoplasmic reticular stress or apoptosis. However, MCT8 overexpression significantly repressed cell proliferation in each cell type in both the presence and absence of the active thyroid hormone T(3) and in a dose-dependent manner. In contrast, L471P MCT8 showed no such influence. Finally, small interfering RNA depletion of endogenous MCT8 resulted in increased cell survival and decreased T(3) uptake. Given that T(3) stimulated proliferation in embryonic neuronal NT2 cells, whereas MCT8 repressed cell growth, these data suggest an entirely novel role for MCT8 in addition to T(3) transport, mediated through the modulation of cell proliferation in the developing brain.
Collapse
Affiliation(s)
- S R James
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Thyroid hormone metabolism and action are largely intracellular events that require transport of iodothyronines across the plasma membrane. It has been assumed for a long time that this occurs by passive diffusion, but it has become increasingly clear that cellular uptake and efflux of thyroid hormone is mediated by transporter proteins. Recently, several active and specific thyroid hormone transporters have been identified, including monocarboxylate transporter 8 (MCT8), MCT10, and organic anion transporting polypeptide 1C1 (OATP1C1). The latter is expressed predominantly in brain capillaries and transports preferentially T(4), whereas MCT8 and MCT10 are expressed in multiple tissues and are capable of transporting different iodothyronines. The pathophysiological importance of thyroid hormone transporters has been established by the demonstration of MCT8 mutations in patients with severe psychomotor retardation and elevated serum T(3) levels. MCT8 appears to play an important role in the transport of thyroid hormone in the brain, which is essential for the crucial action of the hormone during brain development. It is expected that more specific thyroid hormone transporters will be discovered in the near future, which will lead to a better understanding of the tissue-specific regulation of thyroid hormone bioavailability.
Collapse
Affiliation(s)
- Heike Heuer
- Department of Internal Medicine, Section of Endocrinology, Erasmus Medical College, Rotterdam, The Netherlands
| | | |
Collapse
|
24
|
Fuchs O, Pfarr N, Pohlenz J, Schmidt H. Elevated serum triiodothyronine and intellectual and motor disability with paroxysmal dyskinesia caused by a monocarboxylate transporter 8 gene mutation. Dev Med Child Neurol 2009; 51:240-4. [PMID: 19018842 DOI: 10.1111/j.1469-8749.2008.03125.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Monocarboxylate transporter 8 (MCT8 or SLC16A2) is important for the neuronal uptake of triiodothyronine (T3) in its function as a specific and active transporter of thyroid hormones across the cell membrane, thus being essential for human brain development. We report on a German male with Allan-Herndon-Dudley syndrome presenting with severe intellectual and motor disability, paroxysmal dyskinesia combined with truncal muscular hypotonia, and peripheral muscular hypertonia at his current age of 9 years. Additionally, the patient has a lesion in the left putamen region revealed by magnetic resonance imaging and elevated serum T3 levels. The male appeared to have a hemizygous mutation (R271H) in the MCT8 gene that was sequenced directly from genomic DNA and occurred de novo in the maternal germline, as both his mother and his sister were not carriers of the mutation. Ruling out a common polymorphism, 50 normal individuals of the same ethnic background did not harbour the mutation. The identified MCT8 gene mutation (R271H) is very likely to be the genetic cause for neuronal hypothyroidism despite elevated serum T3 levels.
