1
|
Martinez ME, Karaczyn A, Wu Z, Bennett CA, Matoin KL, Daigle HM, Hernandez A. Transgenerational epigenetic self-memory of Dio3 dosage is associated with Meg3 methylation and altered growth trajectories and neonatal hormones. Epigenetics 2024; 19:2376948. [PMID: 38991122 PMCID: PMC11244338 DOI: 10.1080/15592294.2024.2376948] [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: 01/30/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
Intergenerational and transgenerational epigenetic effects resulting from conditions in previous generations can contribute to environmental adaptation as well as disease susceptibility. Previous studies in rodent and human models have shown that abnormal developmental exposure to thyroid hormone affects endocrine function and thyroid hormone sensitivity in later generations. Since the imprinted type 3 deiodinase gene (Dio3) regulates sensitivity to thyroid hormones, we hypothesize its epigenetic regulation is altered in descendants of thyroid hormone overexposed individuals. Using DIO3-deficient mice as a model of developmental thyrotoxicosis, we investigated Dio3 total and allelic expression and growth and endocrine phenotypes in descendants. We observed that male and female developmental overexposure to thyroid hormone altered total and allelic Dio3 expression in genetically intact descendants in a tissue-specific manner. This was associated with abnormal growth and neonatal levels of thyroid hormone and leptin. Descendant mice also exhibited molecular abnormalities in the Dlk1-Dio3 imprinted domain, including increased methylation in Meg3 and altered foetal brain expression of other genes of the Dlk1-Dio3 imprinted domain. These molecular abnormalities were also observed in the tissues and germ line of DIO3-deficient ancestors originally overexposed to thyroid hormone in utero. Our results provide a novel paradigm of epigenetic self-memory by which Dio3 gene dosage in a given individual, and its dependent developmental exposure to thyroid hormone, influences its own expression in future generations. This mechanism of epigenetic self-correction of Dio3 expression in each generation may be instrumental in descendants for their adaptive programming of developmental growth and adult endocrine function.
Collapse
Affiliation(s)
- M. Elena Martinez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Aldona Karaczyn
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Zhaofei Wu
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Christian A. Bennett
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Kassey L. Matoin
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Heather M. Daigle
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Arturo Hernandez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
- Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
2
|
Chincarini G, Walker DW, Wong F, Richardson SJ, Cumberland A, Tolcos M. Thyroid hormone analogues: Promising therapeutic avenues to improve the neurodevelopmental outcomes of intrauterine growth restriction. J Neurochem 2024; 168:2335-2350. [PMID: 38742992 DOI: 10.1111/jnc.16124] [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: 12/14/2023] [Revised: 04/14/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024]
Abstract
Intrauterine growth restriction (IUGR) is a pregnancy complication impairing fetal growth and development. The compromised development is often attributed to disruptions of oxygen and nutrient supply from the placenta, resulting in a number of unfavourable physiological outcomes with impaired brain and organ growth. IUGR is associated with compromised development of both grey and white matter, predisposing the infant to adverse neurodevelopmental outcomes, including long-lasting cognitive and motor difficulties. Cerebral thyroid hormone (TH) signalling, which plays a crucial role in regulating white and grey matter development, is dysregulated in IUGR, potentially contributing to the neurodevelopmental delays associated with this condition. Notably, one of the major TH transporters, monocarboxylate transporter-8 (MCT8), is deficient in the fetal IUGR brain. Currently, no effective treatment to prevent or reverse IUGR exists. Management strategies involve close antenatal monitoring, management of maternal risk factors if present and early delivery if IUGR is found to be severe or worsening in utero. The overall goal is to determine the most appropriate time for delivery, balancing the risks of preterm birth with further fetal compromise due to IUGR. Drug candidates have shown either adverse effects or little to no benefits in this vulnerable population, urging further preclinical and clinical investigation to establish effective therapies. In this review, we discuss the major neuropathology of IUGR driven by uteroplacental insufficiency and the concomitant long-term neurobehavioural impairments in individuals born IUGR. Importantly, we review the existing clinical and preclinical literature on cerebral TH signalling deficits, particularly the impaired expression of MCT8 and their correlation with IUGR. Lastly, we discuss the current evidence on MCT8-independent TH analogues which mimic the brain actions of THs by being metabolised in a similar manner as promising, albeit underappreciated approaches to promote grey and white matter development and improve the neurobehavioural outcomes following IUGR.
Collapse
Affiliation(s)
- Ginevra Chincarini
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - David W Walker
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
- Monash Newborn Health, Monash Medical Centre, Clayton, Melbourne, Victoria, Australia
| | - Flora Wong
- Monash Newborn Health, Monash Medical Centre, Clayton, Melbourne, Victoria, Australia
| | | | - Angela Cumberland
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Martinez ME, Wu Z, Hernandez A. Paternal developmental thyrotoxicosis disrupts neonatal leptin leading to increased adiposity and altered physiology of the melanocortin system. Front Endocrinol (Lausanne) 2023; 14:1210414. [PMID: 37560296 PMCID: PMC10407661 DOI: 10.3389/fendo.2023.1210414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023] Open
Abstract
Background The genetic code does not fully explain individual variability and inheritance of susceptibility to endocrine conditions, suggesting the contribution of epigenetic factors acting across generations. Methods We used a mouse model of developmental thyrotoxicosis (Dio3-/- mouse) to analyze endocrine outcomes in the adult offspring of Dio3-/- males using standard methods for body composition, and baseline and fasting hormonal and gene expression determinations in serum and tissues of relevance to the control of energy balance. Results Compared to controls, adult females with an exposed father (EF females) exhibited higher body weight and fat mass, but not lean mass, a phenotype that was much milder in EF males. After fasting, both EF females and males exhibited a more pronounced decrease in body weight than controls. EF females also showed markedly elevated serum leptin, increased white adipose tissue mRNA expression of leptin and mesoderm-specific transcript but decreased expression of type 2 deiodinase. EF females exhibited decreased serum ghrelin, which showed more pronounced post-fasting changes in EF females than in control females. EF female hypothalami also revealed significant decreases in the expression of pro-opiomelanocortin, agouti-related peptide, neuropeptide Y and melanocortin receptor 4. These markers also showed larger changes in response to fasting in EF females than in control females. Adult EF females showed no abnormalities in serum thyroid hormones, but pituitary expression of thyrotropin-releasing hormone receptor 1 and thyroid gland expression of thyroid-stimulating hormone receptor, thyroid peroxidase and iodotyrosine deiodinase were increased at baseline and showed differential regulation after fasting, with no increase in Trhr1 expression and more pronounced reductions in Tshr, Tpo and Iyd. In EF males, these abnormalities were generally milder. In addition, postnatal day 14 (P14) serum leptin was markedly reduced in EF pups. Discussion A paternal excess of thyroid hormone during development modifies the endocrine programming and energy balance in the offspring in a sexually dimorphic manner, with baseline and dynamic range alterations in the leptin-melanocortin system and thyroid gland, and consequences for adiposity phenotypes. We conclude that thyroid hormone overexposure may have important implications for the non-genetic, inherited etiology of endocrine and metabolic pathologies.
