1
|
Lin P, Liu H, Lou J, Lyu G, Li Y, He P, Fu Y, Zhang R, Zhang Y, Yan T. Novel SLC16A2 Frameshift Mutation as a Cause of Allan-Herndon-Dudley Syndrome and its Implications for Carrier Screening. Pharmgenomics Pers Med 2025; 18:85-94. [PMID: 40291819 PMCID: PMC12034286 DOI: 10.2147/pgpm.s492647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Background Allan-Herndon-Dudley syndrome (AHDS) is a rare X-linked neurodevelopmental disorder caused by mutations in the solute carrier family 16-member 2 (SLC16A2) gene. This syndrome leads to significant psychomotor disabilities, thyroid dysfunction, and abnormal brain development. This case report describes the genetic cause of AHDS in a male proband and to expanding the mutation spectrum of the SLC16A2 gene. Methods A blood specimen was collected from a one-year-old child with delayed development and abnormal thyroid function and this was followed by whole-exome sequencing (WES) was performed on the proband to identify potential genetic mutations. Sanger sequencing was subsequently used to confirm the findings and determine the inheritance pattern of the mutation within the family. Results The proband, who presented with developmental delay, thyroid dysfunction, and abnormal brain development, was found to have a novel hemizygous frameshift mutation, c.513_538del (p.Ile172Cysfs*60), in the SLC16A2 gene (NM_006517.5). This mutation was inherited from his asymptomatic mother, confirming the X-linked inheritance pattern. The mutation is classified as likely pathogenic, contributing to the clinical presentation observed in the proband. Conclusion This study identified a novel frameshift mutation in the SLC16A2 gene associated with AHDS, thereby expanding the known mutation spectrum of this gene. Given the significant impact of AHDS on neural development and hormone secretion, it is recommended that this gene be included in carrier screening panels in China, particularly for families with a history of related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Peng Lin
- Prenatal Diagnostic Centre, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
- Dongguan Key Laboratory of Precision Medicine for Prenatal Diagnosis of Genetic Diseases, Dongguan, Guangdong, People’s Republic of China
| | - Huituan Liu
- Department of Children’s Rehabilitation, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
| | - Jiwu Lou
- Prenatal Diagnostic Centre, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
- Dongguan Key Laboratory of Precision Medicine for Prenatal Diagnosis of Genetic Diseases, Dongguan, Guangdong, People’s Republic of China
| | - Guizhen Lyu
- Dongguan Key Laboratory of Clinical Medical Test Diagnostic Technology for Oncology, Dongguan Labway Medical Testing Laboratory Co., Ltd., Dongguan, Guangdong, People’s Republic of China
- Dongguan Molecular Diagnostic Technology and Infectious Disease Medical Test Engineering Research Centre, Dongguan Labway Medical Testing Laboratory Co., Ltd., Dongguan, Guangdong, People’s Republic of China
| | - Yanwei Li
- Dongguan Key Laboratory of Clinical Medical Test Diagnostic Technology for Oncology, Dongguan Labway Medical Testing Laboratory Co., Ltd., Dongguan, Guangdong, People’s Republic of China
| | - Peiqing He
- Prenatal Diagnostic Centre, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
- Dongguan Key Laboratory of Precision Medicine for Prenatal Diagnosis of Genetic Diseases, Dongguan, Guangdong, People’s Republic of China
| | - Youqing Fu
- Prenatal Diagnostic Centre, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
- Dongguan Key Laboratory of Precision Medicine for Prenatal Diagnosis of Genetic Diseases, Dongguan, Guangdong, People’s Republic of China
| | - Ronghua Zhang
- Prenatal Diagnostic Centre, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
- Dongguan Key Laboratory of Precision Medicine for Prenatal Diagnosis of Genetic Diseases, Dongguan, Guangdong, People’s Republic of China
| | - Yuqiong Zhang
- Department of Children’s Rehabilitation, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
| | - Tizhen Yan
- Prenatal Diagnostic Centre, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
- Dongguan Key Laboratory of Precision Medicine for Prenatal Diagnosis of Genetic Diseases, Dongguan, Guangdong, People’s Republic of China
| |
Collapse
|
2
|
Chakraborty S, Das D. Allan-Herndon-Dudley Syndrome. Indian J Pediatr 2025:10.1007/s12098-025-05507-9. [PMID: 40131620 DOI: 10.1007/s12098-025-05507-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
X-linked MCT 8 mutations cause Allan-Herndon-Dudley syndrome (AHDS), characterized by severe developmental delay and specific thyroid function abnormality. The report describes a 2-y-old boy who presented with severe developmental delay, generalized hypotonia and thyroid function abnormality (high FT3, low FT4 and normal TSH) suggesting a form of impaired thyroid hormone sensitivity. Whole exome sequencing (WES) analysis revealed mutation in exon 3 of MCT 8 gene. Improvement of neurodevelopmental delay and clinical and biochemical features of thyrotoxicosis occurs with use of tri iodoacetic acid.
Collapse
Affiliation(s)
| | - Debaditya Das
- Department of Endocrinology, I.P.G.M.E.R, kolkata, India.
| |
Collapse
|
3
|
Schreiner F, Vollbach H, Sonntag N, Schempp V, Gohlke B, Friese J, Woelfle J, Braun D, Schweizer U. Phenylbutyrate Treatment in a Boy With MCT8 Deficiency: Improvement of Thyroid Function Tests and Possible Hepatotoxicity. J Clin Endocrinol Metab 2025; 110:e992-e999. [PMID: 38781537 DOI: 10.1210/clinem/dgae356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024]
Abstract
CONTEXT Monocarboxylate transporter 8 (MCT8) deficiency is a rare X-chromosomal inherited disease leading to severe cognitive impairment, muscular hypotonia, and symptoms of peripheral thyrotoxicosis. Experimental approaches aiming to functionally rescue mutant MCT8 activity by the chemical chaperone phenylbutyrate (PB) demonstrated promising effects in vitro for several MCT8 missense mutations. OBJECTIVE The objective was to evaluate biochemical and clinical effects of PB in doses equivalent to those approved for the treatment of urea cycle disorders in a boy with MCT8 deficiency due to a novel MCT8 missense mutation c.703G>T (p.V235L). RESULTS During a treatment period of 13 months, PB led to a significant decrease of elevated thyrotropin and triiodothyronine (T3) serum concentrations, while free thyroxine (fT4) increased. The weight z-score of the toddler remained remarkably stable during the treatment period. Neurodevelopmental assessments (BSID-III) revealed a slight increase of gross motor skills from developmental age 4 to 6 months. However, increasing liver enzyme serum activities and accumulation of phenylacetate in urine led to treatment interruptions and dose alterations. In vitro analyses in MDCK1 cells confirmed the pathogenicity of MCT8 p.V235L. However, while PB increased expression of the mutant protein, it did not rescue T3 transport, suggesting a PB effect on thyroid function tests independent of restoring MCT8 activity. CONCLUSION In a clinical attempt of PB treatment in MCT8 deficiency we observed a significant improvement of thyroid hormone function tests, tendencies toward body weight stabilization and slight neurodevelopmental improvement. Hepatotoxicity of PB may be a limiting factor in MCT8 deficiency and requires further investigation.
Collapse
Affiliation(s)
- Felix Schreiner
- Pediatric Endocrinology Division, Children's Hospital, University Hospital Bonn, 53127 Bonn, Germany
| | - Heike Vollbach
- Pediatric Endocrinology Division, Children's Hospital, University Hospital Bonn, 53127 Bonn, Germany
| | - Niklas Sonntag
- Institute of Biochemistry and Molecular Biology, University Hospital Bonn, University of Bonn, 53115 Bonn, Germany
| | - Vera Schempp
- Pediatric Endocrinology Division, Children's Hospital, University Hospital Bonn, 53127 Bonn, Germany
| | - Bettina Gohlke
- Pediatric Endocrinology Division, Children's Hospital, University Hospital Bonn, 53127 Bonn, Germany
| | - Johannes Friese
- Department of Pediatric Neurology, Children's Hospital, University Hospital Bonn, 53127 Bonn, Germany
| | - Joachim Woelfle
- Pediatric Endocrinology Division, Children's Hospital, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Doreen Braun
- Institute of Biochemistry and Molecular Biology, University Hospital Bonn, University of Bonn, 53115 Bonn, Germany
| | - Ulrich Schweizer
- Institute of Biochemistry and Molecular Biology, University Hospital Bonn, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
4
|
Jakka SR, Mugesh G. Emerging Role of Noncovalent Interactions and Disulfide Bond Formation in the Cellular Uptake of Small Molecules and Proteins. Chem Asian J 2025; 20:e202401734. [PMID: 39831847 DOI: 10.1002/asia.202401734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 01/22/2025]
Abstract
Intracellular delivery of proteins and small molecules is an important barrier in the development of strategies to deliver functional proteins and therapeutics into the cells to realize their full potential in biotechnology, biomedicine, cell-based therapies, and gene editing protein systems. Most of the intracellular protein delivery strategies involve the conjugation of cell penetrating peptides to enable the permeability of plasma membrane of mammalian cells to allow proteins to enter cytosol. The conjugations of small molecules such as (p-methylphenyl) glycine, pyrenebutyrate and cysteines are used for the same purpose. Molecular level interactions are governed mostly by ionic (cationic/anionic), covalent and noncovalent interactions with various molecular entities of glycocalyx matrix on plasma membrane lipid bilayer. Although the role of noncovalent interactions in cellular uptake is not fully understood, several recent advances have focused on the noncovalent interaction-based strategies of intracellular delivery of small molecules and proteins into mammalian cells. These are achieved by simple modification of protein surfaces with chemical moieties which can form noncovalent interactions other than hydrogen bonding. In this review, we describe the recent advances and the mechanistic aspects of intracellular delivery and role of noncovalent interactions in the cellular uptake of proteins and small molecules.
Collapse
Affiliation(s)
- Surendar R Jakka
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560 012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560 012, India
| |
Collapse
|
5
|
Wilpert NM, Hewitt AL, Pons R, Henke MT, Dell'Orco A, Bauer M, Grolik C, Menz S, Wahle M, Zink A, Prigione A, Reinauer C, Lange C, Furth C, Brockmann K, Jung-Klawitter S, Christ S, Kaindl AM, Tietze A, Krude H, Opladen T, Schuelke M. Patients with Allan-Herndon-Dudley Syndrome (MCT8 Deficiency) Display Symptoms of Parkinsonism in Childhood and Respond to Levodopa/Carbidopa Treatment. Mov Disord 2025. [PMID: 40088079 DOI: 10.1002/mds.30152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/25/2024] [Accepted: 01/27/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Patients with mutations in the monocarboxylate transporter 8 (MCT8, SLC16A2) suffer from X-linked recessive Allan-Herndon-Dudley syndrome (AHDS), which is characterized by developmental delay and a severe movement disorder. Current trials using thyroid hormone derivatives to overcome the transporter defect have failed to achieve patient-oriented therapeutic goals. OBJECTIVES Our aim was to define the type of movement disorder in AHDS in an observational cohort study and to investigate the causative role of the dopaminergic system. METHODS We present longitudinal clinical data from the DEEPTYPE registry of 11 patients with video documentation, standardized phenotyping, cerebrospinal fluid (CSF) analysis, neuroimaging data, and the treatment response to levodopa/carbidopa supplementation. RESULTS Children presented with signs of childhood parkinsonism, including hypokinesia, hypomimia, inability to sit or stand, rigidity, dystonia, and autonomic dysfunction. CSF homovanillic acid concentrations were decreased (n = 12), suggesting an isolated dopamine pathway impairment. Seven out of 8 patients responded favorably to l-dopa/carbidopa supplementation and we did not observe any adverse drug reactions. CONCLUSIONS AHDS is associated with childhood parkinsonism, which is linked with biochemical abnormalities of dopamine metabolism. It can be treated with l-dopa/carbidopa supplementation. However, further research is needed to elucidate the exact effect of MCT8 deficiency on dopamine metabolism. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Nina-Maria Wilpert
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Center for Chronically Sick Children, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- BIH Charité Junior Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Berlin, Germany
| | - Angela L Hewitt
- Division of Movement Disorders, Department of Neurology, University of Rochester Medical Center, Motor Physiology and Neuromodulation Program, Rochester, New York, USA
- Division of Child Neurology, Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Roser Pons
- First Department of Pediatrics, National and Kapodistrian University of Athens, Hospital Agia Sofía, Pediatric Neurology Unit, Athens, Greece
| | - Marie-Thérèse Henke
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Andrea Dell'Orco
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Neuroradiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Martin Bauer
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Medical Psychology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Christiane Grolik
- Department of Pediatrics, Pediatric Neurology, Kinderkrankenhaus Amsterdamer Strasse, Kliniken der Stadt Köln, Cologne, Germany
| | - Stephan Menz
- Center for Chronically Sick Children, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Monika Wahle
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Annika Zink
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Christina Reinauer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Catharina Lange
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Knut Brockmann
- Department of Pediatrics and Adolescent Medicine, University Medical Center, Göttingen, Germany
| | - Sabine Jung-Klawitter
- Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Heidelberg, Germany
| | - Stine Christ
- Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Heidelberg, Germany
| | - Angela M Kaindl
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Center for Chronically Sick Children, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Institute for Cell and Neurobiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- partner site Berlin, German Center for Child and Adolescent Health (DZKJ), Section CNS Development and Neurological Disease, Berlin, Germany
| | - Anna Tietze
- Institute of Neuroradiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Heiko Krude
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Thomas Opladen
- Department I, Division of Pediatric Neurology and Metabolic Medicine, Heidelberg University, Medical Faculty Heidelberg, Center for Pediatric and Adolescent Medicine, Heidelberg, Germany
| | - Markus Schuelke
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Center for Chronically Sick Children, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- partner site Berlin, German Center for Child and Adolescent Health (DZKJ), Section CNS Development and Neurological Disease, Berlin, Germany
| |
Collapse
|
6
|
Groeneweg S, van Geest FS, Martín M, Dias M, Frazer J, Medina-Gomez C, Sterenborg RBTM, Wang H, Dolcetta-Capuzzo A, de Rooij LJ, Teumer A, Abaci A, van den Akker ELT, Ambegaonkar GP, Armour CM, Bacos I, Bakhtiani P, Barca D, Bauer AJ, van den Berg SAA, van den Berge A, Bertini E, van Beynum IM, Brunetti-Pierri N, Brunner D, Cappa M, Cappuccio G, Castellotti B, Castiglioni C, Chatterjee K, Chesover A, Christian P, Coenen-van der Spek J, de Coo IFM, Coutant R, Craiu D, Crock P, DeGoede C, Demir K, Dewey C, Dica A, Dimitri P, Dremmen MHG, Dubey R, Enderli A, Fairchild J, Gallichan J, Garibaldi L, George B, Gevers EF, Greenup E, Hackenberg A, Halász Z, Heinrich B, Hurst AC, Huynh T, Isaza AR, Klosowska A, van der Knoop MM, Konrad D, Koolen DA, Krude H, Kulkarni A, Laemmle A, LaFranchi SH, Lawson-Yuen A, Lebl J, Leeuwenburgh S, Linder-Lucht M, López Martí A, Lorea CF, Lourenço CM, Lunsing RJ, Lyons G, Malikova JK, Mancilla EE, McCormick KL, McGowan A, Mericq V, Lora FM, Moran C, Muller KE, Nicol LE, Oliver-Petit I, Paone L, Paul PG, Polak M, Porta F, Poswar FO, Reinauer C, Rozenkova K, Seckold R, Seven Menevse T, Simm P, Simon A, Singh Y, Spada M, Stals MAM, Stegenga MT, Stoupa A, et alGroeneweg S, van Geest FS, Martín M, Dias M, Frazer J, Medina-Gomez C, Sterenborg RBTM, Wang H, Dolcetta-Capuzzo A, de Rooij LJ, Teumer A, Abaci A, van den Akker ELT, Ambegaonkar GP, Armour CM, Bacos I, Bakhtiani P, Barca D, Bauer AJ, van den Berg SAA, van den Berge A, Bertini E, van Beynum IM, Brunetti-Pierri N, Brunner D, Cappa M, Cappuccio G, Castellotti B, Castiglioni C, Chatterjee K, Chesover A, Christian P, Coenen-van der Spek J, de Coo IFM, Coutant R, Craiu D, Crock P, DeGoede C, Demir K, Dewey C, Dica A, Dimitri P, Dremmen MHG, Dubey R, Enderli A, Fairchild J, Gallichan J, Garibaldi L, George B, Gevers EF, Greenup E, Hackenberg A, Halász Z, Heinrich B, Hurst AC, Huynh T, Isaza AR, Klosowska A, van der Knoop MM, Konrad D, Koolen DA, Krude H, Kulkarni A, Laemmle A, LaFranchi SH, Lawson-Yuen A, Lebl J, Leeuwenburgh S, Linder-Lucht M, López Martí A, Lorea CF, Lourenço CM, Lunsing RJ, Lyons G, Malikova JK, Mancilla EE, McCormick KL, McGowan A, Mericq V, Lora FM, Moran C, Muller KE, Nicol LE, Oliver-Petit I, Paone L, Paul PG, Polak M, Porta F, Poswar FO, Reinauer C, Rozenkova K, Seckold R, Seven Menevse T, Simm P, Simon A, Singh Y, Spada M, Stals MAM, Stegenga MT, Stoupa A, Subramanian GM, Szeifert L, Tonduti D, Turan S, Vanderniet J, van der Walt A, Wémeau JL, van Wermeskerken AM, Wierzba J, de Wit MCY, Wolf NI, Wurm M, Zibordi F, Zung A, Zwaveling-Soonawala N, Rivadeneira F, Meima ME, Marks DS, Nicola JP, Chen CH, Medici M, Visser WE. Mapping variants in thyroid hormone transporter MCT8 to disease severity by genomic, phenotypic, functional, structural and deep learning integration. Nat Commun 2025; 16:2479. [PMID: 40075072 PMCID: PMC11904026 DOI: 10.1038/s41467-025-56628-w] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 01/23/2025] [Indexed: 03/14/2025] Open
Abstract
Predicting and quantifying phenotypic consequences of genetic variants in rare disorders is a major challenge, particularly pertinent for 'actionable' genes such as thyroid hormone transporter MCT8 (encoded by the X-linked SLC16A2 gene), where loss-of-function (LoF) variants cause a rare neurodevelopmental and (treatable) metabolic disorder in males. The combination of deep phenotyping data with functional and computational tests and with outcomes in population cohorts, enabled us to: (i) identify the genetic aetiology of divergent clinical phenotypes of MCT8 deficiency with genotype-phenotype relationships present across survival and 24 out of 32 disease features; (ii) demonstrate a mild phenocopy in ~400,000 individuals with common genetic variants in MCT8; (iii) assess therapeutic effectiveness, which did not differ among LoF-categories; (iv) advance structural insights in normal and mutated MCT8 by delineating seven critical functional domains; (v) create a pathogenicity-severity MCT8 variant classifier that accurately predicted pathogenicity (AUC:0.91) and severity (AUC:0.86) for 8151 variants. Our information-dense mapping provides a generalizable approach to advance multiple dimensions of rare genetic disorders.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Ferdy S van Geest