Collapse
Affiliation(s)
- Oliver Fuchs
- Dr von Hauner University Children's Hospital, Pediatric Endocrinology, Munich, Germany
| | | | | | | |
Collapse
|
25
|
Capelo LP, Beber EH, Fonseca TL, Gouveia CHA. The monocarboxylate transporter 8 and L-type amino acid transporters 1 and 2 are expressed in mouse skeletons and in osteoblastic MC3T3-E1 cells. Thyroid 2009; 19:171-80. [PMID: 19133747 DOI: 10.1089/thy.2008.0120] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Several plasma membrane transporters have been shown to mediate the cellular influx and/or efflux of iodothyronines, including the sodium-independent organic anion co-transporting polypeptide 1 (OATP1), the sodium taurocholate co-transporting polypeptide (NTCP), the L-type amino acid transporter 1 (LAT1) and 2 (LAT2), and the monocarboxylate transporter 8 (MCT8). The aim of this study was to investigate if the mRNAs of these transporters were expressed and regulated by thyroid hormone (TH) in mouse calvaria-derived osteoblastic MC3T3-E1 cells and in the fetal and postnatal bones of mice. METHODS The mRNA expression of the iodothyronine transporters was investigated with real-time polymerase chain reaction analysis in euthyroid and hypothyroid fetuses and litters of mice and in MC3T3-E1 cells treated with increasing doses of triiodothyronine (T(3); 10(-10) to 10(-6) M) or with 10(-8) M T(3) for 1-9 days. RESULTS MCT8, LAT1, and LAT2 mRNAs were detected in fetal and postnatal femurs and in MC3T3-E1 cells, while OATP1 and NTCP mRNAs were not. LAT1 and LAT2 mRNAs were not affected by TH status in vivo or in vitro or by the stage of bone development or osteoblast maturation (analyzed by the expression of osteocalcin and alkaline phosphatase, which are key markers of osteoblastic differentiation). In contrast, the femoral mRNA expression of MCT8 decreased significantly during post-natal development, whereas MCT8 mRNA expression increased as MC3T3-E1 cells differentiated. We also showed that MCT8 mRNA was up-regulated in the femur of hypothyroid animals, and that it was down-regulated by treatment with T(3) in MC3T3-E1 cells. CONCLUSIONS This is the first study to demonstrate the mRNA expression of LAT1, LAT2, and MCT8 in the bone tissue of mice and in osteoblast-like cells. In addition, the pattern of MCT8 expression observed in vivo and in vitro suggests that MCT8 may be important to modulate TH effects on osteoblast differentiation and on bone development and metabolism.
Collapse
Affiliation(s)
- Luciane P Capelo
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | | |
Collapse
|
26
|
Nunez J, Celi FS, Ng L, Forrest D. Multigenic control of thyroid hormone functions in the nervous system. Mol Cell Endocrinol 2008; 287:1-12. [PMID: 18448240 PMCID: PMC2486256 DOI: 10.1016/j.mce.2008.03.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 03/07/2008] [Accepted: 03/07/2008] [Indexed: 12/16/2022]
Abstract
Thyroid hormone (TH) has a remarkable range of actions in the development and function of the nervous system. A multigenic picture is emerging of the mechanisms that specify these diverse functions in target tissues. Distinct responses are mediated by alpha and beta isoforms of TH receptor which act as ligand-regulated transcription factors. Receptor activity can be regulated at several levels including that of uptake of TH ligand and the activation or inactivation of ligand by deiodinase enzymes in target tissues. Processes under the control of TH range from learning and anxiety-like behaviour to sensory function. At the cellular level, TH controls events as diverse as axonal outgrowth, hippocampal synaptic activity and the patterning of opsin photopigments necessary for colour vision. Overall, TH coordinates this variety of events in both central and sensory systems to promote the function of the nervous system as a complete entity.
Collapse
|
27
|
Sijens PE, Rödiger LA, Meiners LC, Lunsing RJ. 1H magnetic resonance spectroscopy in monocarboxylate transporter 8 gene deficiency. J Clin Endocrinol Metab 2008; 93:1854-9. [PMID: 18319316 DOI: 10.1210/jc.2007-2441] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT In monocarboxylate transporter 8 (MCT8) gene deficiency, a syndrome combining thyroid and neurological abnormalities, the central nervous system has not yet been characterized by magnetic resonance (MR) spectroscopy. OBJECTIVE We studied whether the degree of dysmyelinization in MCT8 gene deficiency according to MR imaging (MRI) is coupled with abnormalities in brain metabolism. DESIGN MRI and MR spectroscopy of the brain were performed twice in two MCT8 gene deficiency patients, for the first time at age 8-10 months and for the second time at age 17-28 months. The results were compared with those obtained in controls of a similar age. RESULTS Compared with controls, young children with MCT8 show choline and myoinositol level increases and N-acetyl aspartate decreases in supraventricular gray and white matter, phenomena associated with the degree of dysmyelinization according to MRI. CONCLUSION MCT8 gene deficiency results in deviant myelinization and general atrophy, which is substantiated by the MR spectroscopy findings of increased choline and myoinositol levels and decreased N-acetyl aspartate. The observations suggest that different mutations in the MCT8 gene lead to differences in the severity of the clinical spectrum, dysmyelinization, and MR spectroscopy-detectable changes in brain metabolism.