Collapse
Affiliation(s)
- Maria Elena Martinez
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
| | - Zhaofei Wu
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
| | - Arturo Hernandez
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
- Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
6
|
Yamauchi I, Hakata T, Ueda Y, Sugawa T, Omagari R, Teramoto Y, Nakayama SF, Nakajima D, Kubo T, Inagaki N. TRIAC disrupts cerebral thyroid hormone action via negative feedback and heterogenous distribution among organs. iScience 2023; 26:107135. [PMID: 37408688 PMCID: PMC10319255 DOI: 10.1016/j.isci.2023.107135] [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: 12/20/2022] [Revised: 04/25/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023] Open
Abstract
As 3,3',5-triiodothyroacetic acid (TRIAC), a metabolite of thyroid hormones (THs), was previously detected in sewage effluent, we aimed to investigate exogenous TRIAC's potential for endocrine disruption. We administered either TRIAC or 3,3',5-triiodo-L-thyronine (LT3) to euthyroid mice and 6-propyl-2-thiouracil-induced hypothyroid mice. In hypothyroid mice, TRIAC administration suppressed the hypothalamus-pituitary-thyroid (HPT) axis and upregulated TH-responsive genes in the pituitary gland, the liver, and the heart. We observed that, unlike LT3, TRIAC administration did not upregulate cerebral TH-responsive genes. Measurement of TRIAC contents suggested that TRIAC was not efficiently trafficked into the cerebrum. By analyzing euthyroid mice, we found that cerebral TRIAC content did not increase despite TRIAC administration at higher concentrations, whereas serum levels and cerebral contents of THs were substantially decreased. Disruption by TRIAC is due to the additive effects of circulating endogenous THs being depleted via a negative feedback loop involving the HPT axis and heterogeneous distribution of TRIAC among different organs.
Collapse
Affiliation(s)
- Ichiro Yamauchi
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takuro Hakata
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yohei Ueda
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Taku Sugawa
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ryo Omagari
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba, Ibaraki 305-8506, Japan
| | - Yasuo Teramoto
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba, Ibaraki 305-8506, Japan
| | - Shoji F. Nakayama
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba, Ibaraki 305-8506, Japan
| | - Daisuke Nakajima
- Health and Environmental Risk Division, National Institute for Environmental Studies, Tsukuba, Ibaraki 305-8506, Japan
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Takuya Kubo
- Department of Material Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto 615-8530, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
- Medical Research Institute KITANO HOSPITAL, PIIF Tazuke-kofukai, Osaka 530-8480, Japan
| |
Collapse
|
7
|
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
|
8
|
Grijota-Martínez C, Montero-Pedrazuela A, Hidalgo-Álvarez J, Bárez-López S, Guadaño-Ferraz A. Intracerebroventricular High Doses of 3,3',5-Triiodothyroacetic Acid at Juvenile Stages Improve Peripheral Hyperthyroidism and Mediate Thyromimetic Effects in Limited Brain Regions in a Mouse Model of Monocarboxylate Transporter 8 Deficiency. Thyroid 2023; 33:501-510. [PMID: 36565029 DOI: 10.1089/thy.2022.0562] [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] [Indexed: 12/25/2022]
Abstract
Introduction: Patients lacking functional monocarboxylate transporter 8 (MCT8), a highly specific thyroid hormone (TH) transporter, present severe psychomotor disabilities. MCT8 deficiency leads to peripheral hyperthyroidism and brain hypothyroidism, the latter due to impaired transport of TH across brain barriers. Available treatments for patients are limited and aim to overcome the limited TH transport across brain barriers. The use of TH analogues such as 3,3',5-triiodothyroacetic acid (TRIAC) that do not require MCT8 to cross the cellular membranes is considered a potential therapy for MCT8 deficiency. Previous studies have shown that systemic administration of TRIAC at therapeutic doses does not increase TRIAC content in the brain, while intracerebroventricular (ICV) administration of therapeutic doses of TRIAC increases TRIAC content in the brain but does not mediate thyromimetic effects. In view of this, we hypothesize that ICV administration of high doses of TRIAC can mediate thyromimetic effects in the brain without worsening the brain hypothyroidism or peripheral hyperthyroidism of patients. Methods: We administered 400 ng/g of body weight per day of ICV TRIAC in a mouse model of MCT8 deficiency: Mct8-/y and deiodinase 2 (Dio2)-/- double knockout mice. The effects of this treatment on TH and TRIAC levels/content in blood and tissues were determined by radioimmunoassay and effects on TH-regulated genes were assessed by real time-quantitative polymerase chain reaction in peripheral and central tissues. Results: ICV administration of high doses of TRIAC ameliorated the peripheral hyperthyroidism. In the brain, this treatment did not further aggravate brain hypothyroidism and increased TRIAC content in several brain regions; however, only moderate thyromimetic activity was observed in restricted brain areas. Conclusion: Administration of high doses of TRIAC by ICV delivery at juvenile stages in a mouse model of MCT8 deficiency is effective in normalizing peripheral hyperthyroidism but exerts minimal thyromimetic activity in the brain.
Collapse
Affiliation(s)
- Carmen Grijota-Martínez
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Montero-Pedrazuela
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Jorge Hidalgo-Álvarez
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Soledad Bárez-López
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Guadaño-Ferraz
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| |
Collapse
|
9
|
Martinez ME, Pinz I, Preda M, Norton CR, Gridley T, Hernandez A. DIO3 protects against thyrotoxicosis-derived cranio-encephalic and cardiac congenital abnormalities. JCI Insight 2022; 7:e161214. [PMID: 36166296 PMCID: PMC9675556 DOI: 10.1172/jci.insight.161214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/21/2022] [Indexed: 12/15/2022] Open
Abstract
Maternal hyperthyroidism is associated with an increased incidence of congenital abnormalities at birth, but it is not clear which of these defects arise from a transient developmental excess of thyroid hormone and which depend on pregnancy stage, antithyroid drug choice, or unwanted subsequent fetal hypothyroidism. To address this issue, we studied a mouse model of comprehensive developmental thyrotoxicosis secondary to a lack of type 3 deiodinase (DIO3). Dio3-/- mice exhibited reduced neonatal viability on most genetic backgrounds and perinatal lethality on a C57BL/6 background. Dio3-/- mice exhibited severe growth retardation during the neonatal period and cartilage loss. Mice surviving after birth manifested brain and cranial dysmorphisms, severe hydrocephalus, choanal atresia, and cleft palate. These abnormalities were noticeable in C57BL/6J Dio3-/- mice at fetal stages, in addition to a thyrotoxic heart with septal defects and thin ventricular walls. Our findings stress the protecting role of DIO3 during development and support the hypothesis that human congenital abnormalities associated with hyperthyroidism during pregnancy are caused by transient thyrotoxicosis before clinical intervention. Our results also suggest thyroid hormone involvement in the etiology of idiopathic pathologies including cleft palate, choanal atresia, Chiari malformations, Kaschin-Beck disease, and Temple and other cranio-encephalic and heart syndromes.