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Mariano Martín
- Department of Clinical Biochemistry (CIBICI-CONICET), Faculty of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mafalda Dias
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jonathan Frazer
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rosalie B T M Sterenborg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hao Wang
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Anna Dolcetta-Capuzzo
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Linda J de Rooij
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Alexander Teumer
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Ayhan Abaci
- Division of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Erica L T van den Akker
- Department of Paediatrics, Division of Endocrinology, Erasmus Medical Centre -Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Gautam P Ambegaonkar
- Department of Paediatric Neurology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Christine M Armour
- Regional Genetics Program, Children's Hospital of Eastern Ontario and Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Iiuliu Bacos
- Centrul Medical Dr. Bacos Cosma, Timisoara, Romania
| | - Priyanka Bakhtiani
- University of Louisville, Louisville, KY, USA
- Childrens Hospital Los Angeles, Los Angeles, CA, USA
| | - Diana Barca
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest, Romania
| | - Andrew J Bauer
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sjoerd A A van den Berg
- Diagnostic Laboratory for Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Amanda van den Berge
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesu' Children's Research Hospital IRCCS, Rome, Italy
| | - Ingrid M van Beynum
- Department of Pediatrics, Division of Pediatric Cardiology, Erasmus Medical Centre - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, 80131, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy
| | - Doris Brunner
- Gottfried Preyer's Children Hospital, Vienna, Austria
| | - Marco Cappa
- Research Area for Innovative Therapies in Endocrinopathies, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gerarda Cappuccio
- Department of Translational Medicine, Federico II University, 80131, Naples, Italy
- Neurological Research Institute and Baylor College of Medicine, Houston, TX, USA
| | - Barbara Castellotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudia Castiglioni
- Department of Neurology, Clinica Meds, School of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Krishna Chatterjee
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Alexander Chesover
- Division of Endocrinology, The Hospital for Sick Children and Department of Paediatrics, University of Toronto, Toronto, M5G 1×8, Canada
- Department of Endocrinology, Great Ormond Street Hospital for Children, London, UK
| | - Peter Christian
- East Kent Hospitals University NHS Foundation Trust, Ashford, UK
| | - Jet Coenen-van der Spek
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Irenaeus F M de Coo
- Department of Toxicogenomics, Unit Clinical Genomics, Maastricht University, MHeNs School for Mental Health and Neuroscience, Maastricht, The Netherlands
| | - Regis Coutant
- Department of Pediatric Endocrinology and Diabetology, University Hospital, Angers, France
| | - Dana Craiu
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest, Romania
| | - Patricia Crock
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Christian DeGoede
- Department of Paediatric Neurology, Clinical Research Facility, Lancashire Teaching Hospitals NHS Trust, Lancashire, UK
| | - Korcan Demir
- Division of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Cheyenne Dewey
- Genomics Institute Mary Bridge Children's Hospital, MultiCare Health System, Tacoma, WA, USA
| | - Alice Dica
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest, Romania
| | - Paul Dimitri
- The Department of Oncology and Metabolism, The University of Sheffield, Western Bank, Sheffield, S10, 2TH, UK
| | - Marjolein H G Dremmen
- Division of Paediatric Radiology, Erasmus Medical Centre - Sophia's Children Hospital, Rotterdam, The Netherlands
| | | | - Anina Enderli
- Department of Neuropediatrics, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Jan Fairchild
- Department of Diabetes and Endocrinology, Women's and Children's Hospital, North Adelaide, 5066, South Australia, Australia
| | | | | | - Belinda George
- Department of Endocrinology, St. John's Medical College Hospital, Bengaluru, India
| | - Evelien F Gevers
- Centre for Endocrinology, William Harvey Research institute, Queen Mary University of London, London, UK
| | - Erin Greenup
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Pediatric Endocrinology, Department of Pediatrics, Orlando Health Arnold Palmer Hospital for Children, Orlando, FL, USA
| | - Annette Hackenberg
- Department of Neuropediatrics, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Zita Halász
- Pediatric Center, Semmelweis University Budapest, Budapest, Hungary
| | - Bianka Heinrich
- Department of Neuropediatrics, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Anna C Hurst
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tony Huynh
- Department of Endocrinology & Diabetes, Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - Amber R Isaza
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anna Klosowska
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - David A Koolen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Heiko Krude
- Institute of Experimental Paediatric Endocrinology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Abhishek Kulkarni
- Department of Paediatric Endocrinology, SRCC Children's Hospital, Mumbai, India
| | - Alexander Laemmle
- Institute of Clinical Chemistry and Department of Pediatrics, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Stephen H LaFranchi
- Department of Pediatrics, Doernbecher Children's Hospital, Oregon Health & Sciences University, Portland, OR, USA
| | - Amy Lawson-Yuen
- Genomics Institute Mary Bridge Children's Hospital, MultiCare Health System, Tacoma, WA, USA
- Department of Genetics, Kaiser Permanente Washington, Seattle, WA, USA
| | - Jan Lebl
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Selmar Leeuwenburgh
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Michaela Linder-Lucht
- Division of Neuropediatrics and Muscular Disorders, Department of Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Anna López Martí
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Cláudia F Lorea
- Teaching Hospital of Universidade Federal de Pelotas, Pelotas, Brazil
- Federal University of Rio Grande do Sul, Porto Alegre-RS, Brazil
| | - Charles M Lourenço
- National Reference Center for Rare Diseases, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
- Personalized Medicine area -Special Education Sector at DLE/Grupo Pardini, Rio de Janeiro, Brazil
| | - Roelineke J Lunsing
- Department of Child Neurology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Greta Lyons
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jana Krenek Malikova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Edna E Mancilla
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kenneth L McCormick
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anne McGowan
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Veronica Mericq
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile, Department of Pediatrics, Clinica Las Condes, Santiago, Chile
| | - Felipe Monti Lora
- Pediatric Endocrinology Group, Sabara Children's Hospital, São Paulo, Brazil
| | - Carla Moran
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | | | - Lindsey E Nicol
- Department of Pediatrics, Doernbecher Children's Hospital, Oregon Health & Sciences University, Portland, OR, USA
| | - Isabelle Oliver-Petit
- Department of Paediatric Endocrinology and Genetics, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Laura Paone
- Endocrinology and Diabetology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Praveen G Paul
- Department of Paediatrics, Christian Medical College, Vellore, India
| | - Michel Polak
- Paediatric Endocrinology, Diabetology and Gynaecology, Department, Necker Children's University Hospital, Imagine Institute Affiliate, Université de Paris Cité, Paris, France
| | - Francesco Porta
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Turin, Italy
| | - Fabiano O Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Christina Reinauer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Medical Faculty, Dusseldorf, Germany
| | - Klara Rozenkova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Rowen Seckold
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Tuba Seven Menevse
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Peter Simm
- Royal Children's Hospital/University of Melbourne, Parkville, Australia
| | - Anna Simon
- Department of Paediatrics, Christian Medical College, Vellore, India
| | - Yogen Singh
- Department of Paediatric Cardiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pediatrics, University of California - UC Davis Children's Hospital, Sacramento, CA, USA
| | - Marco Spada
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Turin, Italy
| | - Milou A M Stals
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Merel T Stegenga
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Athanasia Stoupa
- Paediatric Endocrinology, Diabetology and Gynaecology, Department, Necker Children's University Hospital, Imagine Institute Affiliate, Université de Paris Cité, Paris, France
| | - Gopinath M Subramanian
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Lilla Szeifert
- Pediatric Center, Semmelweis University Budapest, Budapest, Hungary
| | - Davide Tonduti
- Child Neurology Unit - C.O.A.L.A. (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Milano, Italy
- Department of Clinical and Biomedical Science, Università degli Studi di Milano, Milano, Italy
| | - Serap Turan
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Joel Vanderniet
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Adri van der Walt
- Private paediatric Neurology practice Dr A van der Walt, Durbanville, South Africa
| | | | | | - Jolanta Wierzba
- Department of Internal and Pediatric Nursing, Institute of Nursing and Midwifery, Medical University of Gdańsk, Gdańsk, Poland
| | - Marie-Claire Y de Wit
- Department of Paediatric Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Michael Wurm
- University Children's Hospital Regensburg (KUNO), University of Regensburg, Campus St. Hedwig, Regensburg, Germany
| | - Federica Zibordi
- Child Neurology Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Amnon Zung
- Pediatric Endocrinology Unit, Kaplan Medical center, Rehovot and the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nitash Zwaveling-Soonawala
- Emma Children's Hospital, Department of Paediatric Endocrinology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marcel E Meima
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Debora S Marks
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Juan P Nicola
- Department of Clinical Biochemistry (CIBICI-CONICET), Faculty of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Chi-Hua Chen
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Marco Medici
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - W Edward Visser
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Yoon JG, Lee S, Park S, Jang SS, Cho J, Kim MJ, Kim SY, Kim WJ, Lee JS, Chae JH. Identification of a novel non-coding deletion in Allan-Herndon-Dudley syndrome by long-read HiFi genome sequencing. BMC Med Genomics 2025; 18:41. [PMID: 40033291 DOI: 10.1186/s12920-024-02058-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 11/27/2024] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Allan-Herndon-Dudley syndrome (AHDS) is an X-linked disorder caused by pathogenic variants in the SLC16A2 gene. Although most reported variants are found in protein-coding regions or adjacent junctions, structural variations (SVs) within non-coding regions have not been previously reported. METHODS We investigated two male siblings with severe neurodevelopmental disorders and spasticity, who had remained undiagnosed for over a decade and were negative from exome sequencing, utilizing long-read HiFi genome sequencing. We conducted a comprehensive analysis including short-tandem repeats (STRs) and SVs to identify the genetic cause in this familial case. RESULTS While coding variant and STR analyses yielded negative results, SV analysis revealed a novel hemizygous deletion in intron 1 of the SLC16A2 gene (chrX:74,460,691 - 74,463,566; 2,876 bp), inherited from their carrier mother and shared by the siblings. Determination of the breakpoints indicates that the deletion probably resulted from Alu/Alu-mediated rearrangements between homologous AluY pairs. The deleted region is predicted to include multiple transcription factor binding sites, such as Stat2, Zic1, Zic2, and FOXD3, which are crucial for the neurodevelopmental process, as well as a regulatory element including an eQTL (rs1263181) that is implicated in the tissue-specific regulation of SLC16A2 expression, notably in skeletal muscle and thyroid tissues. CONCLUSIONS This report, to our knowledge, is the first to describe a non-coding deletion associated with AHDS, demonstrating the potential utility of long-read sequencing for undiagnosed patients. Although interpreting variants in non-coding regions remains challenging, our study highlights this region as a high priority for future investigation and functional studies.
Collapse
Affiliation(s)
- Jihoon G Yoon
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seungbok Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soojin Park
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Se Song Jang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jaeso Cho
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Man Jin Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Soo Yeon Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woo Joong Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin Sook Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong-Hee Chae
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Garrelfs MR, Mooij CF, Boelen A, van Trotsenburg ASP, Zwaveling-Soonawala N. Newborn screening for central congenital hypothyroidism: past, present and future. Eur Thyroid J 2025; 14:e240329. [PMID: 39913280 PMCID: PMC11883867 DOI: 10.1530/etj-24-0329] [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: 10/14/2024] [Revised: 01/17/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Congenital hypothyroidism (CH) is defined as thyroid hormone deficiency at birth and constitutes one of the most common causes of preventable intellectual disability worldwide. Central CH is caused by insufficient pituitary or hypothalamic control of thyroid function, biochemically characterized by a low serum free thyroxine (fT4), in combination with a low, normal or mildly elevated thyroid-stimulating hormone (TSH). Central CH is less common than primary CH and is part of multiple pituitary hormone deficiencies (MPHD) in most of the cases. MPHD at birth, also known as 'congenital hypopituitarism', is a potentially life-threatening condition due to the possible co-occurrence of adrenocorticotropin hormone and growth hormone deficiency that can result in severe hypoglycemia and adrenal crisis. To date, central CH is the only pituitary hormone deficiency suitable for newborn screening (NBS), providing an opportunity for early detection of MPHD. Even though the first NBS programs utilized T4-based methods that were able to identify central CH, most countries have since transitioned to TSH-based approaches due to the high rate of false positives associated with T4-based strategies. Now, 50 years after the introduction of NBS for CH, only a few countries around the world have a screening program capable of detecting central CH. In this paper, we review the past, present and future of NBS for central CH. We will outline the importance of early detection of central CH and discuss the challenges and opportunities of screening for this condition.
Collapse
Affiliation(s)
- Mark R Garrelfs
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam and Vrije Universiteit, European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Christiaan F Mooij
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam and Vrije Universiteit, European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Anita Boelen
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A S Paul van Trotsenburg
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam and Vrije Universiteit, European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Nitash Zwaveling-Soonawala
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam and Vrije Universiteit, European Reference Network on Rare Endocrine Conditions (Endo-ERN), Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Scholtes N, Jelesch E, Diesener P, Stoffels JC, Völkl TMK. Swallowing Assessment in a Pediatric Case of Allan-Herndon-Dudley Syndrome (MCT8 Deficiency): Advanced Insights into Dysphagia via Flexible Endoscopic Evaluation of Swallowing. Neuropediatrics 2025. [PMID: 39681133 DOI: 10.1055/a-2502-6417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Patients with MCT8 deficiency often present with underweight and are prone to frequent pulmonary infections, including aspiration pneumonia. Despite commonly reported swallowing difficulties in this population, specific dysphagia symptoms have not been well-documented. We conducted a flexible endoscopic evaluation of swallowing (FEES) on a young boy diagnosed with MCT8 deficiency, who exhibited recurrent pulmonary infections and failed to achieve substantial weight gain despite an oral energy intake appropriate for his age and height. The FEES revealed generally weakened swallowing mechanisms, characterized by prolonged swallow and cough sequences, along with penetration and aspiration of both fluid and semi-solid test boluses. Given the considerable effort associated with oral intake, we hypothesize that dysphagia contributes to his underweight status, alongside peripheral thyrotoxicosis. In conclusion, FEES proved to be an invaluable tool in identifying underlying swallowing impairments and assessing the need for gastrostomy in this patient. For MCT8 deficiency, patients presenting with underweight, frequent pulmonary infections, and swallowing difficulties, it is recommended that diagnostic evaluations include FEES to thoroughly assess their swallowing function and airway protection.