Collapse
Affiliation(s)
- Paul E Sijens
- Department of Radiology, University Medical Center Groningen and University of Groningen, Hanzeplein 1, Groningen, The Netherlands.
| | | | | | | |
Collapse
|
28
|
Jansen J, Friesema ECH, Kester MHA, Schwartz CE, Visser TJ. Genotype-phenotype relationship in patients with mutations in thyroid hormone transporter MCT8. Endocrinology 2008; 149:2184-90. [PMID: 18187543 PMCID: PMC2734492 DOI: 10.1210/en.2007-1475] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Loss-of-function mutations in thyroid hormone transporter monocarboxylate transporter 8 (MCT8) lead to severe X-linked psychomotor retardation and elevated serum T(3) levels. Most patients, for example those with mutations V235M, S448X, insI189, or delF230, cannot stand, walk, or speak. Patients with mutations L434W, L568P, and S194F, however, walk independently and/or develop some dysarthric speech. To study the relationship between mutation and phenotype, we transfected JEG3 and COS1 cells with wild-type or mutant MCT8. Expression and function of the transporter were studied by analyzing T(3) and T(4) uptake, T(3) metabolism (by cotransfected type 3 deiodinase), Western blotting, affinity labeling with N-bromoacetyl-T(3), immunocytochemistry, and quantitative RT-PCR. Wild-type MCT8 increased T(3) uptake and metabolism about 5-fold compared with empty vector controls. Mutants V235M, S448X, insI189, and delF230 did not significantly increase transport. However, S194F, L568P, and L434W showed about 20, 23, and 37% of wild-type activity. RT-PCR did not show significant differences in mRNA expression between wild-type and mutant MCT8. Immunocytochemistry detected the nonfunctional mutants V235M, insI189, and delF230 mostly in the cytoplasm, whereas mutants with residual function were expressed at the plasma membrane. Mutants S194F and L434W showed high protein expression but low affinity for N-bromoacetyl-T(3); L568P was detected in low amounts but showed relatively high affinity. Mutations in MCT8 cause loss of function through reduced protein expression, impaired trafficking to the plasma membrane, or reduced substrate affinity. Mutants L434W, L568P, and S194F showed significant residual transport capacity, which may underlie the more advanced psychomotor development observed in patients with these mutations.
Collapse
Affiliation(s)
- Jurgen Jansen
- Department of Internal Medicine, Erasmus Medical Center, Room Ee502, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
29
|
Frints SGM, Lenzner S, Bauters M, Jensen LR, Van Esch H, des Portes V, Moog U, Macville MVE, van Roozendaal K, Schrander-Stumpel CTRM, Tzschach A, Marynen P, Fryns JP, Hamel B, van Bokhoven H, Chelly J, Beldjord C, Turner G, Gecz J, Moraine C, Raynaud M, Ropers HH, Froyen G, Kuss AW. MCT8 mutation analysis and identification of the first female with Allan–Herndon–Dudley syndrome due to loss of MCT8 expression. Eur J Hum Genet 2008; 16:1029-37. [DOI: 10.1038/ejhg.2008.66] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
30
|
Friesema ECH, Jansen J, Jachtenberg JW, Visser WE, Kester MHA, Visser TJ. Effective cellular uptake and efflux of thyroid hormone by human monocarboxylate transporter 10. Mol Endocrinol 2008; 22:1357-69. [PMID: 18337592 DOI: 10.1210/me.2007-0112] [Citation(s) in RCA: 200] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cellular entry of thyroid hormone is mediated by plasma membrane transporters, among others a T-type (aromatic) amino acid transporter. Monocarboxylate transporter 10 (MCT10) has been reported to transport aromatic amino acids but not iodothyronines. Within the MCT family, MCT10 is most homologous to MCT8, which is a very important iodothyronine transporter but does not transport amino acids. In view of this paradox, we decided to reinvestigate the possible transport of thyroid hormone by human (h) MCT10 in comparison with hMCT8. Transfection of COS1 cells with hMCT10 cDNA resulted in 1) the production of an approximately 55 kDa protein located to the plasma membrane as shown by immunoblotting and confocal microscopy, 2) a strong increase in the affinity labeling of intracellular type I deiodinase by N-bromoacetyl-[(125)I]T(3), 3) a marked stimulation of cellular T(4) and, particularly, T(3) uptake, 4) a significant inhibition of T(3) uptake by phenylalanine, tyrosine, and tryptophan of 12.5%, 22.2%, and 51.4%, respectively, and 5) a marked increase in the intracellular deiodination of T(4) and T(3) by different deiodinases. Cotransfection studies using the cytosolic thyroid hormone-binding protein micro-crystallin (CRYM) indicated that hMCT10 facilitates both cellular uptake and efflux of T(4) and T(3). In the absence of CRYM, hMCT10 and hMCT8 increased T(3) uptake after 5 min incubation up to 4.0- and 1.9-fold, and in the presence of CRYM up to 6.9- and 5.8-fold, respectively. hMCT10 was less active toward T(4) than hMCT8. These findings establish that hMCT10 is at least as active a thyroid hormone transporter as hMCT8, and that both transporters facilitate iodothyronine uptake as well as efflux.