Collapse
Affiliation(s)
- M. Elena Martinez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, Maine, USA
| | - Ilka Pinz
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
- Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Marilena Preda
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, Maine, USA
| | - Christine R. Norton
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, Maine, USA
| | - Thomas Gridley
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
- Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Arturo Hernandez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
- Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Emamnejad R, Dass M, Mahlis M, Bozkurt S, Ye S, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. Thyroid hormone-dependent oligodendroglial cell lineage genomic and non-genomic signaling through integrin receptors. Front Pharmacol 2022; 13:934971. [PMID: 36133808 PMCID: PMC9483185 DOI: 10.3389/fphar.2022.934971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous autoimmune disease whereby the pathological sequelae evolve from oligodendrocytes (OLs) within the central nervous system and are targeted by the immune system, which causes widespread white matter pathology and results in neuronal dysfunction and neurological impairment. The progression of this disease is facilitated by a failure in remyelination following chronic demyelination. One mediator of remyelination is thyroid hormone (TH), whose reliance on monocarboxylate transporter 8 (MCT8) was recently defined. MCT8 facilitates the entry of THs into oligodendrocyte progenitor cell (OPC) and pre-myelinating oligodendrocytes (pre-OLs). Patients with MS may exhibit downregulated MCT8 near inflammatory lesions, which emphasizes an inhibition of TH signaling and subsequent downstream targeted pathways such as phosphoinositide 3-kinase (PI3K)-Akt. However, the role of the closely related mammalian target of rapamycin (mTOR) in pre-OLs during neuroinflammation may also be central to the remyelination process and is governed by various growth promoting signals. Recent research indicates that this may be reliant on TH-dependent signaling through β1-integrins. This review identifies genomic and non-genomic signaling that is regulated through mTOR in TH-responsive pre-OLs and mature OLs in mouse models of MS. This review critiques data that implicates non-genomic Akt and mTOR signaling in response to TH-dependent integrin receptor activation in pre-OLs. We have also examined whether this can drive remyelination in the context of neuroinflammation and associated sequelae. Importantly, we outline how novel therapeutic small molecules are being designed to target integrin receptors on oligodendroglial lineage cells and whether these are viable therapeutic options for future use in clinical trials for MS.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Michael Mahlis
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Paschalis Theotokis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
- *Correspondence: Steven Petratos,
| |
Collapse
|
11
|
Braun D, Schweizer U. The Protein Translocation Defect of MCT8 L291R Is Rescued by Sodium Phenylbutyrate. Eur Thyroid J 2020; 9:269-280. [PMID: 33088796 PMCID: PMC7548921 DOI: 10.1159/000507439] [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: 12/20/2019] [Accepted: 03/24/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The monocarboxylate transporter 8 (MCT8; SLC16A2) is a specific transporter for thyroid hormones. MCT8 deficiency, formerly known as the Allan-Herndon-Dudley syndrome, is a rare genetic disease that leads to neurological impairments and muscle weakness. Current experimental treatment options rely on thyromimetic agonists that do not depend on MCT8 for cellular uptake. Another approach comes from studies with the chemical chaperone sodium phenylbutyrate (NaPB), which was able to stabilize MCT8 mutants having protein folding defects in vitro. In addition, NaPB is known as a compound that assists with plasma membrane translocation. OBJECTIVE The pathogenic MCT8L291R leads to the same severe neurological impairments found for other MCT8-deficient patients but, unexpectedly, lacks alterations in plasma 3,3',5-triiodothyronine (T3) levels. Here we tried to unravel the underlying mechanism of MCT8 deficiency and tested whether the pathogenic MCT8L291R mutant responds to NaPB treatment. Therefore, we overexpressed the mutant in Madin-Darby canine kidney cells in the human choriocarcinoma cell line JEG1 and in COS7 cells of African green monkey origin. RESULTS In our recent study we describe that the MCT8L291R mutation most likely leads to a translocation defect. The pathogenic mutant is not located at the plasma membrane, but shows overlapping expression with a marker protein of the lysosome. Mutation of the corresponding amino acid in murine Mct8 (Mct8L223R) displays a similar effect on cell surface expression and transport function as seen before for MCT8L291R. NaPB was able to correct the translocation defect of MCT8L291R/Mct8L223R and restored protein function by increasing T3 transport activity. Furthermore, we detected enhanced mRNA levels of wild-type and mutant MCT8/Mct8 after NaPB treatment. The increase in mRNA levels could be an explanation for the positive effect on protein expression and function detected for wild-type MCT8. CONCLUSION NaPB is not only suitable for the treatment of mutations leading to misfolding and protein degradation, but also for a mutant wrongly sorted inside a cell which is otherwise functional.
Collapse
Affiliation(s)
- Doreen Braun
- *Doreen Braun, Institut für Biochemie und Molekularbiologie, Medizinische Fakultät, Rheinische Friedrich-Wilhelms-Universität Bonn, Nussallee 11, DE–53115 Bonn (Germany),
| | | |
Collapse
|
12
|
Grijota-Martínez C, Bárez-López S, Gómez-Andrés D, Guadaño-Ferraz A. MCT8 Deficiency: The Road to Therapies for a Rare Disease. Front Neurosci 2020; 14:380. [PMID: 32410949 PMCID: PMC7198743 DOI: 10.3389/fnins.2020.00380] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Allan-Herndon-Dudley syndrome is a rare disease caused by inactivating mutations in the SLC16A2 gene, which encodes the monocarboxylate transporter 8 (MCT8), a transmembrane transporter specific for thyroid hormones (T3 and T4). Lack of MCT8 function produces serious neurological disturbances, most likely due to impaired transport of thyroid hormones across brain barriers during development resulting in severe brain hypothyroidism. Patients also suffer from thyrotoxicity in other organs due to the presence of a high concentration of T3 in the serum. An effective therapeutic strategy should restore thyroid hormone serum levels (both T3 and T4) and should address MCT8 transporter deficiency in brain barriers and neural cells, to enable the access of thyroid hormones to target neural cells. Unfortunately, targeted therapeutic options are currently scarce and their effect is limited to an improvement in the thyrotoxic state, with no sign of any neurological improvement. The use of thyroid hormone analogs such as TRIAC, DITPA, or sobetirome, that do not require MCT8 to cross cell membranes and whose controlled thyromimetic activity could potentially restore the normal function of the affected organs, are being explored to improve the cerebral availability of these analogs. Other strategies aiming to restore the transport of THs through MCT8 at the brain barriers and the cellular membranes include gene replacement therapy and the use of pharmacological chaperones. The design of an appropriate therapeutic strategy in combination with an early diagnosis (at prenatal stages), will be key aspects to improve the devastating alterations present in these patients.