Collapse
Affiliation(s)
- Nina Scholtes
- Division of Neuropediatrics and Integrated Health Care, Department of Pediatrics and Adolescent Medicine, KJF Klinikum Josefinum, Augsburg, Germany
| | - Evelyn Jelesch
- Division of Neuropediatrics and Integrated Health Care, Department of Pediatrics and Adolescent Medicine, KJF Klinikum Josefinum, Augsburg, Germany
| | - Paul Diesener
- Center for Dysphagia and Tracheostomy Care, Hegau-Jugendwerk, Gailingen am Hochrhein, Germany
| | - Johannes C Stoffels
- Division of Neuropediatrics and Integrated Health Care, Department of Pediatrics and Adolescent Medicine, KJF Klinikum Josefinum, Augsburg, Germany
| | - Thomas M K Völkl
- Division of Pediatric Endocrinology and Diabetology, Department of Pediatrics and Adolescent Medicine, KJF Klinikum Josefinum, Augsburg, Germany
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
10
|
Çelik N, Demir K, Dibeklioğlu SE, Dündar BN, Hatipoğlu N, Mutlu GY, Arslan E, Yıldırımçakar D, Çayır A, Hacıhamdioğlu B, Sütçü ZK, Ünsal Y, Karagüzel G. Clinical and genetic characteristics of patients with monocarboxylate transporter-8 deficiency: a multicentre retrospective study. Eur J Pediatr 2024; 184:92. [PMID: 39699593 DOI: 10.1007/s00431-024-05931-7] [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: 08/26/2024] [Revised: 12/01/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Allan-Herndon-Dudley syndrome is a neurodevelopmental disorder characterized by motor and intellectual disabilities. Despite its rarity, there has been a rise in interest due to ongoing research and emerging therapy suggestions. In this multicenter, retrospective, cross-sectional study, the genetic characteristics and clinical data of twenty-one cases of genetically confirmed MCT8 deficiency were evaluated. The median age at the diagnosis was 2.4 (1.29; 5.9) years, which ranged from 0.5 to 14.0 years. The median follow-up period was 2.34 years, ranging from four months to 7.9 years. In 21 patients, 17 different variants were detected in the SLC16A2 gene. Eleven of these variants (c.1456delC, c.439G > T, c.949C > A, c.1392dupC, c.1612C > T, c.407dup, c.781del, c.589C > A, c.712G > A, c.311 T > A, c.1461del) have not been previously reported. In this study, with the exception of three cases with fT3/fT4 ratios of 4.95, 3.58, and 4.52, all cases exhibited fT3/fT4 ratios higher than five (9.9 (7.9; 12.0)). CONCLUSION MCT8 deficiency is a rare and devastating disorder characterized by central hypothyroidism and peripheral thyrotoxicosis. The fT3/fT4 ratio can be used as a useful diagnostic indicator of MCT8 deficiency in males with mental and motor retardation. There is a need to raise clinicians' awareness of this potentially treatable condition with the emergence of new and promising treatments. WHAT IS KNOWN • Allan-Herndon-Dudley syndrome, also known as MCT8 deficiency is a rare and devastating disorder characterized by central hypothyroidism and peripheral thyrotoxicosis. WHAT IS NEW • In this study, seventeen different variants were detected in the SLC16A2 gene, eleven of which (c.1456delC; c.439G>T; c.949C>A; c.1392dupC; c.1612C>T; c.407dup; c.781del; c.589C>A; c.712G>A; c.311T>A; c.1461del) have not been reported before. • The fT3/fT4 ratio can be used as a useful diagnostic indicator of MCT8 deficiency in males with mental and motor retardation.
Collapse
Affiliation(s)
- Nurullah Çelik
- Department of Pediatric Endocrinology, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey.
| | - Korcan Demir
- Department of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | | | - Bumin Nuri Dündar
- Department of Pediatric Endocrinology, Faculty of Medicine, İzmir Katip Celebi University, Izmir, Turkey
| | - Nihal Hatipoğlu
- Department of Pediatric Endocrinology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Gül Yeşiltepe Mutlu
- Department of Pediatric Endocrinology and Diabetes, Koç University School of Medicine, Istanbul, Turkey
| | - Emrullah Arslan
- Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, Ege University, Izmir, Turkey
| | - Didem Yıldırımçakar
- Department of Pediatric Endocrinology, Denizli State Hospital, Denizli, Turkey
| | - Atilla Çayır
- Department of Pediatric Endocrinology and Diabetes, Erzurum Education and Research Hospital, University of Health Science, Erzurum, Turkey
| | - Bülent Hacıhamdioğlu
- Department of Pediatric Endocrinology, Faculty of Medicine, İstanbul Aydın University, Istanbul, Turkey
| | - Zümrüt Kocabey Sütçü
- Başakşehir Çam and Sakura City Hospital, Pediatric Endocrinology, Istanbul, Turkey
| | - Yağmur Ünsal
- Clinic of Pediatric Endocrinology, Şanlıurfa Education and Research Hospital, Şanlıurfa, Turkey
| | - Gülay Karagüzel
- Department of Pediatric Endocrinology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey.
| |
Collapse
|
11
|
Weiss RE, Lemos JRN, Dumitrescu AM, Islam MS, Hirani K, Refetoff S. Combined Levothyroxine and Propylthiouracil Treatment in Children with Monocarboxylate Transporter 8 Deficiency: A Multicenter Case Series of 12 Patients. Thyroid 2024; 34:1435-1443. [PMID: 39283825 PMCID: PMC11631802 DOI: 10.1089/thy.2024.0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Objective: To evaluate the combined administration of propylthiouracil (PTU) and levothyroxine (LT4) in managing monocarboxylate transporter 8 (MCT8) deficiency and identify optimal therapeutic dosages. Methods: This multicenter case series involved 12 male patients with MCT8 deficiency whose parents/guardians consented to PTU and LT4 treatment. Data were collected from January 2008 to June 24, 2024. The study focused on treatment safety and outcomes, analyzing baseline and last encounter biochemical, metabolic, and anthropometric parameters. Statistical analyses included Wilcoxon signed ranks tests and generalized estimated equations to assess effects on thyroid and metabolic markers, and receiver operating characteristics curves to predict optimal dose. Results: Patients showed a significant reduction in serum total triiodothyronine (TT3) concentration and TT3/TT4 ratio, with increased serum TT4 and free T4 (fT4) concentrations. The use of PTU effectively reduced TT3 concentration by 25% at an average dose of 6.8 mg/kg/day, while LT4 increased fT4 concentration by 40% from baseline at an average dose of 4.3 µg/kg/day. Thyrotropin concentration was undetectable on treatment. No statistical differences were observed in metabolic and physical parameters between baseline and last encounter overall for the group, but six of eight patients for whom these data were available had an increase in weight (z-score). There were no adverse effects on liver function or granulocyte numbers noted throughout the period of observation. Conclusion: Combined treatment with PTU and LT4 normalized serum T3, fT4, and TT4 in patients with MCT8 deficiency. Individualized dose adjustments were crucial for achieving therapeutic goals, indicating the need for personalized treatment plans.
Collapse
Affiliation(s)
- Roy E. Weiss
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Joana R. N. Lemos
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alexandra M. Dumitrescu
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
- Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, Illinois, USA
| | - Mohammad S. Islam
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Khemraj Hirani
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
- Department of Pediatrics and Committee on Genetics, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
12
|
Yu L, Liu Y, Wang Y, Wang G, Xiao X, Wang H, Wang H, Sun H, Wang G. Increased thyroid hormone sensitivity is correlated with visceral obesity in patients with type 2 diabetes. Lipids Health Dis 2024; 23:337. [PMID: 39415187 PMCID: PMC11481250 DOI: 10.1186/s12944-024-02320-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
OBJECTIVE The study aimed to assess whether thyroid hormone (TH) sensitivity is related to visceral fat area (VFA) and visceral obesity in euthyroid subjects with type 2 diabetes (T2D). METHODS 750 euthyroid patients with T2D were enrolled. A VFA of 80 cm2 or more was considered visceral obesity. Central TH sensitivity was conducted using thyrotrophic thyroxine resistance index (TT4RI), thyrotropin index (TSHI), and thyroid feedback quantile-based index (TFQI). Free triiodothyronine to free thyroxine (FT3/FT4) was utilized for assessing peripheral TH sensitivity. RESULTS The subjects had a mean age of 51.5 ± 11.1 years, and 540 (72.0%) of them were men. In multivariable regression analyses, there was a positive correlation of FT3/FT4 tertile with visceral obesity, after full adjustment for confounding variables (P < 0.05). The middle and highest FT3/FT4 tertiles were correlated with a 134% [95% CI (1.24, 4.44)] and 98% [95% CI (1.04, 3.78)] higher prevalence of visceral obesity than the lowest tertile, respectively. Conversely, elevated TFQI levels were linked to a decreased prevalence of visceral obesity. Stratified analysis revealed that these associations were particularly pronounced in participants who are neither overweight nor obese and those aged less than 60 years (all P < 0.05). CONCLUSIONS Higher TH sensitivity is correlated with visceral obesity and elevated VFA in euthyroid patients with T2D, particularly among those younger than 60 years and individuals who are neither overweight nor obese.
Collapse
Affiliation(s)
- Lu Yu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
- Department of Endocrinology and Metabolism, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Yujia Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yingxuan Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Gang Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xianchao Xiao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Huan Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China
| | - Hanyu Wang
- Department of Endocrinology and Metabolism, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Hui Sun
- Department of Endocrinology and Metabolism, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
13
|
Zung A, Sonntag N, Schweizer U, Banne E, Braun D. Glycerol Phenylbutyrate Treatment of 2 Patients With Monocarboxylate Transporter 8 Deficiency. J Clin Endocrinol Metab 2024; 109:2589-2601. [PMID: 38469646 DOI: 10.1210/clinem/dgae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
CONTEXT Monocarboxylate transporter 8 (MCT8) deficiency is a rare genetic disease that leads to severe global developmental delay. MCT8 facilitates thyroid hormone (TH) transport across the cell membrane, and the serum TH profile is characterized by high T3 and low T4 levels. Recent studies have shown that the chemical chaperone sodium phenylbutyrate (NaPB) restored mutant MCT8 function and increased TH content in patient-derived induced pluripotent stem cells, making it a potential treatment for MCT8 deficiency. OBJECTIVE We aimed to assess the efficacy and safety of glycerol phenylbutyrate (GPB) in MCT8 deficiency. METHODS We treated 2 monozygotic twins aged 14.5 years with MCT8 deficiency due to P321L mutation with escalating doses of GPB over 13 months. We recorded TH, vital signs, anthropometric measurements, and neurocognitive functions. Resting metabolic rate (RMR) was measured by indirect calorimetry. Serum metabolites of GPB were monitored as a safety measure. In vitro effects of NaPB were evaluated in MDCK1 cells stably expressing the MCT8P321L mutation. The effects of GPB were compared to the effects of DITPA and TRIAC, thyromimetic medications that the patients had received in the past. RESULTS NaPB restored mutant MCT8 expression in MDCK1 cells and increased T3 transport into cells carrying the P321L mutation. GPB treatment reduced high T3 and increased low T4 levels. The patients showed a significant weight gain simultaneously with a reduction in RMR. Only minor neurocognitive improvement was observed, in hyperreflexia score and in cognitive functions. Serum metabolites did not exceed the toxic range, but elevated liver transaminases were observed. CONCLUSION In the first report of GPB treatment in MCT8 deficiency we found an improvement in TH profile and body mass index, with minor neurodevelopmental changes.
Collapse
Affiliation(s)
- Amnon Zung
- Pediatric Endocrinology Unit, Kaplan Medical Center, Rehovot 76100, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 12000, Israel
| | - Niklas Sonntag
- Institut für Biochemie und Molekularbiologie, Medizinische Fakultät, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn 53115, Germany
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Medizinische Fakultät, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn 53115, Germany
| | - Ehud Banne
- The Genetic Institute, Edith Wolfson Medical Center, Holon 5822012, Israel
| | - Doreen Braun
- Institut für Biochemie und Molekularbiologie, Medizinische Fakultät, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn 53115, Germany
| |
Collapse
|
14
|
Masser BE, Brinkmeier ML, Lin Y, Liu Q, Miyazaki A, Nayeem J, Cheung LYM. Gene Misexpression in a Smoc2+ve/ Sox2-Low Population in Juvenile Prop1-Mutant Pituitary Gland. J Endocr Soc 2024; 8:bvae146. [PMID: 39253355 PMCID: PMC11382140 DOI: 10.1210/jendso/bvae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Indexed: 09/11/2024] Open
Abstract
Mutations in the pituitary-specific transcription factor Prophet of Pit-1 (PROP1) are the most common genetic etiology of combined pituitary hormone deficiency (CPHD). CPHD is associated with short stature, attributable to growth hormone deficiency and/or thyroid-stimulating hormone deficiency, as well as hypothyroidism and infertility. Pathogenic lesions impair pituitary development and differentiation of endocrine cells. We performed single-cell RNA sequencing of pituitary cells from a wild-type and a Prop1-mutant P4 female mouse to elucidate population-specific differential gene expression. We observed a Smoc2+ve population that expressed low Sox2, which trajectory analyses suggest are a transitional cell state as stem cells differentiate into endocrine cells. We also detected ectopic expression of Sox21 in these cells in the Prop1df/df mutant. Prop1-mutant mice are known to overexpress Pou3f4, which we now show to be also enriched in this Smoc2+ve population. We sought to elucidate the role of Pou3f4 during pituitary development and to determine the contributions of Pou3f4 upregulation to pituitary disease by utilizing double-mutant mice lacking both Prop1 and Pou3f4. However, our data showed that Pou3f4 is not required for normal pituitary development and function. Double mutants further demonstrated that the upregulation of Pou3f4 was not causative for the overexpression of Sox21. These data indicate loss of Pou3f4 is not a potential cause of CPHD, and further studies may investigate the functional consequence of upregulation of Pou3f4 and Sox21, if any, in the novel Smoc2+ve cell population.
Collapse
Affiliation(s)
- Bailey E Masser
- Department of Human Genetics, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48105, USA
| | - Michelle L Brinkmeier
- Department of Human Genetics, University of Michigan Medical School, University of Michigan, Ann Arbor, MI 48105, USA
| | - Yuxuan Lin
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Qin Liu
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Aya Miyazaki
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jannatun Nayeem
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Leonard Y M Cheung
- Department of Physiology & Biophysics, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
15
|
Cheetham T, Wood C. Paediatric thyroid disease. Clin Endocrinol (Oxf) 2024. [PMID: 39072866 DOI: 10.1111/cen.15110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/30/2024]
Abstract
The spectrum of thyroid disorders presenting to paediatricians is different to that seen by adult physicians. Referrals reflect cases detected by the neonatal screening programme for congenital hypothyroidism and many of the inherited defects of thyroid hormone generation or action will be manifest in early life. Autoimmune thyroid disease can be particularly challenging to manage in the young and the potential impact of thyroid status on neurodevelopment and schooling are key considerations throughout childhood and adolescence.
Collapse
Affiliation(s)
- Timothy Cheetham
- Newcastle University and Great North Children's Hospital, Newcastle upon Tyne, UK
| | - Claire Wood
- Newcastle University and Great North Children's Hospital, Newcastle upon Tyne, UK
| |
Collapse
|
16
|
Bauer AJ, Auble B, Clark AL, Hu TY, Isaza A, McNerney KP, Metzger DL, Nicol L, Pierce SR, Sidlow R. Unmet patient needs in monocarboxylate transporter 8 (MCT8) deficiency: a review. Front Pediatr 2024; 12:1444919. [PMID: 39132310 PMCID: PMC11310894 DOI: 10.3389/fped.2024.1444919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/03/2024] [Indexed: 08/13/2024] Open
Abstract
Monocarboxylate transporter 8 (MCT8) deficiency is a rare, X-linked disorder arising from mutations in the SLC16A2 gene and resulting from dysfunctional thyroid hormone transport. This disorder is characterized by profound neurodevelopmental delay and motor disability due to a lack of thyroid hormone in the brain, and coexisting endocrinological symptoms, due to chronic thyrotoxicosis, resulting from elevated thyroid hormone outside the central nervous system (CNS). In February 2024, we reviewed the published literature to identify relevant articles reporting on the current unmet needs of patients with MCT8 deficiency. There are several main challenges in the diagnosis and treatment of MCT8 deficiency, with decreased awareness and recognition of MCT8 deficiency among healthcare professionals (HCPs) associated with misdiagnosis and delays in diagnosis. Diagnostic delay may also be attributed to other factors, including the complex symptomology of MCT8 deficiency only becoming apparent several months after birth and pathognomonic serum triiodothyronine (T3) testing not being routinely performed. For patients with MCT8 deficiency, multidisciplinary team care is vital to optimize the support provided to patients and their caregivers. Although there are currently no approved treatments specifically for MCT8 deficiency, earlier identification and diagnosis of this disorder enables earlier access to supportive care and developing treatments focused on improving outcomes and quality of life for both patients and caregivers.