Collapse
Affiliation(s)
- Edith C H Friesema
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
31
|
Visser WE, Friesema ECH, Jansen J, Visser TJ. Thyroid hormone transport in and out of cells. Trends Endocrinol Metab 2008; 19:50-6. [PMID: 18291666 DOI: 10.1016/j.tem.2007.11.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Revised: 11/01/2007] [Accepted: 11/07/2007] [Indexed: 12/13/2022]
Abstract
Thyroid hormone (TH) is essential for the proper development of numerous tissues, notably the brain. TH acts mostly intracellularly, which requires transport by TH transporters across the plasma membrane. Although several transporter families have been identified, only monocarboxylate transporter (MCT)8, MCT10 and organic anion-transporting polypeptide (OATP)1C1 demonstrate a high degree of specificity towards TH. Recently, the biological importance of MCT8 has been elucidated. Mutations in MCT8 are associated with elevated serum T(3) levels and severe psychomotor retardation, indicating a pivotal role for MCT8 in brain development. MCT8 knockout mice lack neurological damage, but mimic TH abnormalities of MCT8 patients. The exact pathophysiological mechanisms in MCT8 patients remain to be elucidated fully. Future research will probably identify novel TH transporters and disorders based on TH transporter defects.
Collapse
Affiliation(s)
- W Edward Visser
- Department of Internal Medicine, Erasmus University Medical Centre, Dr Molewaterplein 50, Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
32
|
Papadimitriou A, Dumitrescu AM, Papavasiliou A, Fretzayas A, Nicolaidou P, Refetoff S. A novel monocarboxylate transporter 8 gene mutation as a cause of severe neonatal hypotonia and developmental delay. Pediatrics 2008; 121:e199-202. [PMID: 18166539 DOI: 10.1542/peds.2007-1247] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Monocarboxylate transporter 8 acts as a specific cell membrane transporter for thyroxine and especially triiodothyronine into target cells. It is expressed in brain neurons and in many other tissues. The monocarboxylate transporter 8 gene resides on chromosome Xq13.2. An 11-month-old male infant was referred because of severe hypotonia from early life and global developmental delay. Thyroid-function tests showed normal thyrotropin levels and the characteristic for the disorder, including high serum triiodothyronine and low thyroxine concentrations. Molecular analysis of the monocarboxylate transporter 8 gene showed that the patient was hemizygous for a novel missense mutation P537L. This case highlights the importance of determining thyroid hormone levels, especially triiodothyronine, in infants with severe neonatal hypotonia.
Collapse
Affiliation(s)
- Anastasios Papadimitriou
- Pediatric Endocrinology Unit, Third Department of Pediatrics, University of Athens School of Medicine, Attikon University Hospital, Rimini 1, Haidari, Athens 124 62, Greece.
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Thyroid hormones are vital for fetal development and can act directly on placental tissues to modify their metabolism, differentiation and development. There is evidence that maternal thyroid hormones can cross the human placenta and act to modulate fetal development before the onset of the fetus's own thyroid hormone production. Plasma membrane transport of thyroid hormones has now been shown to require specific transporter proteins. Several proteins have recently been identified as specific thyroid hormone transporters. However, as yet few data are available to define the functionally important transporter proteins in the human placenta. To date, members of the organic anion-transporting polypeptide, L-type amino acid, and of the monocarboxylate transporter families, have been identified as thyroid hormone transporters that are active in a variety of placental cell types. However, further research is necessary to determine the role of these and other proteins in placental transport of thyroid hormone, and to investigate how modulations of their function could affect fetal pathologies such as intrauterine growth restriction.