Collapse
Affiliation(s)
- Carmen Grijota-Martínez
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain.,Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid, Madrid, Spain
| | - Soledad Bárez-López
- Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain.,Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, United Kingdom
| | - David Gómez-Andrés
- Pediatric Neurology, Vall d'Hebron University Hospital and VHIR (Euro-NMD, ERN-RND), Barcelona, Spain
| | - Ana Guadaño-Ferraz
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
13
|
Krude H, Biebermann H, Schuelke M, Müller TD, Tschöp M. Allan-Herndon-Dudley-Syndrome: Considerations about the Brain Phenotype with Implications for Treatment Strategies. Exp Clin Endocrinol Diabetes 2020; 128:414-422. [PMID: 32242326 DOI: 10.1055/a-1108-1456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite its first description more than 75 years ago, effective treatment for "Allan-Herndon-Dudley-Syndrome (AHDS)", an X-linked thyroid hormone transporter defect, is unavailable. Mutations in the SLC16A2 gene have been discovered to be causative for AHDS in 2004, but a comprehensive understanding of the function of the encoded protein, monocarboxylate transporter 8 (MCT8), is incomplete. Patients with AHDS suffer from neurodevelopmental delay, as well as extrapyramidal (dystonia, chorea, athetosis), pyramidal (spasticity), and cerebellar symptoms (ataxia). This suggests an affection of the pyramidal tracts, basal ganglia, and cerebellum, most likely already during fetal brain development. The function of other brain areas relevant for mood, behavior, and vigilance seems to be intact. An optimal treatment strategy should thus aim to deliver T3 to these relevant structures at the correct time points during development. A potential therapeutic strategy meeting these needs might be the delivery of T3 via a "Trojan horse mechanism" by which T3 is delivered into target cells by a thyroid hormone transporter independent T3 internalization.
Collapse
Affiliation(s)
- Heiko Krude
- Institute of Experimental Pediatric Endocrinology, Charité - Universitätsmedizin, Berlin, Germany
| | - Heike Biebermann
- Institute of Experimental Pediatric Endocrinology, Charité - Universitätsmedizin, Berlin, Germany
| | - Markus Schuelke
- Department of Neuropediatrics, Charité - Universitätsmedizin, Berlin, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Centre Munich, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany
| | - Matthias Tschöp
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Helmholtz Zentrum München, Germany
| |
Collapse
|
14
|
Groeneweg S, van Geest FS, Peeters RP, Heuer H, Visser WE. Thyroid Hormone Transporters. Endocr Rev 2020; 41:5637505. [PMID: 31754699 DOI: 10.1210/endrev/bnz008] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ferdy S van Geest
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - W Edward Visser
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
15
|
Bárez-López S, Grijota-Martínez C, Liao XH, Refetoff S, Guadaño-Ferraz A. Intracerebroventricular administration of the thyroid hormone analog TRIAC increases its brain content in the absence of MCT8. PLoS One 2019; 14:e0226017. [PMID: 31809508 PMCID: PMC6897405 DOI: 10.1371/journal.pone.0226017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/19/2019] [Indexed: 01/06/2023] Open
Abstract
Patients lacking the thyroid hormone (TH) transporter MCT8 present abnormal serum levels of TH: low thyroxine and high triiodothyronine. They also have severe neurodevelopmental defects resulting from cerebral hypothyroidism, most likely due to impaired TH transport across the brain barriers. The use of TH analogs, such as triiodothyroacetic acid (TRIAC), that can potentially access the brain in the absence of MCT8 and restore at least a subset of cerebral TH actions could improve the neurological defects in these patients. We hypothesized that direct administration of TRIAC into the brain by intracerebroventricular delivery to mice lacking MCT8 could bypass the restriction at the brain barriers and mediate TH action without causing hypermetabolism. We found that intracerebroventricular administration of therapeutic doses of TRIAC does not increase further plasma triiodothyronine or further decrease plasma thyroxine levels and does not alter TH content in the cerebral cortex. Although TRIAC content increased in the brain, it did not induce TH-mediated actions on selected target genes. Our data suggest that intracerebroventricular delivery of TRIAC has the ability to target the brain in the absence of MCT8 and should be further investigated to address its potential therapeutic use in MCT8 deficiency.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Grijota-Martínez
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid, Madrid, Spain
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States of America
- Department of Pediatrics, The University of Chicago, Chicago, Illinois, United States of America
- Committee on Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Ana Guadaño-Ferraz
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
16
|
Groeneweg S, Peeters RP, Moran C, Stoupa A, Auriol F, Tonduti D, Dica A, Paone L, Rozenkova K, Malikova J, van der Walt A, de Coo IFM, McGowan A, Lyons G, Aarsen FK, Barca D, van Beynum IM, van der Knoop MM, Jansen J, Manshande M, Lunsing RJ, Nowak S, den Uil CA, Zillikens MC, Visser FE, Vrijmoeth P, de Wit MCY, Wolf NI, Zandstra A, Ambegaonkar G, Singh Y, de Rijke YB, Medici M, Bertini ES, Depoorter S, Lebl J, Cappa M, De Meirleir L, Krude H, Craiu D, Zibordi F, Oliver Petit I, Polak M, Chatterjee K, Visser TJ, Visser WE. Effectiveness and safety of the tri-iodothyronine analogue Triac in children and adults with MCT8 deficiency: an international, single-arm, open-label, phase 2 trial. Lancet Diabetes Endocrinol 2019; 7:695-706. [PMID: 31377265 PMCID: PMC7611958 DOI: 10.1016/s2213-8587(19)30155-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/18/2019] [Accepted: 04/18/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Deficiency of the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) causes severe intellectual and motor disability and high serum tri-iodothyronine (T3) concentrations (Allan-Herndon-Dudley syndrome). This chronic thyrotoxicosis leads to progressive deterioration in bodyweight, tachycardia, and muscle wasting, predisposing affected individuals to substantial morbidity and mortality. Treatment that safely alleviates peripheral thyrotoxicosis and reverses cerebral hypothyroidism is not yet available. We aimed to investigate the effects of treatment with the T3 analogue Triac (3,3',5-tri-iodothyroacetic acid, or tiratricol), in patients with MCT8 deficiency. METHODS In this investigator-initiated, multicentre, open-label, single-arm, phase 2, pragmatic trial, we investigated the effectiveness and safety of oral Triac in male paediatric and adult patients with MCT8 deficiency in eight countries in Europe and one site in South Africa. Triac was administered in a predefined escalating dose schedule-after the initial dose of once-daily 350 μg Triac, the daily dose was increased progressively in 350 μg increments, with the goal of attaining serum total T3 concentrations within the target range of 1·4-2·5 nmol/L. We assessed changes in several clinical and biochemical signs of hyperthyroidism between baseline and 12 months of treatment. The prespecified primary endpoint was the change in serum T3 concentrations from baseline to month 12. The co-primary endpoints were changes in concentrations of serum thyroid-stimulating hormone (TSH), free and total thyroxine (T4), and total reverse T3 from baseline to month 12. These analyses were done in patients who received at least one dose of Triac and had at least one post-baseline evaluation of serum throid function. This trial is registered with ClinicalTrials.gov, number NCT02060474. FINDINGS Between Oct 15, 2014, and June 1, 2017, we screened 50 patients, all of whom were eligible. Of these patients, four (8%) patients decided not to participate because of travel commitments. 