Collapse
Affiliation(s)
- Andrew J. Bauer
- The Thyroid Center, Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Bethany Auble
- Medical College of Wisconsin, Children’s Wisconsin, Milwaukee, WI, United States
| | - Amy L. Clark
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO, United States
- Department of Pediatric Endocrinology and Diabetes, SSM Health Cardinal Glennon, St. Louis, MO, United States
| | - Tina Y. Hu
- Department of Pediatrics, Division of Endocrinology, University of California San Francisco, San Francisco, CA, United States
| | - Amber Isaza
- The Thyroid Center, Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Kyle P. McNerney
- Diabetes Education Program, Washington University in St. Louis, St. Louis, MO, United States
| | - Daniel L. Metzger
- The Endocrinology & Diabetes Unit, British Columbia Children’s Hospital, Vancouver, BC, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Lindsey Nicol
- Department of Pediatric Endocrinology, Oregon Health & Science University Doernbecher Children’s Hospital, Portland, OR, United States
- Division of Endocrinology, Oregon Health & Science University, Portland, OR, United States
| | - Samuel R. Pierce
- Division of Rehabilitation Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Richard Sidlow
- Department of Medical Genetics and Metabolism, Valley Children’s Hospital, Madera, CA, United States
| |
Collapse
|
17
|
Peng W, Shi S, Yang L, Liu D. Identification of a novel nonsense SLC16A2 gene mutation in an infant with severe neurologic phenotype: A case report. Medicine (Baltimore) 2024; 103:e39047. [PMID: 39029020 PMCID: PMC11398793 DOI: 10.1097/md.0000000000039047] [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: 04/17/2024] [Accepted: 07/02/2024] [Indexed: 07/21/2024] Open
Abstract
RATIONALE Allan-Herndon-Dudley syndrome (AHDS) results from a pathogenic variant in the hemizygous subunit of the SLC16A2 gene, which encodes monocarboxylate transporter 8 and follows an X-linked recessive pattern. AHDS manifests as neuropsychomotor developmental delay, intellectual disability, movement disorders, and thyroid hormone abnormalities. It is frequently misdiagnosed as cerebral palsy or hypothyroidism. PATIENT CONCERNS A 9-month-old male infant exhibited poor head control, hypodynamia, motor retardation, hypertonic limbs, and thyroid abnormalities. Despite levothyroxine supplementation and rehabilitation therapy, no improvements were observed. Whole-exome sequencing identified a novel nonsense mutation in SLC16A2 (c.124G > T, p.E42X), which unequivocally established the diagnosis. DIAGNOSES AHDS was confirmed. INTERVENTIONS Levothyroxine treatment commenced early in infancy, followed by 3 months of rehabilitation therapy, starting at 5 months of age. The combined administration of levothyroxine and methimazole was initiated at 1 year and 10 months of age, respectively. OUTCOMES While improvements were noted in thyroid hormone levels, neurological developmental delays persisted. LESSONS AHDS should be considered in patients presenting with atypical neurological features and thyroid hormone abnormalities such as elevated triiodothyronine and decreased thyroxine levels. The early utilization of exome sequencing aids in prompt diagnosis. The identified SLC16A2 nonsense mutation correlates with severe neurological phenotypes and adds to the spectrum of genetic variations associated with AHDS.
Collapse
Affiliation(s)
- Wu Peng
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuxia Shi
- Department of Clinical Medicine, Anhui Medical College, Hefei, China
| | - Liqi Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Deyun Liu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
18
|
Anthofer L, Gmach P, Uretmen Kagiali ZC, Kleinau G, Rotter J, Opitz R, Scheerer P, Beck-Sickinger AG, Wolf P, Biebermann H, Bechmann I, Kühnen P, Krude H, Paisdzior S. Melanocortin-4 Receptor PLC Activation Is Modulated by an Interaction with the Monocarboxylate Transporter 8. Int J Mol Sci 2024; 25:7565. [PMID: 39062808 PMCID: PMC11277258 DOI: 10.3390/ijms25147565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
The melanocortin-4 receptor (MC4R) is a key player in the hypothalamic leptin-melanocortin pathway that regulates satiety and hunger. MC4R belongs to the G protein-coupled receptors (GPCRs), which are known to form heterodimers with other membrane proteins, potentially modulating receptor function or characteristics. Like MC4R, thyroid hormones (TH) are also essential for energy homeostasis control. TH transport across membranes is facilitated by the monocarboxylate transporter 8 (MCT8), which is also known to form heterodimers with GPCRs. Based on the finding in single-cell RNA-sequencing data that both proteins are simultaneously expressed in hypothalamic neurons, we investigated a putative interplay between MC4R and MCT8. We developed a novel staining protocol utilizing a fluorophore-labeled MC4R ligand and demonstrated a co-localization of MC4R and MCT8 in human brain tissue. Using in vitro assays such as BRET, IP1, and cAMP determination, we found that MCT8 modulates MC4R-mediated phospholipase C activation but not cAMP formation via a direct interaction, an effect that does not require a functional MCT8 as it was not altered by a specific MCT8 inhibitor. This suggests an extended functional spectrum of MCT8 as a GPCR signaling modulator and argues for the investigation of further GPCR-protein interactions with hitherto underrepresented physiological functions.
Collapse
Affiliation(s)
- Larissa Anthofer
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- Institute of Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Philipp Gmach
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Zeynep Cansu Uretmen Kagiali
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Gunnar Kleinau
- Group Structural Biology of Cellular Signaling, Institute of Medical Physics and Biophysics, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Jonas Rotter
- Institute of Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Robert Opitz
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Patrick Scheerer
- Group Structural Biology of Cellular Signaling, Institute of Medical Physics and Biophysics, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | | | - Philipp Wolf
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, D-04103 Leipzig, Germany
| | - Heike Biebermann
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Peter Kühnen
- Department for Pediatric Endocrinology and Diabetology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Heiko Krude
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Sarah Paisdzior
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| |
Collapse
|
19
|
Sriram S, Shahid N, Mysliwiec D D, Lichter-Konecki U, Yatsenko SA, Garibaldi LR. Late diagnosis of the X-linked MCT8 deficiency (Allan-Herndon-Dudley syndrome) in a teenage girl with primary ovarian insufficiency. J Pediatr Endocrinol Metab 2024; 37:371-374. [PMID: 38345890 DOI: 10.1515/jpem-2023-0070] [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: 02/16/2023] [Accepted: 11/27/2023] [Indexed: 04/11/2024]
Abstract
OBJECTIVES To report an unusual case of MCT8 deficiency (Allan-Herndon-Dudley syndrome), an X-linked condition caused by pathogenic variants in the SLC16A2 gene. Defective transport of thyroid hormones (THs) in this condition leads to severe neurodevelopmental impairment in males, while heterozygous females are usually asymptomatic or have mild TH abnormalities. CASE PRESENTATION A girl with profound developmental delay, epilepsy, primary amenorrhea, elevated T3, low T4 and free T4 levels was diagnosed with MCT8-deficiency at age 17 years, during evaluation for primary ovarian insufficiency (POI). Cytogenetic analysis demonstrated balanced t(X;16)(q13.2;q12.1) translocation with a breakpoint disrupting SLC16A2. X-chromosome inactivation studies revealed a skewed inactivation of the normal X chromosome. CONCLUSIONS MCT8-deficiency can manifest clinically and phenotypically in women with SLC16A2 aberrations when nonrandom X inactivation occurs, while lack of X chromosome integrity due to translocation can cause POI.
Collapse
Affiliation(s)
- Swetha Sriram
- Division of Pediatric Endocrinology, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nabiha Shahid
- Division of Pediatric Endocrinology, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Diana Mysliwiec D
- Division of Pediatric Endocrinology, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Uta Lichter-Konecki
- Division of Genetics and Inborn Errors of Metabolism, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Svetlana A Yatsenko
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Luigi R Garibaldi
- Division of Pediatric Endocrinology, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Graffunder AS, Bresser AAJ, Fernandez Vallone V, Megges M, Stachelscheid H, Kühnen P, Opitz R. Spatiotemporal expression of thyroid hormone transporter MCT8 and THRA mRNA in human cerebral organoids recapitulating first trimester cortex development. Sci Rep 2024; 14:9355. [PMID: 38654093 PMCID: PMC11039642 DOI: 10.1038/s41598-024-59533-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Thyroid hormones (TH) play critical roles during nervous system development and patients carrying coding variants of MCT8 (monocarboxylate transporter 8) or THRA (thyroid hormone receptor alpha) present a spectrum of neurological phenotypes resulting from perturbed local TH action during early brain development. Recently, human cerebral organoids (hCOs) emerged as powerful in vitro tools for disease modelling recapitulating key aspects of early human cortex development. To begin exploring prospects of this model for thyroid research, we performed a detailed characterization of the spatiotemporal expression of MCT8 and THRA in developing hCOs. Immunostaining showed MCT8 membrane expression in neuronal progenitor cell types including early neuroepithelial cells, radial glia cells (RGCs), intermediate progenitors and outer RGCs. In addition, we detected robust MCT8 protein expression in deep layer and upper layer neurons. Spatiotemporal SLC16A2 mRNA expression, detected by fluorescent in situ hybridization (FISH), was highly concordant with MCT8 protein expression across cortical cell layers. FISH detected THRA mRNA expression already in neuroepithelium before the onset of neurogenesis. THRA mRNA expression remained low in the ventricular zone, increased in the subventricular zone whereas strong THRA expression was observed in excitatory neurons. In combination with a robust up-regulation of known T3 response genes following T3 treatment, these observations show that hCOs provide a promising and experimentally tractable model to probe local TH action during human cortical neurogenesis and eventually to model the consequences of impaired TH function for early cortex development.
Collapse
Affiliation(s)
- Adina Sophie Graffunder
- Department of Pediatric Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Audrey Amber Julie Bresser
- Department of Pediatric Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Valeria Fernandez Vallone
- Core Unit Pluripotent Stem Cells and Organoids (CUSCO), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Megges
- Department of Pediatric Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Harald Stachelscheid
- Core Unit Pluripotent Stem Cells and Organoids (CUSCO), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Kühnen
- Department of Pediatric Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Opitz
- Institute of Experimental Pediatric Endocrinology, Charité Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
21
|
Moran C, Schoenmakers N, Halsall D, Oddy S, Lyons G, van den Berg S, Gurnell M, Chatterjee K. Approach to the Patient With Raised Thyroid Hormones and Nonsuppressed TSH. J Clin Endocrinol Metab 2024; 109:1094-1108. [PMID: 37988295 PMCID: PMC10940260 DOI: 10.1210/clinem/dgad681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/24/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
Measurement of free thyroid hormones (THs) and thyrotropin (TSH) using automated immunoassays is central to the diagnosis of thyroid dysfunction. Using illustrative cases, we describe a diagnostic approach to discordant thyroid function tests, focusing on entities causing elevated free thyroxine and/or free triiodothyronine measurements with nonsuppressed TSH levels. Different types of analytical interference (eg, abnormal thyroid hormone binding proteins, antibodies to iodothyronines or TSH, heterophile antibodies, biotin) or disorders (eg, resistance to thyroid hormone β or α, monocarboxylate transporter 8 or selenoprotein deficiency, TSH-secreting pituitary tumor) that can cause this biochemical pattern will be considered. We show that a structured approach, combining clinical assessment with additional laboratory investigations to exclude assay artifact, followed by genetic testing or specialized imaging, can establish a correct diagnosis, potentially preventing unnecessary investigation or inappropriate therapy.
Collapse
Affiliation(s)
- Carla Moran
- Endocrine Section, Beacon Hospital, Dublin, D18 AK68, Ireland
- Endocrine Department, St. Vincent's University Hospital, Dublin, D04 T6F4, Ireland
- School of Medicine, University College Dublin, Dublin, D04 V1W8, Ireland
| | - Nadia Schoenmakers
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - David Halsall
- Department of Clinical Biochemistry, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Susan Oddy
- Department of Clinical Biochemistry, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Greta Lyons
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Sjoerd van den Berg
- Department of Clinical Chemistry, Erasmus Medical Center, 3015 GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Mark Gurnell
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Krishna Chatterjee
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
22
|
Freund ME, van der Most F, Visser WE. Diagnosis and Therapy in MCT8 Deficiency: Ongoing Challenges. J Clin Res Pediatr Endocrinol 2024; 16:1-3. [PMID: 38345399 PMCID: PMC10938520 DOI: 10.4274/jcrpe.galenos.2024.2024-1-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/16/2024] Open
Affiliation(s)
- Matthijs E.T. Freund
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Floor van der Most
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - W. Edward Visser
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
23
|
Ünsal Y, Hayran G. Impact of Early Intervention with Triiodothyroacetic Acid on Peripheral and Neurodevelopmental Findings in a Boy with MCT8 Deficiency. J Clin Res Pediatr Endocrinol 2024; 16:116-122. [PMID: 38054413 PMCID: PMC10938514 DOI: 10.4274/jcrpe.galenos.2023.2023-10-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/25/2023] [Indexed: 12/07/2023] Open
Abstract
Monocarboxylate transporter 8 (MCT8) deficiency is a rare genetic disorder characterized by peripheral thyrotoxicosis and severe cognitive and motor disability due to cerebral hypothyroidism. 3,3’,5-triiodothyroacetic acid (Triac) was shown to improve peripheral thyrotoxicosis but data on neurodevelopmental outcome are scarce. We present a case of MCT8 deficiency and the experience with Triac focusing on change in neurodevelopmental and peripheral features. A five-month-old boy was referred because of feeding difficulty, central hypotonia and global developmental delay. Despite six months of physiotherapy, physical developmental milestones did not improve, and distal muscle tone was increased. A hemizygous pathogenic variant in SLC16A2 was found and MCT8 deficiency was confirmed at 19-months. Thyroid stimulating hormone was 2.83 mIU/mL, free thyroxine 6.24 pmol/L (N=12-22) and free triiodothyronine (FT3) 15.65pmol/L (N=3.1-6.8). He had tachycardia, blood pressure and transaminases were elevated. Triac was started at 21-months. Two weeks after treatment, FT3 dramatically decreased, steady normal serum FT3 was achieved at 28-months. Assessment of neurodevelopmental milestones and signs of hyperthyroidism were evaluated at baseline, 6 months and 12 months after treatment. Signs of hyperthyroidism were improved by 6 months. Developmental composite scores of Bayley Scales of Infant Developmental 3rd Edition remained the same but important developmental milestones (head control, recognition of caregiver, response to his name) were attained, regression in the attained milestones were not observed. Initial dose, management protocol for Triac and research into its efficacy on neurodevelopmental signs in MCT8 deficiency are progressing. This case presents evidence that Triac may resolve peripheral thyrotoxicosis successfully and may slow neurodevelopmental regression, while some developmental milestones were achieved after one year of treatment.
Collapse
Affiliation(s)
- Yağmur Ünsal
- Şanlıurfa Training and Research Hospital, Clinic of Pediatric Endocrinology, Şanlıurfa, Turkey
| | - Gamze Hayran
- Şanlıurfa Training and Research Hospital, Clinic of Developmental Pediatrics, Şanlıurfa, Turkey
| |
Collapse
|
24
|
van Geest FS, Groeneweg S, Popa VM, Stals MAM, Visser WE. Parent Perspectives on Complex Needs in Patients With MCT8 Deficiency: An International, Prospective, Registry Study. J Clin Endocrinol Metab 2023; 109:e330-e335. [PMID: 37450560 PMCID: PMC10735299 DOI: 10.1210/clinem/dgad412] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/22/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
CONTEXT Monocarboxylate transporter 8 (MCT8) deficiency is a rare neurodevelopmental and metabolic disorder, with daily care posing a heavy burden on caregivers. A comprehensive overview of these complex needs and daily care challenges is lacking. DESIGN We established an international prospective registry to systemically capture data from parents and physicians caring for patients with MCT8 deficiency. Parent-reported data on complex needs and daily care challenges were extracted. RESULTS Between July 17, 2018, and May 16, 2022, 51 patients were registered. Difficulties in daily life care were mostly related to feeding and nutritional status (17/33 patients), limited motor skills (12/33 patients), and sleeping (11/33 patients). Dietary advice was provided for 11/36 patients. Two of 32 patients were under care of a cardiologist. Common difficulties in the diagnostic trajectory included late diagnosis (20/35 patients) and visiting a multitude of specialists (15/35 patients). Median diagnostic delay was significantly shorter in patients born in or after 2017 vs before 2017 (8 vs 19 months, P < .0001). CONCLUSIONS Feeding and sleeping problems and limited motor skills mostly contribute to difficulties in daily care. The majority of patients did not receive professional dietary advice, although being underweight is a key disease feature, strongly linked with poor survival. Despite sudden death being a prominent cause of death, potentially related to the cardiovascular abnormalities frequently observed, patients were hardly seen by cardiologists. These findings can directly improve patient-centered multidisciplinary care and define patient-centered outcome measures for intervention studies in patients with MCT8 deficiency.