Collapse
Affiliation(s)
- Sally R James
- Division of Reproductive and Child Health, University of Birmingham, Birmingham, UK
| | | | | |
Collapse
|
34
|
Abstract
Thyroid hormone is essential for the proper development and function of the brain. The active form of thyroid hormone is T(3), which binds to nuclear receptors. Recently, a transporter specific for T(3), MCT8 (monocarboxylate transporter 8) was identified. MCT8 is highly expressed in liver and brain. The gene is located in Xq13 and mutations in MCT8 are responsible for an X-linked condition, Allan-Herndon-Dudley syndrome (AHDS). This syndrome is characterized by congenital hypotonia that progresses to spasticity with severe psychomotor delays. Affected males also present with muscle hypoplasia, generalized muscle weakness, and limited speech. Importantly, these patients have elevated serum levels of free T(3), low to below normal serum levels of free T(4), and levels of thyroid stimulating hormone that are within the normal range. This constellation of measurements of thyroid function enables quick screening for AHDS in males presenting with cognitive impairment, congenital hypotonia, and generalized muscle weakness.
Collapse
Affiliation(s)
- Charles E Schwartz
- JC Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, SC, USA.
| | | |
Collapse
|
35
|
Refetoff S, Dumitrescu AM. Syndromes of reduced sensitivity to thyroid hormone: genetic defects in hormone receptors, cell transporters and deiodination. Best Pract Res Clin Endocrinol Metab 2007; 21:277-305. [PMID: 17574009 DOI: 10.1016/j.beem.2007.03.005] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
At least six major steps are required for secreted thyroid hormone (TH) to exert its action on target tissues. Mutations interfering with three of these steps have been so far identified. The first recognized defect, which causes resistance to TH, involves the TH receptor beta gene and has been given the acronym RTH. Occurring in approximately 1 per 40,000 newborns, more than 1000 affected subjects, from 339 families, have been identified. The gene defect remains unknown in 15% of subjects with RTH. Two novel syndromes causing reduced sensitivity to TH were recently identified. One, producing severe psychomotor defects in > 100 males from 26 families, is caused by mutations in the cell-membrane transporter of TH, MCT8; the second, affecting the intracellular metabolism of TH in four individuals from two families, is caused by mutations in the SECISBP2 gene, which is required for the synthesis of selenoproteins, including TH deiodinases.
Collapse
Affiliation(s)
- Samuel Refetoff
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | | |
Collapse
|
36
|
Jansen J, Friesema ECH, Kester MHA, Milici C, Reeser M, Grüters A, Barrett TG, Mancilla EE, Svensson J, Wemeau JL, Busi da Silva Canalli MH, Lundgren J, McEntagart ME, Hopper N, Arts WF, Visser TJ. Functional analysis of monocarboxylate transporter 8 mutations identified in patients with X-linked psychomotor retardation and elevated serum triiodothyronine. J Clin Endocrinol Metab 2007; 92:2378-81. [PMID: 17356046 DOI: 10.1210/jc.2006-2570] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
CONTEXT T(3) action in neurons is essential for brain development. Recent evidence indicates that monocarboxylate transporter 8 (MCT8) is important for neuronal T(3) uptake. Hemizygous mutations have been identified in the X-linked MCT8 gene in boys with severe psychomotor retardation and elevated serum T(3) levels. OBJECTIVE The objective of this study was to determine the functional consequences of MCT8 mutations regarding transport of T(3). DESIGN MCT8 function was studied in wild-type or mutant MCT8-transfected JEG3 cells by analyzing: 1) T(3) uptake, 2) T(3) metabolism in cells cotransfected with human type 3 deiodinase, 3) immunoblotting, and 4) immunocytochemistry. RESULTS The mutations identified in MCT8 comprise four deletions (24.5 kb, 2.4 kb, 14 bp, and 3 bp), three missense mutations (Ala224Val, Arg271His, and Leu471Pro), a nonsense mutation (Arg245stop), and a splice site mutation (94 amino acid deletion). All tested mutants were inactive in uptake and metabolism assays, except MCT8 Arg271His, which showed approximately 20% activity vs. wild-type MCT8. CONCLUSION These findings support the hypothesis that the severe psychomotor retardation and elevated serum T(3) levels in these patients are caused by inactivation of the MCT8 transporter, preventing action and metabolism of T(3) in central neurons.