46 (92%) patients were therefore enrolled in the trial to receive Triac (median age 7·1 years [range 0·8-66·8]). 45 (98%) participants received Triac and had at least one follow-up measurement of thyroid function and thus were included in the analyses of the primary endpoints. Of these 45 patients, five did not complete the trial (two patients withdrew [travel burden, severe pre-existing comorbidity], one was lost to follow-up, one developed of Graves disease, and one died of sepsis). Patients required a mean dose of 38.3 μg/kg of bodyweight (range 6·4-84·3) to attain T3 concentrations within the target range. Serum T3 concentration decreased from 4·97 nmol/L (SD 1·55) at baseline to 1·82 nmol/L (0·69) at month 12 (mean decrease 3·15 nmol/L, 95% CI 2·68-3·62; p<0·0001), while serum TSH concentrations decreased from 2·91 mU/L (SD 1·68) to 1·02 mU/L (1·14; mean decrease 1·89 mU/L, 1·39-2·39; p<0·0001) and serum free T4 concentrations decreased from 9·5 pmol/L (SD 2·5) to 3·4 (1·6; mean decrease 6·1 pmol/L (5·4-6·8; p<0·0001). Additionally, serum total T4 concentrations decreased by 31·6 nmol/L (28·0-35·2; p<0·0001) and reverse T3 by 0·08 nmol/L (0·05-0·10; p<0·0001). Seven treatment-related adverse events (transiently increased perspiration or irritability) occurred in six (13%) patients. 26 serious adverse events that were considered unrelated to treatment occurred in 18 (39%) patients (mostly hospital admissions because of infections). One patient died from pulmonary sepsis leading to multi-organ failure, which was unrelated to Triac treatment. INTERPRETATION Key features of peripheral thyrotoxicosis were alleviated in paediatric and adult patients with MCT8 deficiency who were treated with Triac. Triac seems a reasonable treatment strategy to ameliorate the consequences of untreated peripheral thyrotoxicosis in patients with MCT8 deficiency. FUNDING Dutch Scientific Organization, Sherman Foundation, NeMO Foundation, Wellcome Trust, UK National Institute for Health Research Cambridge Biomedical Centre, Toulouse University Hospital, and Una Vita Rara ONLUS.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Academic Center for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Robin P Peeters
- Academic Center for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Carla Moran
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Athanasia Stoupa
- Paediatric Endocrinology, Diabetology and Gynaecology Department, Necker Children's University Hospital, Imagine Institute, Paris, France
| | - Françoise Auriol
- Department of Paediatric Endocrinology and Genetics, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Davide Tonduti
- Child Neurology Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alice Dica
- Paediatric Neurology Clinic, Alexandru Obregia Hospital, Bucharest, Romania
| | - Laura Paone
- Division of Endocrinology, Bambino Gesu' Children's Research Hospital IRCCS, Rome, Italy
| | - Klara Rozenkova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Jana Malikova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | | | - Irenaeus F M de Coo
- Sophia Children's Hospital, Department of Paediatric Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Anne McGowan
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Greta Lyons
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Femke K Aarsen
- Sophia Children's Hospital, Department of Paediatric Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Diana Barca
- Paediatric Neurology Clinic, Alexandru Obregia Hospital, Bucharest, Romania; Department of Neurosciences, Paediatric Neurology Discipline II, Carol Davila University of Medicine, Bucharest, Romania
| | - Ingrid M van Beynum
- Sophia Children's Hospital, Division of Paediatric Cardiology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Marieke M van der Knoop
- Sophia Children's Hospital, Department of Paediatric Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Jurgen Jansen
- Department of Paediatrics, Meander Medical Center, Amersfoort, Netherlands
| | | | - Roelineke J Lunsing
- Department of Child Neurology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Stan Nowak
- Department of Paediatrics, Refaja Hospital, Stadskanaal, Netherlands
| | - Corstiaan A den Uil
- Department of Cardiology and Intensive Care Medicine, Erasmus Medical Centre, Rotterdam, Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, Netherlands
| | | | | | - Marie Claire Y de Wit
- Sophia Children's Hospital, Department of Paediatric Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Amsterdam Neuroscience, Amsterdam, Netherlands
| | | | - Gautam Ambegaonkar
- Department of Paediatric Neurology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Yogen Singh
- Department of Paediatric Cardiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Marco Medici
- Academic Center for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Enrico S Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesu' Children's Research Hospital IRCCS, Rome, Italy
| | - Sylvia Depoorter
- Department of Paediatrics, Algemeen Ziekenhuis Sint-Jan, Bruges, Belgium
| | - Jan Lebl
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Marco Cappa
- Division of Endocrinology, Bambino Gesu' Children's Research Hospital IRCCS, Rome, Italy
| | - Linda De Meirleir
- Paediatric Neurology Unit, Department of Paediatrics, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Heiko Krude
- Department of Paediatric Endocrinology and Diabetology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dana Craiu
- Paediatric Neurology Clinic, Alexandru Obregia Hospital, Bucharest, Romania; Department of Neurosciences, Paediatric Neurology Discipline II, Carol Davila University of Medicine, Bucharest, Romania
| | - Federica Zibordi
- Child Neurology Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Isabelle Oliver Petit
- Department of Paediatric Endocrinology and Genetics, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Michel Polak
- Paediatric Endocrinology, Diabetology and Gynaecology Department, Necker Children's University Hospital, Imagine Institute, Paris, France
| | - Krishna Chatterjee
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Theo J Visser
- Academic Center for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, Netherlands
| | - W Edward Visser
- Academic Center for Thyroid Diseases, Erasmus Medical Centre, Rotterdam, Netherlands.