Collapse
Affiliation(s)
- Ferdy S van Geest
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Stefan Groeneweg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Veronica M Popa
- Patient Advisory Council of RD Connect and MCT8-AHDS Foundation, Oklahoma, OK 74464, USA
| | - Milou A M Stals
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| | - W Edward Visser
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, 3015 CN Rotterdam, The Netherlands
| |
Collapse
|
25
|
Yiu RSW, Ling TK, Ko CH, Poon SWY, Poon GWK, Wong FCK, Law CY, Iwayama H, Lam CW. Allan-Herndon-Dudley syndrome in Hong Kong: Implication for newborn screening. Clin Chim Acta 2023; 551:117621. [PMID: 37925810 DOI: 10.1016/j.cca.2023.117621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/21/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Allan-Herndon-Dudley syndrome (MCT 8 deficiency) is an X-linked recessive condition caused by hemizygous pathogenic variants in SLC16A2 encoding the monocarboxylate transporter 8 (MCT8). Patients present with global developmental delay and neurological impairment, and abnormal serum thyroid function tests. The drug, 3,3',5 triiodothyroacetic acid (TRIAC), was recently demonstrated to improve the endocrinological profile. Improvement in diagnostic approach is key to earlier start of treatment. PATIENT FINDINGS We described four Chinese patients with MCT8 deficiency undergoing different diagnostic odysseys. Their initial presentation included global developmental delay and dystonia. Patient 2 also had epilepsy. Patients 1 and 2 presented with two novel variants: (1)hemizygous NM_006517.4(SLC16A2):c.1170 + 2 T > A; p.(?), and (2)hemizygous NM_006517.4(SLC16A2):c.305dupT; p.(Val103GlyfsTer17) respectively. Patients 3 and 4 were biological brothers harboring hemizygous NM_006517.4(SLC16A2):c.305dupT; p.(Val103GlyfsTer17), which was first reported in 2004. We obtained the measurement of triiodothyronine (T3) and reverse T3 (rT3) from dried blood spot samples collected on Day 1 of life from Patient 1 and studied the biomarkers (rT3 and T3/rT3 ratio) proposed by Iwayama et al. for the detection of MCT8 deficiency at birth. Our data verified the significantly reduced rT3 level in Patient 1, compared with healthy newborns, although low T3 level and comparable T3/rT3 ratio with controls were detected. SUMMARY Patients with MCT8 deficiency often undergo diagnostic odysseys. An early diagnosis could be missed by a normal newborn thyroid function screening result based on biochemical measurement of TSH and/or T4/fT4. Early detection of rT3 is key to improving current diagnostic approach. CONCLUSION We recommend that full thyroid function profile (TSH, T4/fT4, T3/fT3, rT3) be considered early for all pediatric patients presenting with unexplained developmental delay and/or dystonia. The potential inclusion of rT3 measurement in newborn screening may prove promising.
Collapse
Affiliation(s)
- Rachel Sze-Wan Yiu
- Division of Chemical Pathology, Department of Pathology, Queen Mary Hospital, Hong Kong, China
| | - Tsz-Ki Ling
- Division of Chemical Pathology, Department of Pathology, Queen Mary Hospital, Hong Kong, China
| | - Chun-Hung Ko
- Department of Paediatrics and Adolescent Medicine, Caritas Medical Centre, Hong Kong, China
| | - Sarah Wing-Yiu Poon
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong, China
| | - Grace Wing-Kit Poon
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong, China
| | - Felix Chi-Kin Wong
- Division of Chemical Pathology, Department of Pathology, Queen Mary Hospital, Hong Kong, China
| | - Chun-Yiu Law
- Division of Chemical Pathology, Department of Pathology, Queen Mary Hospital, Hong Kong, China
| | - Hideyuki Iwayama
- Department of Pediatrics, Aichi Medical University, Nagakute, Japan
| | - Ching-Wan Lam
- Division of Chemical Pathology, Department of Pathology, Queen Mary Hospital, Hong Kong, China; Department of Pathology, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
26
|
Wilpert NM, Tonduti D, Vaia Y, Krude H, Sarret C, Schuelke M. Establishing Patient-Centered Outcomes for MCT8 Deficiency: Stakeholder Engagement and Systematic Literature Review. Neuropsychiatr Dis Treat 2023; 19:2195-2216. [PMID: 37881807 PMCID: PMC10595182 DOI: 10.2147/ndt.s379703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/07/2023] [Indexed: 10/27/2023] Open
Abstract
Introduction The SCL16A2 gene encodes the thyroid hormone (TH) transporter MCT8. Pathogenic variants result in a reduced TH uptake into the CNS despite high serum T3 concentrations. Patients suffer from severe neurodevelopmental delay and require multidisciplinary care. Since a first compassionate use study in 2008, the development of therapies has recently gained momentum. Treatment strategies range from symptom-based approaches, supplementation with TH or TH-analogs, to gene therapy. All these studies have mainly used surrogate endpoints and clinical outcomes. However, the EMA and FDA strongly encourage researchers to involve patients and their advocacy groups in the design of clinical trials. This should strengthen the patients' perspective and identify clinical endpoints that are clinically relevant to their daily life. Methods We involved patient families to define patient-relevant outcomes for MCT8 deficiency. In close collaboration with patient families, we designed a questionnaire asking for their five most preferred therapeutic goals, which, if achieved at least, make a difference in their lives. In addition, we performed a systematic review according to Cochrane recommendations of the published treatment trials. Results We obtained results from 15 families with completed questionnaires from 14 mothers and 8 fathers. Improvement in development, especially in gross motor skills, was most important to the parents. 59% wished for head control and 50% for sitting ability. Another 36% wished for weight gain, 32% for improvement of expressive language skills, and 18% for a reduction of dystonia/spasticity, less dysphagia, and reflux. Paraclinical aspects were least important (5-9%). In a treatment trial (n=46) and compassionate use cases (n=83), the results were mainly inconclusive, partly due to a lack of predefined patient-centered clinical endpoints. Discussion We recommend that future trials should define a relevant improvement in "development" and/or other patient-relevant outcomes compared to natural history as treatment goals.
Collapse
Affiliation(s)
- Nina-Maria Wilpert
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Pediatric Neurology, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Center for Chronically Sick Children, Berlin, Germany
| | - Davide Tonduti
- Unit of Pediatric Neurology, C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children’s Hospital, Università Degli Studi Di Milano, Milan, Italy
| | - Ylenia Vaia
- Unit of Pediatric Neurology, C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children’s Hospital, Università Degli Studi Di Milano, Milan, Italy
| | - Heiko Krude
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Experimental Pediatric Endocrinology, Berlin, Germany
| | - Catherine Sarret
- Centre de Compétence des Leucodystrophies et Leucoencéphalopathies de Cause Rare, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Markus Schuelke
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Pediatric Neurology, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Center for Chronically Sick Children, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), NeuroCure Clinical Research Center, Berlin, Germany
| |
Collapse
|
27
|
Richard S, Ren J, Flamant F. Thyroid hormone action during GABAergic neuron maturation: The quest for mechanisms. Front Endocrinol (Lausanne) 2023; 14:1256877. [PMID: 37854197 PMCID: PMC10579935 DOI: 10.3389/fendo.2023.1256877] [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: 07/11/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Thyroid hormone (TH) signaling plays a major role in mammalian brain development. Data obtained in the past years in animal models have pinpointed GABAergic neurons as a major target of TH signaling during development, which opens up new perspectives to further investigate the mechanisms by which TH affects brain development. The aim of the present review is to gather the available information about the involvement of TH in the maturation of GABAergic neurons. After giving an overview of the kinds of neurological disorders that may arise from disruption of TH signaling during brain development in humans, we will take a historical perspective to show how rodent models of hypothyroidism have gradually pointed to GABAergic neurons as a main target of TH signaling during brain development. The third part of this review underscores the challenges that are encountered when conducting gene expression studies to investigate the molecular mechanisms that are at play downstream of TH receptors during brain development. Unravelling the mechanisms of action of TH in the developing brain should help make progress in the prevention and treatment of several neurological disorders, including autism and epilepsy.
Collapse
Affiliation(s)
| | | | - Frédéric Flamant
- Institut de Génomique Fonctionnelle de Lyon, UMR5242, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard-Lyon 1, USC1370 Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Lyon, France
| |
Collapse
|
28
|
Lv F, Cai X, Li Y, Zhang X, Zhou X, Han X, Ji L. Sensitivity to thyroid hormone and risk of components of metabolic syndrome in a Chinese euthyroid population. J Diabetes 2023; 15:900-910. [PMID: 37429739 PMCID: PMC10590679 DOI: 10.1111/1753-0407.13441] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/29/2023] [Accepted: 06/17/2023] [Indexed: 07/12/2023] Open
Abstract
INTRODUCTION To evaluate the association of sensitivity to thyroid hormone with metabolic syndrome (MetS) and its components in a Chinese euthyroid population. METHODS A total of 3573 participants from Pinggu Metabolic Disease Study were analyzed. Serum-free triiodothyronine (FT3), free thyroxine (FT4), thyrotropin (TSH), total adipose tissue (TAT), visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT) area of abdominal, and lumbar skeletal muscle area (SMA) were measured. Central thyroid hormone resistance was calculated by the Thyroid Feedback Quantile-based Index (TFQI) and Chinese-referenced Parametric TFQI (PTFQI), Thyrotroph T4 Resistance Index (TT4RI) and TSH Index (TSHI). Peripheral thyroid hormone resistance was assessed by FT3/FT4 ratio. RESULTS Higher values of TSHI (odds ratio [OR] = 1.167, 95% confidence interval [CI]: 1.079-1.262, p < .001), TT4RI (OR = 1.115, 95% CI: 1.031-1.206, p = .006), TFQI (OR = 1.196, 95% CI: 1.106-1.294, p < .001), PTFQI (OR = 1.194, 95% CI: 1.104-1.292, p < .001), and lower values of FT3/FT4 ratio (OR = 0.914, 95% CI: 0.845-0.990, p = .026) were associated with MetS. Increased levels of TFQI and PTFQI were associated with abdominal obesity, hypertriglyceridemia, and hypertension. Increased levels of TSHI and TT4RI were associated with hypertriglyceridemia, abdominal obesity, low high-density lipoprotein cholesterol. Reduced levels of FT3/FT4 ratio were associated with hyperglycemia, hypertension, and hypertriglyceridemia. The levels of TSHI, TFQI, and PTFQI were negatively related to SMA and positively related to VAT, SAT, and TAT (all p < .05). CONCLUSIONS Reduced thyroid hormone sensitivity was associated with MetS and its components. Impaired thyroid hormone sensitivity might affect the distribution of adipose tissue and muscle.
Collapse
Affiliation(s)
- Fang Lv
- Department of Endocrinology and MetabolismPeking University People's HospitalBeijingChina
| | - Xiaoling Cai
- Department of Endocrinology and MetabolismPeking University People's HospitalBeijingChina
| | - Yufeng Li
- Department of EndocrinologyBeijing Pinggu HospitalBeijingChina
| | - Xiuying Zhang
- Department of Endocrinology and MetabolismPeking University People's HospitalBeijingChina
| | - Xianghai Zhou
- Department of Endocrinology and MetabolismPeking University People's HospitalBeijingChina
| | - Xueyao Han
- Department of Endocrinology and MetabolismPeking University People's HospitalBeijingChina
| | - Linong Ji
- Department of Endocrinology and MetabolismPeking University People's HospitalBeijingChina
| |
Collapse
|
29
|
Thomas J, Sairoz, Jose A, Poojari VG, Shetty S, K SP, Prabhu R V K, Rao M. Role and Clinical Significance of Monocarboxylate Transporter 8 (MCT8) During Pregnancy. Reprod Sci 2023; 30:1758-1769. [PMID: 36595209 PMCID: PMC10229697 DOI: 10.1007/s43032-022-01162-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023]
Abstract
The review aims to summarize the available research focusing on the importance of monocarboxylate transporter (MCT8) in thyroid hormone trafficking across the placenta and fetal development. A systematic search was carried out in PubMed; studies available in English related to "monocarboxylate transporter", "adverse pregnancy", "fetal development," and "thyroid hormone" were identified and assessed. The references within the resulting articles were manually searched. MCT8 is a highly active and selective thyroid hormone transporter that facilitates the cellular uptake of triiodothyronine (T3), thyroxine (T4), reverse triiodothyronine (rT3), and diiodothyronine (T2) in different tissues. MCT8 is expressed in the placenta from the first trimester onwards, allowing the transport of thyroid hormone from mother to fetus. Mutations in MCT8 cause an X-linked disorder known as Allan-Herndon-Dudley syndrome (AHDS), characterized by severe psychomotor impairment and peripheral thyrotoxicosis. Hence, any maternal thyroid dysfunction may cause severe consequences for the fetus and newborn. Further research regarding MCT8 gene expression, polymorphic variation, and adverse pregnancy outcomes must be done to establish that MCT8 is a novel prognostic marker for the early detection of pregnancy-related complications.
Collapse
Affiliation(s)
- Jinsu Thomas
- Department of Pharmacy Practice, Center for Translational Research, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sairoz
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Anmi Jose
- Department of Pharmacy Practice, Center for Translational Research, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vidyashree G Poojari
- Department of Reproductive Medicine and Surgery, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sahana Shetty
- Department of Endocrinology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Shama Prasada K
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Krishnananda Prabhu R V
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Center for Translational Research, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
30
|
Wang T, Wang Y, Montero-Pedrazuela A, Prensa L, Guadaño-Ferraz A, Rausell E. Thyroid Hormone Transporters MCT8 and OATP1C1 Are Expressed in Projection Neurons and Interneurons of Basal Ganglia and Motor Thalamus in the Adult Human and Macaque Brains. Int J Mol Sci 2023; 24:9643. [PMID: 37298594 PMCID: PMC10254002 DOI: 10.3390/ijms24119643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Monocarboxylate transporter 8 (MCT8) and organic anion-transporting polypeptide 1C1 (OATP1C1) are thyroid hormone (TH) transmembrane transporters relevant for the availability of TH in neural cells, crucial for their proper development and function. Mutations in MCT8 or OATP1C1 result in severe disorders with dramatic movement disability related to alterations in basal ganglia motor circuits. Mapping the expression of MCT8/OATP1C1 in those circuits is necessary to explain their involvement in motor control. We studied the distribution of both transporters in the neuronal subpopulations that configure the direct and indirect basal ganglia motor circuits using immunohistochemistry and double/multiple labeling immunofluorescence for TH transporters and neuronal biomarkers. We found their expression in the medium-sized spiny neurons of the striatum (the receptor neurons of the corticostriatal pathway) and in various types of its local microcircuitry interneurons, including the cholinergic. We also demonstrate the presence of both transporters in projection neurons of intrinsic and output nuclei of the basal ganglia, motor thalamus and nucleus basalis of Meynert, suggesting an important role of MCT8/OATP1C1 for modulating the motor system. Our findings suggest that a lack of function of these transporters in the basal ganglia circuits would significantly impact motor system modulation, leading to clinically severe movement impairment.
Collapse
Affiliation(s)
- Ting Wang
- School of Medicine, Department Anatomy Histology & Neuroscience, Autónoma de Madrid University (UAM), 28029 Madrid, Spain; (T.W.); (Y.W.); (L.P.)
- PhD Program in Neuroscience, Autónoma de Madrid University (UAM)-Cajal Institute, 28029 Madrid, Spain
| | - Yu Wang
- School of Medicine, Department Anatomy Histology & Neuroscience, Autónoma de Madrid University (UAM), 28029 Madrid, Spain; (T.W.); (Y.W.); (L.P.)