Collapse
Affiliation(s)
- Jurgen Jansen
- Department of Internal Medicine, Erasmus Medical Center, Room Ee502, Dr Molewaterplein 50, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Heuer H. The importance of thyroid hormone transporters for brain development and function. Best Pract Res Clin Endocrinol Metab 2007; 21:265-76. [PMID: 17574008 DOI: 10.1016/j.beem.2007.03.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Thyroid hormone is essential for proper brain development and function. As a prerequisite for its action, transporters must exist to mediate its cellular entry. As impaired uptake of thyroid hormone into the CNS causes severe neurological symptoms, it is of utmost importance to identify these carriers. The monocarboxylate transporter 8 (MCT8) was recently characterized as a very specific thyroid hormone transporter. Inactivating mutations in the MCT8 gene are associated with a severe syndrome of psychomotor retardation and abnormal thyroid hormone parameters. To elucidate the underlying pathogenic mechanisms, MCT8-deficient mice that replicate the human thyroid phenotype, despite the absence of overt neurological symptoms, have been generated. Here, we summarize recent findings obtained by analyzing these animals and discuss their potential impact for the treatment of affected patients.
Collapse
Affiliation(s)
- Heike Heuer
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Beutenbergstrasse 11, Jena, Germany.
| |
Collapse
|
38
|
Tjørve E, Tjørve KMC, Olsen JO, Senum R, Oftebro H. On commonness and rarity of thyroid hormone resistance: a discussion based on mechanisms of reduced sensitivity in peripheral tissues. Med Hypotheses 2007; 69:913-21. [PMID: 17383828 DOI: 10.1016/j.mehy.2006.12.056] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2006] [Accepted: 01/10/2007] [Indexed: 10/23/2022]
Abstract
Reduced sensitivity to thyroid hormone (TH) in peripheral tissues can occur as defects in TH transport into the cell, intracellular TH metabolism, cytosolic mechanisms, TH entry into the nucleus, thyroxin receptors (TRs) and receptor binding, transcription and post-transcriptional mechanisms. Current literature reveals an extensive list of mutations, drugs, toxins, metabolites and autoimmune antibodies that may impair TH action in the cell, but such impairment may not be picked up by assays of TH and TSH in blood plasma. Substances may induce tissue specific resistance to thyroid hormone (RTH), e.g. by affecting numbers of different TR isoforms. Recent literature also indicates mechanisms by which different conditions, for example, chronic fatigue syndrome (CFS), chronic renal failure (CRF) and nonthyroidal illness, can be accompanied by acquired RTH caused by inhibition of TH metabolism, cell uptake, TR binding and transcription. This prompts us to reassess commonness and rarity of congenital vs. acquired RTH. We hypothesise that observed clinical symptoms of hypothyroidism in chemically euthyroid patients are typically caused by changes in hormonal systems, autoimmune antibodies, metabolites or other substances in the body, leading to reduced sensitivity to TH in peripheral tissues. These changes may be a by-product of other processes and a reversible biological response in the body, and may also result in chronic acquired RTH. Antibodies may prove to be the most common cause of chronic reduction in TH sensitivity. It is argued that the acquired form of RTH, caused by endogenous and exogenous sources, may indeed be more common than the congenital, as in insulin resistance. If acquired RTH exists, then it may not be picked up by blood assays of TH and TSH. An appropriate test to assess TH action in peripheral tissues is therefore greatly desired.
Collapse
Affiliation(s)
- E Tjørve
- Lillehammer University College, 2626 Lillehammer, Norway.