| |
Collapse
|
17
|
Köhrle J. The Colorful Diversity of Thyroid Hormone Metabolites. Eur Thyroid J 2019; 8:115-129. [PMID: 31259154 PMCID: PMC6587369 DOI: 10.1159/000497141] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/22/2019] [Indexed: 12/17/2022] Open
Abstract
Since the discovery of L-thyroxine, the main secretory product of the thyroid gland, and its major metabolite T3, which exerts the majority of thyroid hormone action via ligand-dependent modulation of the function of T3 receptors in nuclei, mitochondria, and other subcellular compartments, various other T4-derived endogenous metabolites have been identified in blood and tissues of humans, animals, and early protochordates. This review addresses major historical milestones and experimental findings resulting in the discovery of the key enzymes of thyroid hormone metabolism, the three selenoprotein deiodinases, as well as the decarboxylases and amine oxidases involved in formation and degradation of recently identified endogenous thyroid hormone metabolites, i.e. 3-iodothyronamine and 3-thyroacetic acid. The concerted action of deiodinases 2 and 3 in regulation of local T3 availability is discussed. Special attention is given to the role of the thyromimetic "hot" metabolite 3,5-T2 and the "cool" 3-iodothyronamine, especially after administration of pharmacological doses of these endogenous thyroid hormone metabolites in various animal experimental models. In addition, available information on the biological roles of the two major acetic acid derivatives of thyroid hormones, i.e. Tetrac and Triac, as well as sulfated metabolites of thyroid hormones is reviewed. This review addresses the consequences of the existence of this broad spectrum of endogenous thyroid hormone metabolites, the "thyronome," beyond the classical thyroid hormone profile comprising T4, T3, and rT3 for appropriate analytical coverage and clinical diagnostics using mass spectrometry versus immunoassays for determination of total and free concentrations of thyroid hormone metabolites in blood and tissues.
Collapse
Affiliation(s)
- Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité Campus Virchow-Klinikum (CVK), Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
18
|
Kim MJ, Petratos S. Oligodendroglial Lineage Cells in Thyroid Hormone-Deprived Conditions. Stem Cells Int 2019; 2019:5496891. [PMID: 31182964 PMCID: PMC6515029 DOI: 10.1155/2019/5496891] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/20/2019] [Indexed: 01/06/2023] Open
Abstract
Oligodendrocytes are supporting glial cells that ensure the metabolism and homeostasis of neurons with specific synaptic axoglial interactions in the central nervous system. These require key myelinating glial trophic signals important for growth and metabolism. Thyroid hormone (TH) is one such trophic signal that regulates oligodendrocyte maturation, myelination, and oligodendroglial synaptic dynamics via either genomic or nongenomic pathways. The intracellular and extracellular transport of TH is facilitated by a specific transmembrane transporter known as the monocarboxylate transporter 8 (MCT8). Dysfunction of the MCT8 due to mutation, inhibition, or downregulation during brain development leads to inherited hypomyelination, which manifests as psychomotor retardation in the X-linked inherited Allan-Herndon-Dudley syndrome (AHDS). In particular, oligodendroglial-specific MCT8 deficiency may restrict the intracellular T3 availability, culminating in deficient metabolic communication between the oligodendrocytes and the neurons they ensheath, potentially promulgating neurodegenerative adult diseases such as multiple sclerosis (MS). Based on the therapeutic effects exhibited by TH in various preclinical studies, particularly related to its remyelinating potential, TH has now entered the initial stages of a clinical trial to test the therapeutic efficacy in relapsing-remitting MS patients (NCT02506751). However, TH analogs, such as DITPA or Triac, may well serve as future therapeutic options to rescue mature oligodendrocytes and/or promote oligodendrocyte precursor cell differentiation in an environment of MCT8 deficiency within the CNS. This review outlines the therapeutic strategies to overcome the differentiation blockade of oligodendrocyte precursors and maintain mature axoglial interactions in TH-deprived conditions.
Collapse
Affiliation(s)
- Min Joung Kim
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria 3004, Australia
| |
Collapse
|
19
|
Rurale G, Cicco ED, Dentice M, Salvatore D, Persani L, Marelli F, Luongo C. Thyroid Hormone Hyposensitivity: From Genotype to Phenotype and Back. Front Endocrinol (Lausanne) 2019; 10:912. [PMID: 32038483 PMCID: PMC6992580 DOI: 10.3389/fendo.2019.00912] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/16/2019] [Indexed: 01/24/2023] Open
Abstract
Thyroid hormone action defects (THADs) have been classically considered conditions of impaired sensitivity to thyroid hormone (TH). They were originally referring to alterations in TH receptor genes (THRA and THRB), but the discovery of genetic mutations and polymorphisms causing alterations in cell membrane transport (e.g., MCT8) and metabolism (e.g., SECISBP2, DIO2) led recently to a new and broader definition of TH hyposensitivity (THH), including not only THADs but all defects that could interfere with the activity of TH. Due to the different functions and tissue-specific expression of these genes, affected patients exhibit highly variable phenotypes. Some of them are characterized by a tissue hypothyroidism or well-recognizable alterations in the thyroid function tests (TFTs), whereas others display a combination of hypo- and hyperthyroid manifestations with normal or only subtle biochemical defects. The huge effort of basic research has greatly aided the comprehension of the molecular mechanisms underlying THADs, dissecting the morphological and functional alterations on target tissues, and defining the related-changes in the biochemical profile. In this review, we describe different pictures in which a specific alteration in the TFTs (TSH, T4, and T3 levels) is caused by defects in a specific gene. Altogether these findings can help clinicians to early recognize and diagnose THH and to perform a more precise genetic screening and therapeutic intervention. On the other hand, the identification of new genetic variants will allow the generation of cell-based and animal models to give novel insight into thyroid physiology and establish new therapeutic interventions.