- PhD Program in Neuroscience, Autónoma de Madrid University (UAM)-Cajal Institute, 28029 Madrid, Spain
| | - Ana Montero-Pedrazuela
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Autónoma de Madrid University (UAM), 28029 Madrid, Spain;
| | - Lucía Prensa
- School of Medicine, Department Anatomy Histology & Neuroscience, Autónoma de Madrid University (UAM), 28029 Madrid, Spain; (T.W.); (Y.W.); (L.P.)
| | - Ana Guadaño-Ferraz
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Autónoma de Madrid University (UAM), 28029 Madrid, Spain;
| | - Estrella Rausell
- School of Medicine, Department Anatomy Histology & Neuroscience, Autónoma de Madrid University (UAM), 28029 Madrid, Spain; (T.W.); (Y.W.); (L.P.)
| |
Collapse
|
31
|
Silva N, Campinho MA. In a zebrafish biomedical model of human Allan-Herndon-Dudley syndrome impaired MTH signaling leads to decreased neural cell diversity. Front Endocrinol (Lausanne) 2023; 14:1157685. [PMID: 37214246 PMCID: PMC10194031 DOI: 10.3389/fendo.2023.1157685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/04/2023] [Indexed: 05/24/2023] Open
Abstract
Background Maternally derived thyroid hormone (T3) is a fundamental factor for vertebrate neurodevelopment. In humans, mutations on the thyroid hormones (TH) exclusive transporter monocarboxylic acid transporter 8 (MCT8) lead to the Allan-Herndon-Dudley syndrome (AHDS). Patients with AHDS present severe underdevelopment of the central nervous system, with profound cognitive and locomotor consequences. Functional impairment of zebrafish T3 exclusive membrane transporter Mct8 phenocopies many symptoms observed in patients with AHDS, thus providing an outstanding animal model to study this human condition. In addition, it was previously shown in the zebrafish mct8 KD model that maternal T3 (MTH) acts as an integrator of different key developmental pathways during zebrafish development. Methods Using a zebrafish Mct8 knockdown model, with consequent inhibition of maternal thyroid hormones (MTH) uptake to the target cells, we analyzed genes modulated by MTH by qPCR in a temporal series from the start of segmentation through hatching. Survival (TUNEL) and proliferation (PH3) of neural progenitor cells (dla, her2) were determined, and the cellular distribution of neural MTH-target genes in the spinal cord during development was characterized. In addition, in-vivo live imaging was performed to access NOTCH overexpression action on cell division in this AHDS model. We determined the developmental time window when MTH is required for appropriate CNS development in the zebrafish; MTH is not involved in neuroectoderm specification but is fundamental in the early stages of neurogenesis by promoting the maintenance of specific neural progenitor populations. MTH signaling is required for developing different neural cell types and maintaining spinal cord cytoarchitecture, and modulation of NOTCH signaling in a non-autonomous cell manner is involved in this process. Discussion The findings show that MTH allows the enrichment of neural progenitor pools, regulating the cell diversity output observed by the end of embryogenesis and that Mct8 impairment restricts CNS development. This work contributes to the understanding of the cellular mechanisms underlying human AHDS.
Collapse
Affiliation(s)
- Nádia Silva
- Centre for Marine Sciences of the University of the Algarve, Faro, Portugal
- Algarve Biomedical Center-Research Institute, University of the Algarve, Faro, Portugal
| | - Marco António Campinho
- Centre for Marine Sciences of the University of the Algarve, Faro, Portugal
- Algarve Biomedical Center-Research Institute, University of the Algarve, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of the Algarve, Faro, Portugal
| |
Collapse
|
32
|
Gevers EF, de Winter JP. New developments and therapies in pediatric endocrinology. Eur J Pediatr 2023; 182:1439-1443. [PMID: 36567374 DOI: 10.1007/s00431-022-04772-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Evelien F Gevers
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
- Department of Paediatric Endocrinology and Diabetes, Barts Health NHS Trust - The Royal London Children's Hospital, London, United Kingdom.
| | - J Peter de Winter
- Department of Pediatrics, Spaarne Gasthuis, Haarlem/Hoofddorp, The Netherlands
- Leuven Child and Health Institute, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Triac Treatment Prevents Neurodevelopmental and Locomotor Impairments in Thyroid Hormone Transporter Mct8/Oatp1c1 Deficient Mice. Int J Mol Sci 2023; 24:ijms24043452. [PMID: 36834863 PMCID: PMC9966820 DOI: 10.3390/ijms24043452] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Patients with inactive thyroid hormone (TH) transporter MCT8 display intellectual disability due to compromised central TH transport and action. As a therapeutic strategy, application of thyromimetic, MCT8-independent compounds Triac (3,5,3'-triiodothyroacetic acid), and Ditpa (3,5-diiodo-thyropropionic acid) was proposed. Here, we directly compared their thyromimetic potential in Mct8/Oatp1c1 double knock-out mice (Dko) modeling human MCT8 deficiency. Dko mice received either Triac (50 ng/g or 400 ng/g) or Ditpa (400 ng/g or 4000 ng/g) daily during the first three postnatal weeks. Saline-injected Wt and Dko mice served as controls. A second cohort of Dko mice received Triac (400 ng/g) daily between postnatal weeks 3 and 6. Thyromimetic effects were assessed at different postnatal stages by immunofluorescence, ISH, qPCR, electrophysiological recordings, and behavior tests. Triac treatment (400 ng/g) induced normalized myelination, cortical GABAergic interneuron differentiation, electrophysiological parameters, and locomotor performance only when administered during the first three postnatal weeks. Ditpa (4000 ng/g) application to Dko mice during the first three postnatal weeks resulted in normal myelination and cerebellar development but only mildly improved neuronal parameters and locomotor function. Together, Triac is highly-effective and more efficient than Ditpa in promoting CNS maturation and function in Dko mice yet needs to be initiated directly after birth for the most beneficial effects.
Collapse
|
34
|
Reinwald JR, Weber-Fahr W, Cosa-Linan A, Becker R, Sack M, Falfan-Melgoza C, Gass N, Braun U, Clemm von Hohenberg C, Chen J, Mayerl S, Muente TF, Heuer H, Sartorius A. TRIAC Treatment Improves Impaired Brain Network Function and White Matter Loss in Thyroid Hormone Transporter Mct8/Oatp1c1 Deficient Mice. Int J Mol Sci 2022; 23:15547. [PMID: 36555189 PMCID: PMC9779161 DOI: 10.3390/ijms232415547] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Dysfunctions of the thyroid hormone (TH) transporting monocarboxylate transporter MCT8 lead to a complex X-linked syndrome with abnormal serum TH concentrations and prominent neuropsychiatric symptoms (Allan-Herndon-Dudley syndrome, AHDS). The key features of AHDS are replicated in double knockout mice lacking MCT8 and organic anion transporting protein OATP1C1 (Mct8/Oatp1c1 DKO). In this study, we characterize impairments of brain structure and function in Mct8/Oatp1c1 DKO mice using multimodal magnetic resonance imaging (MRI) and assess the potential of the TH analogue 3,3',5-triiodothyroacetic acid (TRIAC) to rescue this phenotype. Structural and functional MRI were performed in 11-weeks-old male Mct8/Oatp1c1 DKO mice (N = 10), wild type controls (N = 7) and Mct8/Oatp1c1 DKO mice (N = 13) that were injected with TRIAC (400 ng/g bw s.c.) daily during the first three postnatal weeks. Grey and white matter volume were broadly reduced in Mct8/Oatp1c1 DKO mice. TRIAC treatment could significantly improve white matter thinning but did not affect grey matter loss. Network-based statistic showed a wide-spread increase of functional connectivity, while graph analysis revealed an impairment of small-worldness and whole-brain segregation in Mct8/Oatp1c1 DKO mice. Both functional deficits could be substantially ameliorated by TRIAC treatment. Our study demonstrates prominent structural and functional brain alterations in Mct8/Oatp1c1 DKO mice that may underlie the psychomotor deficiencies in AHDS. Additionally, we provide preclinical evidence that early-life TRIAC treatment improves white matter loss and brain network dysfunctions associated with TH transporter deficiency.
Collapse
Affiliation(s)
- Jonathan Rochus Reinwald
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
- Research Group Systems Neuroscience and Mental Health, Department of Psychiatry and Psychotherapy, University Medical Center Mainz, 55131 Mainz, Germany
| | - Wolfgang Weber-Fahr
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Alejandro Cosa-Linan
- Research Group in Silico Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Robert Becker
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
- Center for Innovative Psychiatry and Psychotherapy Research, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Markus Sack
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
- Center for Innovative Psychiatry and Psychotherapy Research, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Claudia Falfan-Melgoza
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Natalia Gass
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Urs Braun
- Research Group Systems Neuroscience in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Christian Clemm von Hohenberg
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Jiesi Chen
- Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Steffen Mayerl
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Thomas F. Muente
- Department of Neurology, University of Lübeck, 23538 Lübeck, Germany
| | - Heike Heuer
- Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Alexander Sartorius
- Research Group Translational Imaging, Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| |
Collapse
|
35
|
Thyroid Hormone Transporters in Pregnancy and Fetal Development. Int J Mol Sci 2022; 23:ijms232315113. [PMID: 36499435 PMCID: PMC9737226 DOI: 10.3390/ijms232315113] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Thyroid hormone is essential for fetal (brain) development. Plasma membrane transporters control the intracellular bioavailability of thyroid hormone. In the past few decades, 15 human thyroid hormone transporters have been identified, and among them, mutations in monocarboxylate transporter (MCT)8 and organic anion transporting peptide (OATP)1C1 are associated with clinical phenotypes. Different animal and human models have been employed to unravel the (patho)-physiological role of thyroid hormone transporters. However, most studies on thyroid hormone transporters focus on postnatal development. This review summarizes the research on the thyroid hormone transporters in pregnancy and fetal development, including their substrate preference, expression and tissue distribution, and physiological and pathophysiological role in thyroid homeostasis and clinical disorders. As the fetus depends on the maternal thyroid hormone supply, especially during the first half of pregnancy, the review also elaborates on thyroid hormone transport across the human placental barrier. Future studies may reveal how the different transporters contribute to thyroid hormone homeostasis in fetal tissues to properly facilitate development. Employing state-of-the-art human models will enable a better understanding of their roles in thyroid hormone homeostasis.
Collapse
|
36
|
Wasserman-Bartov T, Admati I, Lebenthal-Loinger I, Sharabany J, Lerer-Goldshtein T, Appelbaum L. Tsh Induces Agrp1 Neuron Proliferation in Oatp1c1-Deficient Zebrafish. J Neurosci 2022; 42:8214-8224. [PMID: 36150888 PMCID: PMC9653277 DOI: 10.1523/jneurosci.0002-22.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/25/2022] [Accepted: 08/30/2022] [Indexed: 11/21/2022] Open
Abstract
Thyroid hormones (THs), thyroxine (T4), and triiodothyronine (T3), regulate growth, metabolism, and neurodevelopment. THs secretion is controlled by the pituitary thyroid-stimulating hormone (TSH) and the hypothalamic-pituitary-thyroid (HPT) axis. The organic anion-transporting polypeptide 1C1 (OATP1C1/SLCO1C1) and the monocarboxylate transporter 8 (MCT8/SLC16A2) actively transport THs, which bind to their nuclear receptors and induce gene expression. A mutation in OATP1C1 is associated with brain hypometabolism, gradual neurodegeneration, and impaired cognitive and motor functioning in adolescent patients. To understand the role of Oatp1c1 and the mechanisms of the disease, we profiled the transcriptome of oatp1c1 mutant (oatp1c1 -/-) and mct8 -/- xoatp1c1 -/- adult male and female zebrafish brains. Among dozens of differentially expressed genes, agouti-related neuropeptide 1 (agrp1) expression increased in oatp1c1 -/- adult brains. Imaging in the hypothalamus revealed enhanced proliferation of Agrp1 neurons in oatp1c1 -/- larvae and adults, and increased food consumption in oatp1c1 -/- larvae. Similarly, feeding and the number of Agrp1 neurons increased in thyroid gland-ablated zebrafish. Pharmacological treatments showed that the T3 analog TRIAC (3,3',5-tri-iodothyroacetic acid), but not T4, normalized the number of Agrp1 neurons in oatp1c1 -/- zebrafish. Since the HPT axis is hyperactive in the oatp1c1 -/- brain, we used the CRISPR-Cas9 system to knockdown tsh in oatp1c1 -/- larvae, and inducibly enhanced the HPT axis in wild-type larvae. These manipulations showed that Tsh promotes proliferation of Agrp1 neurons and increases food consumption in zebrafish. The results revealed upregulation of both the HPT axis-Agrp1 circuitry and feeding in a zebrafish model for OATP1C1 deficiency.SIGNIFICANCE STATEMENT Mutation in the thyroid hormone (TH) transporter OATP1C1 is associated with cognitive and motor functioning disturbances in humans. Here, we used an oatp1c1 -/- zebrafish to understand the role of organic anion-transporting polypeptide 1C1 (Oatp1c1), and the characteristics of OATP1C1 deficiency. Transcriptome profiling identified upregulation of agrp1 expression in the oatp1c1 -/- brain. The oatp1c1 -/- larvae showed increased thyroid-stimulating hormone (tsh) levels, proliferation of Agrp1 neurons and food consumption. Genetic manipulations of the hypothalamic-pituitary-thyroid (HPT) axis showed that Tsh increases the number of Agrp1 neurons and food consumption. The T3 analog TRIAC (3,3',5-tri-iodothyroacetic acid) normalizes the number of Agrp1 neurons and may have potential for the treatment of Oatp1c1 deficiency. The findings demonstrate a functional interaction between the thyroid and feeding systems in the brain of zebrafish and suggest a neuroendocrinological mechanism for OATP1C1 deficiency.
Collapse
Affiliation(s)
- Talya Wasserman-Bartov
- The Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
- The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Inbal Admati
- The Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
- The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | | | - Julia Sharabany
- The Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
- The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Tali Lerer-Goldshtein
- The Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
- The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Lior Appelbaum
- The Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
- The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| |
Collapse
|
37
|
Kubota M, Yakuwa A, Terashima H, Hoshino H. A nationwide survey of monocarboxylate transporter 8 deficiency in Japan: Its incidence, clinical course, MRI and laboratory findings. Brain Dev 2022; 44:699-705. [PMID: 35945102 DOI: 10.1016/j.braindev.2022.07.007] [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: 02/25/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Monocarboxylate transporter 8 (MCT8) deficiency is an X-linked recessive developmental disorder characterized by initially marked truncal hypotonia, later athetotic posturing, and severe intellectual disability caused by mutations in SLC16A2, which is responsible for the transport of triiodothyronine (T3) into neurons. We conducted a nationwide survey of patients with MCT8 deficiency to clarify their current status. METHODS Primary survey: In 2016-2017, we assessed the number of patients diagnosed with MCT8 deficiency from 1027 hospitals. Secondary survey: in 2017-2018, we sent case surveys to 31 hospitals (45 cases of genetic diagnosis), who responded in the primary survey. We asked for: 1) perinatal history, 2) developmental history, 3) head MRI findings, 4) neurophysiological findings, 5) thyroid function tests, and 5) genetic test findings. RESULTS We estimated the prevalence of MCT8 deficiency to be 1 in 1,890,000 and the incidence of MCT8 deficiency per million births to be 2.12 (95 % CI: 0.99-3.25). All patients showed severe psychomotor retardation, and none were able to walk or speak. The significantly higher value of the free T3/free T4 (fT3/fT4) ratio found in our study can be a simple and useful diagnostic biomarker (Our value 11.60 ± 4.14 vs control 3.03 ± 0.38). Initial white matter signal abnormalities on head MRI showed recovery, but somatosensory evoked potentials (SEP) showed no improvement, suggesting that the patient remained dysfunctional. CONCLUSION For early diagnosis, including in mild cases, it might be important to consider the clinical course, early head MRI, SEP, and fT3/fT4 ratio.