| | | | | | | | | |
Collapse
|
39
|
Cheng SY. Thyroid hormone receptor mutations and disease: insights from knock-in mouse models. Expert Rev Endocrinol Metab 2007; 2:47-57. [PMID: 30743748 DOI: 10.1586/17446651.2.1.47] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Thyroid hormone nuclear receptors (TRs) mediate thyroid hormone's activities in growth, differentiation, and development. Two TR genes (α and β ) encode four thyroid hormone-binding receptors that regulate target gene expression. Mutations of the TRβ gene cause the genetic syndrome of resistance to thyroid hormone. Studies indicate a close association between TRβ mutations and several human cancers, suggesting their oncogenic role. A TRβ gene knock-in mutant mouse (TRβPV/PV mouse) that spontaneously develops thyroid cancer allows elucidation of the oncogenic functions in vivo. TRβPV is a potent dominant negative mutant identified in a resistance to thyroid hormone patient. Molecular studies indicate that the PV mutant mediates its oncogenic activities via nucleus-initiated transcription and novel extranuclear actions. Thus, the deleterious effects of the gene mutations go beyond resistance to thyroid hormone and are more severe and extensive than previously envisioned. This newly identified oncogene exerts its tumorigenic effects via multiple signaling mechanisms.
Collapse
Affiliation(s)
- Sheue-Yann Cheng
- a National Cancer Institute, Laboratory of Molecular Biology, 37 Convent Dr., Room 5128, Bethesda, MD 20892-4264, USA.
| |
Collapse
|
40
|
Beck-Peccoz P, Persani L, Calebiro D, Bonomi M, Mannavola D, Campi I. Syndromes of hormone resistance in the hypothalamic-pituitary-thyroid axis. Best Pract Res Clin Endocrinol Metab 2006; 20:529-46. [PMID: 17161330 DOI: 10.1016/j.beem.2006.11.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Forty years have elapsed since the first description of a syndrome of resistance in the hypothalamic-pituitary-thyroid axis, i.e., resistance to thyroid hormone action. In the last two decades many other types of resistance have been discovered, including resistance to the action of thyrotropin-releasing hormone (TRH), of thyroid-stimulating hormone (TSH), and of thyroid hormones (THs); the latter can be due not only to thyroid hormone receptor defects but also to alteration in genes encoding TH-specific transporters or components involved in metabolic pathways of THs. Moreover, alteration in genes encoding for second messengers may cause forms of resistance other than those due to receptor mutations, the most important one being that of an inactivating mutation in the G-protein alpha-subunit leading to TSH resistance in the setting of pseudohypoparathyroidism type 1a. Recognition of these rare thyroid disorders is of great importance not only for informed genetic counselling but also for avoiding diagnostic mistakes that may lead to incorrect and potentially dangerous treatments.
Collapse
Affiliation(s)
- Paolo Beck-Peccoz
- Department of Medical Sciences, Endocrine and Metabolic Unit, Fondazione Ospedale Maggiore IRCCS, Padiglione Granelli, Via Francesco Sforza 35, 20122-Milano, Italy.
| | | | | | | | | | | |
Collapse
|
41
|
Friesema ECH, Jansen J, Heuer H, Trajkovic M, Bauer K, Visser TJ. Mechanisms of disease: psychomotor retardation and high T3 levels caused by mutations in monocarboxylate transporter 8. ACTA ACUST UNITED AC 2006; 2:512-23. [PMID: 16957765 DOI: 10.1038/ncpendmet0262] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2005] [Accepted: 05/23/2006] [Indexed: 11/09/2022]
Abstract
The actions and the metabolism of thyroid hormone are intracellular events that require the transport of iodothyronines across the plasma membrane. It is increasingly clear that this process does not occur by simple diffusion, but is facilitated by transport proteins. Only recently have iodothyronine transporters been identified at the molecular level, of which organic anion transporting polypeptide 1C1 and monocarboxylate transporter 8 (MCT8) deserve special mention, because of their high activity and specificity for iodothyronines. Organic anion transporting polypeptide 1C1 is almost exclusively expressed in brain capillaries, and may be crucial for the transport of the prohormone T4 across the blood-brain barrier. MCT8 is also expressed in the brain--in particular, in neurons--but also in other tissues. MCT8 seems to be especially important for the uptake of active hormone T3 into neurons, which is essential for optimal brain development. T3 is produced from T4 by type 2 deiodinase in neighboring astrocytes. Neurons express type 3 deiodinase, the enzyme that terminates T3 activity. The SLC16A2 (formerly MCT8) gene is located on chromosome Xq13.2 and has recently been associated with a syndrome combining severe, X-linked, psychomotor retardation and high serum T3 levels. In over 20 families, where affected males have developed this syndrome, several mutations in MCT8 have been identified. The disease mechanism is thought to involve a defect in the neuronal entry of T3 and, therefore, in the action and metabolism of T3 in these cells. This defect results in impaired neurological development and a decrease in T3 clearance.