Collapse
Affiliation(s)
- Giuditta Rurale
- Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Emery Di Cicco
- Department of Clinical Medicine & Surgery, University of Naples Federico II, Naples, Italy
| | - Monica Dentice
- Department of Clinical Medicine & Surgery, University of Naples Federico II, Naples, Italy
| | - Domenico Salvatore
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Luca Persani
- Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Federica Marelli
- Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- *Correspondence: Federica Marelli
| | - Cristina Luongo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
20
|
|
21
|
Vancamp P, Darras VM. From zebrafish to human: A comparative approach to elucidate the role of the thyroid hormone transporter MCT8 during brain development. Gen Comp Endocrinol 2018; 265:219-229. [PMID: 29183795 DOI: 10.1016/j.ygcen.2017.11.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
Abstract
Monocarboxylate transporter 8 (MCT8) facilitates transmembrane transport of thyroid hormones (THs) ensuring their action on gene expression during vertebrate neurodevelopment. A loss of MCT8 in humans results in severe psychomotor deficits associated with the Allan-Herndon-Dudley Syndrome (AHDS). However, where and when exactly a lack of MCT8 causes the neurological manifestations remains unclear because of the varying expression pattern of MCT8 between specific brain regions and cells. Here, we elaborate on the animal models that have been generated to elucidate the mechanisms underlying MCT8-deficient brain development. The absence of a clear neurological phenotype in Mct8 knockout mice made it clear that a single species would not suffice. The evolutionary conservation of TH action on neurodevelopment as well as the components regulating TH signalling however offers the opportunity to answer different aspects of MCT8 function in brain development using different vertebrate species. Moreover, the plethora of tools for genome editing available today facilitates gene silencing in these animals as well. Studies in the recently generated mct8-deficient zebrafish and Mct8/Oatp1c1 double knockout mice have put forward the current paradigm of impaired TH uptake at the level of the blood-brain barrier during peri- and postnatal development as being the main pathophysiological mechanism of AHDS. RNAi vector-based, cell-specific induction of MCT8 knockdown in the chicken embryo points to an additional function of MCT8 at the level of the neural progenitors during early brain development. Future studies including also additional in vivo models like Xenopus or in vitro approaches such as induced pluripotent stem cells will continue to help unravelling the exact role of MCT8 in developmental events. In the end, this multispecies approach will lead to a unifying thesis regarding the cellular and molecular mechanisms responsible for the neurological phenotype in AHDS patients.
Collapse
Affiliation(s)
- Pieter Vancamp
- KU Leuven, Laboratory of Comparative Endocrinology, Department of Biology, B-3000 Leuven, Belgium
| | - Veerle M Darras
- KU Leuven, Laboratory of Comparative Endocrinology, Department of Biology, B-3000 Leuven, Belgium.
| |
Collapse
|
22
|
Bárez-López S, Hartley MD, Grijota-Martínez C, Scanlan TS, Guadaño-Ferraz A. Sobetirome and its Amide Prodrug Sob-AM2 Exert Thyromimetic Actions in Mct8-Deficient Brain. Thyroid 2018; 28:1211-1220. [PMID: 29845892 PMCID: PMC6154442 DOI: 10.1089/thy.2018.0008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Loss of function mutations in the thyroid hormone (TH)-specific cell membrane transporter, the monocarboxylate transporter 8 (MCT8), lead to profound psychomotor retardation and abnormal TH serum levels, with low thyroxine (T4) and high triiodothyronine (T3). Several studies point to impaired TH transport across brain barriers as a crucial pathophysiological mechanism resulting in cerebral hypothyroidism. Treatment options for MCT8-deficient patients are limited and are focused on overcoming the brain barriers. The aim of this study was to evaluate the ability of the TH analog sobetirome and its prodrug Sob-AM2 to access the brain and exert thyromimetic actions in the absence of Mct8. METHODS Juvenile wild-type (Wt) mice and mice lacking Mct8 and deiodinase type 2 (Mct8/Dio2KO) were treated systemically with daily injections of vehicle, 1 mg of sobetirome/kg body weight/day, or 0.3 mg of Sob-AM2/kg body weight/day for seven days. Sobetirome content was measured using liquid chromatography-tandem mass spectrometry, and T4 and T3 levels by specific radioimmunoassays. The effect of sobetirome treatment in the expression of T3-dependent genes was measured in the heart, liver, and cerebral cortex by real-time polymerase chain reaction. RESULTS Sob-AM2 treatment in Mct8/Dio2KO animals led to 1.8-fold more sobetirome content in the brain and 2.5-fold less in plasma in comparison to the treatment with the parent drug sobetirome. Both sobetirome and Sob-AM2 treatments in Mct8/Dio2KO mice greatly decreased plasma T4 and T3 levels. Dio1 and Ucp2 gene expression was altered in the liver of Mct8/Dio2KO mice and was not affected by the treatments. In the heart, Hcn2 but not Atp2a2 expression was increased after treatment with the analogs. Interestingly, both sobetirome and Sob-AM2 treatments increased the expression of several T3-dependent genes in the brain such as Hr, Abcd2, Mme, and Flywch2 in Mct8/Dio2KO mice. CONCLUSIONS Sobetirome and its amide prodrug Sob-AM2 can access the brain in the absence of Mct8 and exert thyromimetic actions modulating the expression of T3-dependent genes. At the peripheral level, the administration of these TH analogs results in the depletion of circulating T4 and T3. Therefore, sobetirome and Sob-AM2 have the potential to address the cerebral hypothyroidism and the peripheral hyperthyroidism characteristic of MCT8 deficiency.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Center for Biomedical Research on Rare Diseases (Ciberer), Unit 708, Instituto de Salud Carlos III, Madrid, Spain
| | - Meredith D. Hartley
- Department of Physiology and Pharmacology and Program in Chemical Biology, Oregon Health and Science University, Portland, Oregon
| | - Carmen Grijota-Martínez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Thomas S. Scanlan
- Department of Physiology and Pharmacology and Program in Chemical Biology, Oregon Health and Science University, Portland, Oregon
- Address correspondence to:Thomas S. Scanlan, PhDDepartment of Physiology and Pharmacology and Program in Chemical BiologyOregon Health and Science UniversityPortland, OR 97239
| | - Ana Guadaño-Ferraz
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Center for Biomedical Research on Rare Diseases (Ciberer), Unit 708, Instituto de Salud Carlos III, Madrid, Spain
- Ana Guadaño-Ferraz, PhDDepartment of Endocrine and Nervous System PathophysiologyInstituto de Investigaciones Biomédicas Alberto SolsConsejo Superior de Investigaciones Científicas-Universidad Autónoma de MadridArturo Duperier 4E-28029 MadridSpain
| |
Collapse
|
23
|
Bárez-López S, Obregon MJ, Bernal J, Guadaño-Ferraz A. Thyroid Hormone Economy in the Perinatal Mouse Brain: Implications for Cerebral Cortex Development. Cereb Cortex 2018; 28:1783-1793. [PMID: 28407057 DOI: 10.1093/cercor/bhx088] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Indexed: 12/24/2022] Open
Abstract