Collapse
Affiliation(s)
- Masaya Kubota
- Division of Neurology, National Center for Child Health and Development, Japan; Department of Pediatrics, Shimada Ryoiku Medical Center for Challenged Children, Japan.
| | - Akiko Yakuwa
- Department of Pediatrics, National Rehabilitation Center for Children with Disabilities, Japan.
| | - Hiroshi Terashima
- Division of Neurology, National Center for Child Health and Development, Japan.
| | | |
Collapse
|
38
|
Valcárcel-Hernández V, Guillén-Yunta M, Bueno-Arribas M, Montero-Pedrazuela A, Grijota-Martínez C, Markossian S, García-Aldea Á, Flamant F, Bárez-López S, Guadaño-Ferraz A. A CRISPR/Cas9-engineered avatar mouse model of monocarboxylate transporter 8 deficiency displays distinct neurological alterations. Neurobiol Dis 2022; 174:105896. [DOI: 10.1016/j.nbd.2022.105896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/30/2022] [Accepted: 10/11/2022] [Indexed: 10/31/2022] Open
|
39
|
Moran C, Schoenmakers N, Visser WE, Schoenmakers E, Agostini M, Chatterjee K. Genetic disorders of thyroid development, hormone biosynthesis and signalling. Clin Endocrinol (Oxf) 2022; 97:502-514. [PMID: 35999191 PMCID: PMC9544560 DOI: 10.1111/cen.14817] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/24/2022] [Accepted: 07/04/2022] [Indexed: 11/28/2022]
Abstract
Development and differentiation of the thyroid gland is directed by expression of specific transcription factors in the thyroid follicular cell which mediates hormone biosynthesis. Membrane transporters are rate-limiting for cellular entry of thyroid hormones (TH) (T4 and T3) into some tissues, with selenocysteine-containing, deiodinase enzymes (DIO1 and DIO2) converting T4 to the biologically active hormone T3. TH regulate expression of target genes via hormone-inducible nuclear receptors (TRα and TRβ) to exert their physiological effects. Primary congenital hypothyroidism (CH) due to thyroid dysgenesis may be mediated by defects in thyroid transcription factors or impaired thyroid stimulating hormone receptor function. Dyshormonogenic CH is usually due to mutations in genes mediating thyroidal iodide transport, organification or iodotyrosine synthesis and recycling. Disorders of TH signalling encompass conditions due to defects in membrane TH transporters, impaired hormone metabolism due to deficiency of deiodinases and syndromes of Resistance to thyroid hormone due to pathogenic variants in either TRα or TRβ. Here, we review the genetic basis, pathogenesis and clinical features of congenital, dysgenetic or dyshormonogenic hypothyroidism and disorders of TH transport, metabolism and action.
Collapse
Affiliation(s)
- Carla Moran
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- Present address:
Beacon Hospital and School of MedicineUniversity CollegeDublinIreland
| | - Nadia Schoenmakers
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - W. Edward Visser
- Department of Internal MedicineErasmus Medical Center, Academic Center for Thyroid DiseasesRotterdamThe Netherlands
| | - Erik Schoenmakers
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - Maura Agostini
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - Krishna Chatterjee
- Wellcome Trust‐MRC Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| |
Collapse
|
40
|
Sundaram SM, Arrulo Pereira A, Müller-Fielitz H, Köpke H, De Angelis M, Müller TD, Heuer H, Körbelin J, Krohn M, Mittag J, Nogueiras R, Prevot V, Schwaninger M. Gene therapy targeting the blood-brain barrier improves neurological symptoms in a model of genetic MCT8 deficiency. Brain 2022; 145:4264-4274. [PMID: 35929549 DOI: 10.1093/brain/awac243] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 06/03/2022] [Accepted: 06/22/2022] [Indexed: 12/27/2022] Open
Abstract
A genetic deficiency of the solute carrier monocarboxylate transporter 8 (MCT8), termed Allan-Herndon-Dudley syndrome, is an important cause of X-linked intellectual and motor disability. MCT8 transports thyroid hormones across cell membranes. While thyroid hormone analogues improve peripheral changes of MCT8 deficiency, no treatment of the neurological symptoms is available so far. Therefore, we tested a gene replacement therapy in Mct8- and Oatp1c1-deficient mice as a well-established model of the disease. Here, we report that targeting brain endothelial cells for Mct8 expression by intravenously injecting the vector AAV-BR1-Mct8 increased tri-iodothyronine (T3) levels in the brain and ameliorated morphological and functional parameters associated with the disease. Importantly, the therapy resulted in a long-lasting improvement in motor coordination. Thus, the data support the concept that MCT8 mediates the transport of thyroid hormones into the brain and indicate that a readily accessible vascular target can help overcome the consequences of the severe disability associated with MCT8 deficiency.
Collapse
Affiliation(s)
- Sivaraj M Sundaram
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Adriana Arrulo Pereira
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Helge Müller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Hannes Köpke
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Meri De Angelis
- Institute for Diabetes and Obesity, Helmholtz Zentrum Munich, Munich, and German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.,Institute of Experimental Genetics, Helmholtz Zentrum Munich, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum Munich, Munich, and German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Jakob Körbelin
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany.,Department of Oncology, Hematology and Bone Marrow Transplantation, UKE Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Markus Krohn
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Jens Mittag
- Institute for Endocrinology and Diabetes, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain
| | - Vincent Prevot
- Université Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, European Genomic Institute for Diabetes (EGID), 59045 Lille Cedex, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Hamburg-Lübeck-Kiel, Germany
| |
Collapse
|
41
|
Brûlé E, Silander TL, Wang Y, Zhou X, Bak B, Groeneweg S, Bernard DJ. IGSF1 Deficiency Leads to Reduced TSH Production Independent of Alterations in Thyroid Hormone Action in Male Mice. Endocrinology 2022; 163:6609251. [PMID: 35708735 PMCID: PMC9258739 DOI: 10.1210/endocr/bqac092] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Indexed: 11/19/2022]
Abstract
Loss of function mutations in IGSF1/Igsf1 cause central hypothyroidism. Igsf1 knockout mice have reduced pituitary thyrotropin-releasing hormone receptor, Trhr, expression, perhaps contributing to the phenotype. Because thyroid hormones negatively regulate Trhr, we hypothesized that IGSF1 might affect thyroid hormone availability in pituitary thyrotropes. Consistent with this idea, IGSF1 coimmunoprecipitated with the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) in transfected cells. This association was impaired with IGSF1 bearing patient-derived mutations. Wild-type IGSF1 did not, however, alter MCT8-mediated thyroid hormone import into heterologous cells. IGSF1 and MCT8 are both expressed in the apical membrane of the choroid plexus. However, MCT8 protein levels and localization in the choroid plexus were unaltered in Igsf1 knockout mice, ruling out a necessary chaperone function for IGSF1. MCT8 expression was low in the pituitary and was similarly unaffected in Igsf1 knockouts. We next assessed whether IGSF1 affects thyroid hormone transport or action, by MCT8 or otherwise, in vivo. To this end, we treated hypothyroid wild-type and Igsf1 knockout mice with exogenous thyroid hormones. T4 and T3 inhibited TSH release and regulated pituitary and forebrain gene expression similarly in both genotypes. Interestingly, pituitary TSH beta subunit (Tshb) expression was consistently reduced in Igsf1 knockouts relative to wild-type regardless of experimental condition, whereas Trhr was more variably affected. Although IGSF1 and MCT8 can interact in heterologous cells, the physiological relevance of their association is not clear. Nevertheless, the results suggest that IGSF1 loss can impair TSH production independently of alterations in TRHR levels or thyroid hormone action.
Collapse
Affiliation(s)
- Emilie Brûlé
- Department of Anatomy and Cell Biology, McGill University, Montreal H3G 1Y6, Canada
| | - Tanya L Silander
- Integrated Program in Neuroscience, McGill University, Montreal H3G 1Y6, Canada
| | - Ying Wang
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Xiang Zhou
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Beata Bak
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Academic Center for Thyroid Diseases, Rotterdam, The Netherlands
| | - Daniel J Bernard
- Correspondence: Daniel J. Bernard, PhD, Department of Pharmacology and Therapeutics, McGill University, McIntyre Medical Building, 3655 Prom. Sir William Osler, Room 1320, Montreal, Quebec H3G 1Y6, Canada.
| |
Collapse
|
42
|
Liao XH, Avalos P, Shelest O, Ofan R, Shilo M, Bresee C, Likhite S, Vit JP, Heuer H, Kaspar B, Meyer K, Dumitrescu AM, Refetoff S, Svendsen CN, Vatine GD. AAV9-MCT8 Delivery at Juvenile Stage Ameliorates Neurological and Behavioral Deficits in a Mouse Model of MCT8-Deficiency. Thyroid 2022; 32:849-859. [PMID: 35350867 PMCID: PMC9469747 DOI: 10.1089/thy.2022.0034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background: Allan-Herndon-Dudley syndrome (AHDS) is a severe psychomotor disability disorder that also manifests characteristic abnormal thyroid hormone (TH) levels. AHDS is caused by inactivating mutations in monocarboxylate transporter 8 (MCT8), a specific TH plasma membrane transporter widely expressed in the central nervous system (CNS). MCT8 mutations cause impaired transport of TH across brain barriers, leading to insufficient neural TH supply. There is currently no successful therapy for the neurological symptoms. Earlier work has shown that intravenous (IV), but not intracerebroventricular adeno-associated virus serotype 9 (AAV9) -based gene therapy given to newborn Mct8 knockout (Mct8-/y) male mice increased triiodothyronine (T3) brain content and partially rescued TH-dependent gene expression, suggesting a promising approach to treat this neurological disorder. Methods: The potential of IV delivery of AAV9 carrying human MCT8 was tested in the well-established Mct8-/y/Organic anion-transporting polypeptide 1c1 (Oatp1c1)-/ - double knockout (dKO) mouse model of AHDS, which, unlike Mct8-/y mice, displays both neurological and TH phenotype. Further, as the condition is usually diagnosed during childhood, treatment was given intravenously to P30 mice and psychomotor tests were carried out blindly at P120-P140 after which tissues were collected and analyzed. Results: Systemic IV delivery of AAV9-MCT8 at a juvenile stage led to improved locomotor and cognitive functions at P120-P140, which was accompanied by a near normalization of T3 content and an increased response of positively regulated TH-dependent gene expression in different brain regions examined (thalamus, hippocampus, and parietal cortex). The effects on serum TH concentrations and peripheral tissues were less pronounced, showing only improvement in the serum T3/reverse T3 (rT3) ratio and in liver deiodinase 1 expression. Conclusion: IV administration of AAV9, carrying the human MCT8, to juvenile dKO mice manifesting AHDS has long-term beneficial effects, predominantly on the CNS. This preclinical study indicates that this gene therapy has the potential to ameliorate the devastating neurological symptoms in patients with AHDS.
Collapse
Affiliation(s)
- Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Pablo Avalos
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Oksana Shelest
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Raz Ofan
- Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- The Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Michael Shilo
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Catherine Bresee
- Biostatistics Core, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shibi Likhite
- The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Jean-Philippe Vit
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University of Duisburg-Essen, Essen, Germany
| | - Brian Kaspar
- The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Kathrin Meyer
- The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | | | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, The University of Chicago, Chicago, Illinois, USA
- Committee on Genetics, The University of Chicago, Chicago, Illinois, USA
- Address correspondence to: Samuel Refetoff, MD, Department of Medicine, The University of Chicago, MC3090, 5841 South Maryland Avenue, Chicago, IL 60637, USA
| | - Clive N. Svendsen
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Address correspondence to: Clive N. Svendsen, PhD, The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Gad D. Vatine
- The Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Address correspondence to: Gad D. Vatine, PhD, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
43
|
Liu Z, Zhao S, Chen J, Ma L, Shi Q, Zhou Y. A novel frameshift mutation in Allan-Herndon-Dudley syndrome. Int J Legal Med 2022; 136:1181-1187. [PMID: 35391604 DOI: 10.1007/s00414-022-02823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/30/2022] [Indexed: 11/25/2022]
Abstract
Allan-Herndon-Dudley syndrome (AHDS) is a very rare, X-linked psychomotor disability syndrome with delayed myelination, almost exclusively affecting boys. We present a case of a 4-year-old boy with AHDS who was found cyanotic, with intermittent vomiting and paroxysmal convulsions about 4 h after his parents went out, and was then taken to the hospital, where he eventually died the next day. The autopsy revealed foreign bodies in the tiny bronchi and alveoli of the deceased, congestion, and punctate hemorrhage in multiple organs, consistent with the diagnosis of asphyxia. Compared with a normally developing 4-year-old boy, the deceased showed cerebral atrophy and cerebral edema, and Luxol Fast Blue (LFB) stain indicated delayed cerebellar, hippocampal, and basal ganglia development and myelination. A novel frameshift mutation c.584delG in the SLC16A2 gene was detected. Family lineage investigation showed that the mutation was also detected in the deceased's 8-year-old brother and biological mother. The present work enriches the profile mutations in SLC16A2 related to AHDS and emphasizes the importance of autopsy and postmortem genetic analysis in such cases.
Collapse
Affiliation(s)
- Zihao Liu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, People's Republic of China
| | - Shuquan Zhao
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, People's Republic of China
| | - Jianyi Chen
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, People's Republic of China
| | - Longda Ma
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, People's Republic of China
| | - Qing Shi
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, People's Republic of China
| | - Yiwu Zhou
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
44
|
Olivati C, Favilla BP, Freitas EL, Santos B, Melaragno MI, Meloni VA, Piazzon F. Allan-Herndon-Dudley syndrome in a female patient and related mechanisms. Mol Genet Metab Rep 2022; 31:100879. [PMID: 35782622 PMCID: PMC9248228 DOI: 10.1016/j.ymgmr.2022.100879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/30/2022] Open
Abstract
Allan-Herndon-Dudley syndrome (AHDS) is characterized by neuropsychomotor developmental delay/intellectual disability, neurological impairment with a movement disorder, and an abnormal thyroid hormone profile. This disease is an X-linked disorder that mainly affects men. We described a female patient with a de novo variant in the SLC16A2 gene, a milder AHDS phenotype, and a skewed X chromosome inactivation profile. We discuss the mechanisms associated with the expression of the phenotypic characteristics in female patients, including SLC16A2 gene variants and cytogenomic alterations, as well as preferential inactivation of the normal X chromosome.
Collapse
Affiliation(s)
- Caroline Olivati
- Rare Rosy Clinic, São Paulo, Brazil
- Fleury Medicina e Saúde, São Paulo, Brazil
- Corresponding author at: Rare Rosy Clinic, Rua Borges Lagoa, 1080, CEP 04038-020 São Paulo, SP, Brazil.
| | - Bianca Pereira Favilla
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - Maria Isabel Melaragno
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Vera Ayres Meloni
- Rare Rosy Clinic, São Paulo, Brazil
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Flavia Piazzon
- Rare Rosy Clinic, São Paulo, Brazil
- Neuromuscular Reference Center, Department of Pediatrics, University Hospital Liège & University of Liège, Belgium
- Neurometabolic Unit, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
Chen X, Liu L, Zeng C. A novel variant in SLC16A2 associated with typical Allan-Herndon-Dudley syndrome: a case report. BMC Pediatr 2022; 22:180. [PMID: 35382784 PMCID: PMC8981932 DOI: 10.1186/s12887-022-03259-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/31/2022] [Indexed: 11/23/2022] Open
Abstract
Background Allan-Herndon-Dudley syndrome (AHDS) is an X-linked recessive neurodegenerative disorder caused by mutations in the SLC16A2 gene that encodes thyroid hormone transporter. AHDS has been rarely reported in China. Case presentation This study reported a novel splicing mutation in the SLC16A2 gene in an 18-month-old male patient with AHDS. The patient was born to non-consanguineous, healthy parents of Chinese origin. He passed new-born screening for hypothyroidism, but failed to reach developmental milestones. He presented with hypotonia, severe mental retardation, dysarthria and ataxia. Genetic analysis identified a novel splicing mutation, NM_006517.4: c.431-2 A > G, in the SLC16A2 gene inherited from his mother. The patient received Triac treatment, (triiodothyroacetic acid), a thyroid hormone analogue for 3 months. Triac treatment effectively reduced serum TSH concentrations and normalized serum T3 concentrations in the patient. Conclusions This study reported the first case of AHDS treated by Triac in China. And the study expanded the mutational spectrum of the SLC16A2 gene in AHDS patients. Supplementary information The online version contains supplementary material available at 10.1186/s12887-022-03259-5.
Collapse
Affiliation(s)
- Xiaodan Chen
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, 510623, Guangzhou, China
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, 510623, Guangzhou, China.
| | - Chunhua Zeng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, 510623, Guangzhou, China.
| |
Collapse
|
46
|
Adams JW, Malicki D, Levy M, Crawford JR. Ganglioglioma with novel molecular features presenting in a child with Allan-Herndon-Dudley syndrome. BMJ Case Rep 2022; 15:e248734. [PMID: 35236707 PMCID: PMC8895953 DOI: 10.1136/bcr-2021-248734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2022] [Indexed: 11/03/2022] Open
Affiliation(s)
- Jason W Adams
- Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Denise Malicki
- Pathology, Rady Children's Hospital University of California San Diego, San Diego, California, USA
| | - Michael Levy
- Neurosurgery, University of California San Diego, San Diego, California, USA
| | - John Ross Crawford
- Neurosciences and Pediatrics, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
47
|
Abstract
Fetal and neonatal dysfunctions include rare serious disorders involving abnormal thyroid function during the second half of gestation, which may persist throughout life, as for most congenital thyroid disorders, or be transient, resolving in the first few weeks of life, as in autoimmune hyperthyroidism or hypothyroidism and some cases of congenital hypothyroidism (CH) with the thyroid gland in situ. Primary CH is diagnosed by neonatal screening, which has been implemented for 40 years in developed countries and should be introduced worldwide, as early treatment prevents irreversible neurodevelopmental delay. Central CH is a rarer entity occurring mostly in association with multiple pituitary hormone deficiencies. Other rare disorders impair the action of thyroid hormones. Neonatal Graves' disease (GD) results from the passage of thyrotropin receptor antibodies (TRAbs) across the placenta, from mother to fetus. It may affect the fetuses and neonates of mothers with a history of current or past GD, but hyperthyroidism develops only in those with high levels of stimulatory TRAb activity. The presence of antibodies predominantly blocking thyroid-stimulating hormone receptors may result in transient hypothyroidism, possibly followed by neonatal hyperthyroidism, depending on the balance between the antibodies present. Antithyroid drugs taken by the mother cross the placenta, treating potential fetal hyperthyroidism, but they may also cause transient fetal and neonatal hypothyroidism. Early diagnosis and treatment are key to optimizing the child's prognosis. This review focuses on the diagnosis and management of these patients during the fetal and neonatal periods. It includes the description of a case of fetal and neonatal autoimmune hyperthyroidism.