Collapse
Affiliation(s)
- Edith C H Friesema
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
42
|
Herzovich V, Vaiani E, Marino R, Dratler G, Lazzati JM, Tilitzky S, Ramirez P, Iorcansky S, Rivarola MA, Belgorosky A. Unexpected Peripheral Markers of Thyroid Function in a Patient with a Novel Mutation of the MCT8 Thyroid Hormone Transporter Gene. Horm Res Paediatr 2006; 67:1-6. [PMID: 16974106 DOI: 10.1159/000095805] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Accepted: 05/15/2006] [Indexed: 11/19/2022] Open
Abstract
The specific thyroid hormone transporter, MCT8, located on the X chromosome, has led to the identification a novel syndrome. The objective is to relate phenotype with several tissue-specific thyroid functions. A 1-year-old boy, who had severe psychological damage and low serum T4, had received l-T4 for 3 months. At admission, body length was normal but weight was low. Off therapy, serum TSH was mildly elevated, serum T4 and free T4 were low, and serum T3 and free T3 were high. Direct sequencing of the MCT8 gene revealed a single nucleotide change that resulted in a novel nonsense mutation at codon 261 (Q261X) in exon 3. Since serum T3 was high, peripheral markers of hyperthyroidism were looked for. Bone age was advanced, despite the presence of malnutrition and low T4. Serum SHBG, a marker of thyroid hormone action in liver, was markedly elevated. Markers of skeletal muscle catabolism, ammonemia and lactic acid, were found to be elevated. The phenotype of MCT 8 mutation might be explained by differences in the entry of thyroid hormones into different cells. In the presence of an inactive MCT8 transporter, the high blood T3 levels might not be enough to prevent brain damage early in life, while they seem to be able to induce a postnatal state of peripheral hyperthyroidism in other tissues, such as liver, bone and skeletal muscle.
Collapse
Affiliation(s)
- V Herzovich
- Endocrinology Service, Hospital de Pediatria Garrahan, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Dumitrescu AM, Liao XH, Weiss RE, Millen K, Refetoff S. Tissue-specific thyroid hormone deprivation and excess in monocarboxylate transporter (mct) 8-deficient mice. Endocrinology 2006; 147:4036-43. [PMID: 16709608 DOI: 10.1210/en.2006-0390] [Citation(s) in RCA: 258] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mutations of the X-linked thyroid hormone (TH) transporter (monocarboxylate transporter, MCT8) produce in humans unusual abnormalities of thyroid function characterized by high serum T3 and low T4 and rT3. The mechanism of these changes remains obscure and raises questions regarding the regulation of intracellular availability and metabolism of TH. To study the pathophysiology of MCT8 deficiency, we generated Mct8 knockout mice. Male mice deficient in Mct8 (Mct8(-/y)) replicate the thyroid abnormalities observed in affected men. TH deprivation and replacement with L-T3 showed that suppression of TSH required higher serum levels T3 in Mct8(-/y) than wild-type (WT) littermates, indicating hypothalamus and/or thyrotroph resistance to T3. Furthermore, T4 is required to maintain the high serum T3 level because the latter was not different between the two genotypes during administration of T3. Mct8(-/y) mice have 2.3-fold higher T3 content in liver associated with 6.1- and 3.1-fold increase in deiodinase 1 mRNA and enzymatic activity, respectively. The relative T3 excess in liver of Mct8(-/y) mice produced a decrease in serum cholesterol (79 +/- 18 vs. 137 +/- 38 mg/dl in WT) and an increase in alkaline phosphatase (107 +/- 23 vs. 58 +/- 3 U/liter in WT) levels. In contrast, T3 content in cerebrum was 1.8-fold lower in Mct8(-/y) mice, associated with a 1.6- and 10.6-fold increase in D2 mRNA and enzymatic activity, respectively, as previously observed in TH-deprived WT mice. We conclude that cell-specific differences in intracellular TH content due to differences in contribution of the various TH transporters are responsible for the unusual clinical presentation of this defect, in contrast to TH deficiency.
Collapse
Affiliation(s)
- Alexandra M Dumitrescu
- Department of Medicine, University of Chicago, MC 3090, 5841 South Maryland Avenue, Chicago, Illinois 60637, USA
| | | | | | | | | |
Collapse
|