Collapse
Affiliation(s)
- Juliane Léger
- Assistance Publique-Hôpitaux de Paris, Robert Debré University Hospital, Pediatric Endocrinology-Diabetology Department, Reference Center for Growth and Development Endocrine Diseases, Paris, France
- Université de Paris; NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Clemence Delcour
- Université de Paris; NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
- Assistance Publique-Hôpitaux de Paris, Robert Debré University Hospital, Gynecology and Obstetric Department, Paris, France
| | - Jean-Claude Carel
- Assistance Publique-Hôpitaux de Paris, Robert Debré University Hospital, Pediatric Endocrinology-Diabetology Department, Reference Center for Growth and Development Endocrine Diseases, Paris, France
- Université de Paris; NeuroDiderot, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| |
Collapse
|
48
|
van Geest FS, Groeneweg S, van den Akker ELT, Bacos I, Barca D, van den Berg SAA, Bertini E, Brunner D, Brunetti-Pierri N, Cappa M, Cappuccio G, Chatterjee K, Chesover AD, Christian P, Coutant R, Craiu D, Crock P, Dewey C, Dica A, Dimitri P, Dubey R, Enderli A, Fairchild J, Gallichan J, Garibaldi LR, George B, Hackenberg A, Heinrich B, Huynh T, Kłosowska A, Lawson-Yuen A, Linder-Lucht M, Lyons G, Monti Lora F, Moran C, Müller KE, Paone L, Paul PG, Polak M, Porta F, Reinauer C, de Rijke YB, Seckold R, Menevşe TS, Simm P, Simon A, Spada M, Stoupa A, Szeifert L, Tonduti D, van Toor H, Turan S, Vanderniet J, de Waart M, van der Wal R, van der Walt A, van Wermeskerken AM, Wierzba J, Zibordi F, Zung A, Peeters RP, Visser WE. Long-Term Efficacy of T3 Analogue Triac in Children and Adults With MCT8 Deficiency: A Real-Life Retrospective Cohort Study. J Clin Endocrinol Metab 2022; 107:e1136-e1147. [PMID: 34679181 PMCID: PMC8852204 DOI: 10.1210/clinem/dgab750] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Patients with mutations in thyroid hormone transporter MCT8 have developmental delay and chronic thyrotoxicosis associated with being underweight and having cardiovascular dysfunction. OBJECTIVE Our previous trial showed improvement of key clinical and biochemical features during 1-year treatment with the T3 analogue Triac, but long-term follow-up data are needed. METHODS In this real-life retrospective cohort study, we investigated the efficacy of Triac in MCT8-deficient patients in 33 sites. The primary endpoint was change in serum T3 concentrations from baseline to last available measurement. Secondary endpoints were changes in other thyroid parameters, anthropometric parameters, heart rate, and biochemical markers of thyroid hormone action. RESULTS From October 15, 2014 to January 1, 2021, 67 patients (median baseline age 4.6 years; range, 0.5-66) were treated up to 6 years (median 2.2 years; range, 0.2-6.2). Mean T3 concentrations decreased from 4.58 (SD 1.11) to 1.66 (0.69) nmol/L (mean decrease 2.92 nmol/L; 95% CI, 2.61-3.23; P < 0.0001; target 1.4-2.5 nmol/L). Body-weight-for-age exceeded that of untreated historical controls (mean difference 0.72 SD; 95% CI, 0.36-1.09; P = 0.0002). Heart-rate-for-age decreased (mean difference 0.64 SD; 95% CI, 0.29-0.98; P = 0.0005). SHBG concentrations decreased from 245 (99) to 209 (92) nmol/L (mean decrease 36 nmol/L; 95% CI, 16-57; P = 0.0008). Mean creatinine concentrations increased from 32 (11) to 39 (13) µmol/L (mean increase 7 µmol/L; 95% CI, 6-9; P < 0.0001). Mean creatine kinase concentrations did not significantly change. No drug-related severe adverse events were reported. CONCLUSIONS Key features were sustainably alleviated in patients with MCT8 deficiency across all ages, highlighting the real-life potential of Triac for MCT8 deficiency.
Collapse
Affiliation(s)
- Ferdy S van Geest
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Stefan Groeneweg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Erica L T van den Akker
- Division of Endocrinology, Department of Pediatrics, Erasmus MC-Sophia Children's Hospital, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Iuliu Bacos
- Centrul Medical Dr. Bacos Cosma, Timisoara 307200, Romania
| | - Diana Barca
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest 050474, Romania
- Paediatric Neurology Clinic, Reference Center for Rare Paediatric Neurological Disorders, ENDO-ERN member, Alexandru Obregia Hospital, Bucharest 041914, Romania
| | - Sjoerd A A van den Berg
- Diagnostic Laboratory for Endocrinology, Department of Internal Medicine, Erasmus Medical Center , 3015 GD Rotterdam, The Netherlands
- Department of Clinical chemistry, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesu' Children's Research Hospital IRCCS, 00165 Rome, Italy
| | - Doris Brunner
- Gottfried Preyer's Children Hospital, 1100 Vienna, Austria
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, 80131 Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078 Naples, Italy
| | - Marco Cappa
- Division of Endocrinology, Bambino Gesu' Children's Research Hospital IRCCS, 00165 Rome, Italy
| | - Gerarda Cappuccio
- Department of Translational Medicine, Federico II University, 80131 Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078 Naples, Italy
| | - Krishna Chatterjee
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Alexander D Chesover
- Division of Endocrinology, The Hospital for Sick Children and Department of Paediatrics, University of Toronto, Toronto, M5G 1X8, Canada
| | - Peter Christian
- East Kent Hospitals University NHS Foundation Trust, Ashford TN24 0LZ, UK
| | - Régis Coutant
- Department of Pediatric Endocrinology and Diabetology, University Hospital, 49100 Angers, France
| | - Dana Craiu
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest 050474, Romania
- Paediatric Neurology Clinic, Reference Center for Rare Paediatric Neurological Disorders, ENDO-ERN member, Alexandru Obregia Hospital, Bucharest 041914, Romania
| | - Patricia Crock
- John Hunter Children's Hospital, New Lambton Heights, NSW 2305, Australia
- Hunter Medical Research Institute, University of Newcastle Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - Cheyenne Dewey
- Genomics Institute Mary Bridge Children's Hospital, MultiCare Health System Tacoma, WA 98403, USA
| | - Alice Dica
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest 050474, Romania
- Paediatric Neurology Clinic, Reference Center for Rare Paediatric Neurological Disorders, ENDO-ERN member, Alexandru Obregia Hospital, Bucharest 041914, Romania
| | - Paul Dimitri
- Sheffield Children's NHS Foundation Trust, Sheffield Hallam University and University of Sheffield, Sheffield, S10 2TH, UK
| | - Rachana Dubey
- Medanta Superspeciality Hospital, Indore 800020, India
| | - Anina Enderli
- Department of Neuropediatrics, University Children's Hospital Zurich, Steinwiesstrasse 75, 8032 Zürich, Switzerland
- Neurology Department, Children's Hospital, St. Gallen, 9000, Switzerland
| | - Jan Fairchild
- Department of Diabetes and Endocrinology, Women's and Children's Hospital, North Adelaide 5066 SouthAustralia
| | | | | | - Belinda George
- Department of Endocrinology, St. John's Medical College Hospital, Bengaluru 560034, India
| | - Annette Hackenberg
- Department of Neuropediatrics, University Children's Hospital Zurich, Steinwiesstrasse 75, 8032 Zürich, Switzerland
| | - Bianka Heinrich
- Department of Neuropediatrics, University Children's Hospital Zurich, Steinwiesstrasse 75, 8032 Zürich, Switzerland
| | - Tony Huynh
- Department of Endocrinology & Diabetes, Queensland Children's Hospital, South Brisbane Queensland 4101, Australia
- Department of Chemical Pathology, Mater Pathology, South Brisbane, Queensland 4101, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Anna Kłosowska
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Amy Lawson-Yuen
- Genomics Institute Mary Bridge Children's Hospital, MultiCare Health System Tacoma, WA 98403, USA
| | - Michaela Linder-Lucht
- Division of Neuropediatrics and Muscular Disorders, Department of Pediatrics and Adolescent Medicine, University Hospital Freiburg, 79106 Freiburg, Germany
| | - Greta Lyons
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Felipe Monti Lora
- Pediatric Endocrinology Group, Santa Catarina Hospital, São Paulo, 01310-000, Brazil
| | - Carla Moran
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Katalin E Müller
- Heim Pal National Institute of Pediatrics, Budapest, 1089, Hungary
- Institute of Translational Medicine, University of Pécs, Pécs, 7622, Hungary
| | - Laura Paone
- Division of Endocrinology, Bambino Gesu' Children's Research Hospital IRCCS, 00165 Rome, Italy
| | - Praveen G Paul
- Department of Paediatrics, Christian Medical College, Vellore 632004, India
| | - Michel Polak
- Paediatric Endocrinology, Diabetology and Gynaecology Department, Necker Children's University Hospital, Imagine Institute, Université de Paris, Paris 75015, France
| | - Francesco Porta
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Torino 10126,Italy
| | - Christina Reinauer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Medical Faculty, Duesseldorf 40225, Germany
| | - Yolanda B de Rijke
- Department of Clinical chemistry, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Rowen Seckold
- John Hunter Children's Hospital, New Lambton Heights, NSW 2305, Australia
- Hunter Medical Research Institute, University of Newcastle Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - Tuba Seven Menevşe
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul 34854, Turkey
| | - Peter Simm
- Royal Children's Hospital/University of Melbourne, Parkville 3052,Australia
| | - Anna Simon
- Department of Paediatrics, Christian Medical College, Vellore 632004, India
| | - Marco Spada
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Torino 10126,Italy
| | - Athanasia Stoupa
- Paediatric Endocrinology, Diabetology and Gynaecology Department, Necker Children's University Hospital, Imagine Institute, Université de Paris, Paris 75015, France
| | - Lilla Szeifert
- 1st Department of Pediatrics, Semmelweis University, Budapest, 1083, Hungary
| | - Davide Tonduti
- Child Neurology Unit - C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children's Hospital, Milano 20154, Italy
| | - Hans van Toor
- Diagnostic Laboratory for Endocrinology, Department of Internal Medicine, Erasmus Medical Center , 3015 GD Rotterdam, The Netherlands
| | - Serap Turan
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul 34854, Turkey
| | - Joel Vanderniet
- John Hunter Children's Hospital, New Lambton Heights, NSW 2305, Australia
- Hunter Medical Research Institute, University of Newcastle Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - Monique de Waart
- Department of Clinical chemistry, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Ronald van der Wal
- Diagnostic Laboratory for Endocrinology, Department of Internal Medicine, Erasmus Medical Center , 3015 GD Rotterdam, The Netherlands
| | - Adri van der Walt
- Private Paediatric Neurology Practice of Dr A van der Walt, Durbanville, South Africa
| | | | - Jolanta Wierzba
- Department of Internal and Pediatric Nursing, Institute of Nursing and Midwifery, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Federica Zibordi
- Child Neurology Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milan 20133, Italy
| | - Amnon Zung
- Pediatric Endocrinology Unit, Kaplan Medical Center, University of Jerusalem, Rehovot 76100, Israel
| | - Robin P Peeters
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - W Edward Visser
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
49
|
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
|
50
|
Masnada S, Sarret C, Antonello CE, Fadilah A, Krude H, Mura E, Mordekar S, Nicita F, Olivotto S, Orcesi S, Porta F, Remerand G, Siri B, Wilpert NM, Amir-Yazdani P, Bertini E, Schuelke M, Bernard G, Boespflug-Tanguy O, Tonduti D. Movement disorders in MCT8 deficiency/Allan-Herndon-Dudley Syndrome. Mol Genet Metab 2022; 135:109-113. [PMID: 34969638 DOI: 10.1016/j.ymgme.2021.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVES MCT8 deficiency is a rare genetic leukoencephalopathy caused by a defect of thyroid hormone transport across cell membranes, particularly through blood brain barrier and into neural cells. It is characterized by a complex neurological presentation, signs of peripheral thyrotoxicosis and cerebral hypothyroidism. Movement disorders (MDs) have been frequently mentioned in this condition, but not systematically studied. METHODS Each patient recruited was video-recorded during a routine outpatient visit according to a predefined protocol. The presence and the type of MDs were evaluated. The type of MD was blindly scored by two child neurologists experts in inherited white matter diseases and in MD. Dystonia was scored according to Burke-Fahn-Marsden Dystonia Rating Scale (BFMDRS). When more than one MD was present, the predominant one was scored. RESULTS 27 patients were included through a multicenter collaboration. In many cases we saw a combination of different MDs. Hypokinesia was present in 25/27 patients and was the predominant MD in 19. It was often associated with hypomimia and global hypotonia. Dystonia was observed in 25/27 patients, however, in a minority of cases (5) it was deemed the predominant MD. In eleven patients, exaggerated startle reactions and/or other paroxysmal non-epileptic events were observed. CONCLUSION MDs are frequent clinical features of MCT8 deficiency, possibly related to the important role of thyroid hormones in brain development and functioning of normal dopaminergic circuits of the basal ganglia. Dystonia is common, but usually mild to moderate in severity, while hypokinesia was the predominant MD in the majority of patients.
Collapse
Affiliation(s)
- Silvia Masnada
- Unit of Pediatric Neurology, V. Buzzi Children's Hospital, Milan, Italy; C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Milan, Italy.
| | - Catherine Sarret
- Centre de Compétence des Leucodystrophies et Leucoencéphalopathies de Cause Rare, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France.
| | - Clara Eleonora Antonello
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Milan, Italy; Department of Paediatric Orthopaedics, V. Buzzi Children's Hospital, Milan, Italy.
| | - Ala Fadilah
- Department of Paediatric Neurology, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom.
| | - Heiko Krude
- Institute of Experimental Pediatric Endocrinology, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Eleonora Mura
- Unit of Pediatric Neurology, V. Buzzi Children's Hospital, Milan, Italy; C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Milan, Italy
| | - Santosh Mordekar
- Department of Paediatric Neurology, Sheffield Children's NHS Foundation Trust, Sheffield, United Kingdom.
| | - Francesco Nicita
- Genetics and Rare Diseases Research Division, Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Sara Olivotto
- Unit of Pediatric Neurology, V. Buzzi Children's Hospital, Milan, Italy; C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Milan, Italy.
| | - Simona Orcesi
- Department of Brain and Behavioural Neurosciences, University of Pavia, Pavia, Italy; Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy.
| | - Francesco Porta
- Pediatric Department, Regina Margherita Hospital, Turin, Italy
| | - Ganaelle Remerand
- Service de Néonatologie, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Barbara Siri
- Pediatric Department, Regina Margherita Hospital, Turin, Italy; Metabolic Unit, Department Pediatrics, Bambino Gesù Children's Hospital, Italy.
| | - Nina-Maria Wilpert
- Department of Neuropediatrics, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Pouneh Amir-Yazdani
- Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montréal, Québec, Canada; Université Laval, Québec, Québec, Canada.
| | - Enrico Bertini
- Genetics and Rare Diseases Research Division, Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Markus Schuelke
- Department of Neuropediatrics, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Geneviève Bernard
- Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montréal, Québec, Canada; Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University, Montreal, Canada; Department Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal, Canada.
| | - Odile Boespflug-Tanguy
- Department of Pediatric Neurology and Metabolic Disorders, French Reference Center for Leukodystrophies, Robert Debré Hospital, Paris, France; Inserm UMR1141 Neuroprotect, Paris Diderot University, Sorbonne Cite, Paris, France
| | - Davide Tonduti
- Unit of Pediatric Neurology, V. Buzzi Children's Hospital, Milan, Italy; C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Milan, Italy.
| |
Collapse
|