Thyroid hormones (THs, T4 and the transcriptionally active hormone T3) play an essential role in neurodevelopment; however, the mechanisms underlying T3 brain delivery during mice fetal development are not well known. This work has explored the sources of brain T3 during mice fetal development using biochemical, anatomical, and molecular approaches. The findings revealed that during late gestation, a large amount of fetal brain T4 is of maternal origin. Also, in the developing mouse brain, fetal T3 content is regulated through the conversion of T4 into T3 by type-2 deiodinase (D2) activity, which is present from earlier prenatal stages. Additionally, D2 activity was found to be essential to mediate expression of T3-dependent genes in the cerebral cortex, and also necessary to generate the transient cerebral cortex hyperthyroidism present in mice lacking the TH transporter Monocarboxylate transporter 8. Notably, the gene encoding for D2 (Dio2) was mainly expressed at the blood-cerebrospinal fluid barrier (BCSFB). Overall, these data signify that T4 deiodinated by D2 may be the only source of T3 during neocortical development. We therefore propose that D2 activity at the BCSFB converts the T4 transported across the choroid plexus into T3, thus supplying the brain with active hormone to maintain TH homeostasis.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), E-28029 Madrid, Spain
- Department of Endocrine, U-708, Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Maria Jesus Obregon
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), E-28029 Madrid, Spain
| | - Juan Bernal
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), E-28029 Madrid, Spain
- Department of Endocrine, U-708, Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Ana Guadaño-Ferraz
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), E-28029 Madrid, Spain
- Department of Endocrine, U-708, Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
24
|
Groeneweg S, Peeters RP, Visser TJ, Visser WE. Therapeutic applications of thyroid hormone analogues in resistance to thyroid hormone (RTH) syndromes. Mol Cell Endocrinol 2017; 458:82-90. [PMID: 28235578 DOI: 10.1016/j.mce.2017.02.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/17/2017] [Accepted: 02/18/2017] [Indexed: 10/20/2022]
Abstract
Thyroid hormone (TH) is crucial for normal development and metabolism of virtually all tissues. TH signaling is predominantly mediated through binding of the bioactive hormone 3,3',5-triiodothyronine (T3) to the nuclear T3-receptors (TRs). The intracellular TH levels are importantly regulated by transporter proteins that facilitate the transport of TH across the cell membrane and by the three deiodinating enzymes. Defects at the level of the TRs, deiodinases and transporter proteins result in resistance to thyroid hormone (RTH) syndromes. Compounds with thyromimetic potency but with different (bio)chemical properties compared to T3 may hold therapeutic potential in these syndromes by bypassing defective transporters or binding to mutant TRs. Such TH analogues have the potential to rescue TH signaling. This review describes the role of TH analogues in the treatment of RTH syndromes. In particular, the application of 3,3',5-triiodothyroacetic acid (Triac) in RTH due to defective TRβ and the role of 3,5-diiodothyropropionic acid (DITPA), 3,3',5,5'-tetraiodothyroacetic acid (Tetrac) and Triac in MCT8 deficiency will be highlighted.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Theo J Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
25
|
Bárez-López S, Montero-Pedrazuela A, Bosch-García D, Venero C, Guadaño-Ferraz A. Increased anxiety and fear memory in adult mice lacking type 2 deiodinase. Psychoneuroendocrinology 2017; 84:51-60. [PMID: 28654773 DOI: 10.1016/j.psyneuen.2017.06.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/02/2017] [Accepted: 06/08/2017] [Indexed: 12/14/2022]
Abstract
A euthyroid state in the brain is crucial for its adequate development and function. Impairments in thyroid hormones (THs; T3 or 3,5,3'-triiodothyronine and T4 or thyroxine) levels and availability in brain can lead to neurological alterations and to psychiatric disorders, particularly mood disorders. The thyroid gland synthetizes mainly T4, which is secreted to circulating blood, however, most actions of THs are mediated by T3, the transcriptionally active form. In the brain, intracellular concentrations of T3 are modulated by the activity of type 2 (D2) and type 3 (D3) deiodinases. In the present work, we evaluated learning and memory capabilities and anxiety-like behavior at adult stages in mice lacking D2 (D2KO) and we analyzed the impact of D2-deficiency on TH content and on the expression of T3-dependent genes in the amygdala and the hippocampus. We found that D2KO mice do not present impairments in spatial learning and memory, but they display emotional alterations with increased anxiety-like behavior as well as enhanced auditory-cued fear memory and spontaneous recovery of fear memory following extinction. D2KO mice also presented reduced T3 content in the hippocampus and decreased expression of the T3-dependent gene Dio3 in the amygdala suggesting a hypothyroid status in this structure. We propose that the emotional dysfunctions found in D2KO mice can arise from the reduced T3 content in their brain, which consequently leads to alterations in gene expression with functional consequences. We found a downregulation in the gene encoding for the calcium-binding protein calretinin (Calb2) in the amygdala of D2KO mice that could affect the GABAergic transmission. The current findings in D2KO mice can provide insight into emotional disorders present in humans with DIO2 polymorphisms.
Collapse
Affiliation(s)
- Soledad Bárez-López
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols'', Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Arturo Duperier, 4, 28029, Madrid, Spain; Department of Endocrine, U-708, Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain.
| | - Ana Montero-Pedrazuela
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols'', Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Arturo Duperier, 4, 28029, Madrid, Spain.
| | - Daniel Bosch-García
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols'', Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Arturo Duperier, 4, 28029, Madrid, Spain.
| | - César Venero
- Department of Psychobiology, Universidad Nacional de Educación a Distancia, Juan del Rosal 10, 28040, Madrid, Spain.
| | - Ana Guadaño-Ferraz
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas "Alberto Sols'', Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Arturo Duperier, 4, 28029, Madrid, Spain; Department of Endocrine, U-708, Center for Biomedical Research on Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
26
|
Groeneweg S, Peeters RP, Visser TJ, Visser WE. Triiodothyroacetic acid in health and disease. J Endocrinol 2017; 234:R99-R121. [PMID: 28576869 DOI: 10.1530/joe-17-0113] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 12/16/2022]
Abstract
Thyroid hormone (TH) is crucial for development and metabolism of many tissues. The physiological relevance and therapeutic potential of TH analogs have gained attention in the field for many years. In particular, the relevance and use of 3,3',5-triiodothyroacetic acid (Triac, TA3) has been explored over the last decades. Although TA3 closely resembles the bioactive hormone T3, differences in transmembrane transport and receptor isoform-specific transcriptional activation potency exist. For these reasons, the application of TA3 as a treatment for resistance to TH (RTH) syndromes, especially MCT8 deficiency, is topic of ongoing research. This review is a summary of all currently available literature about the formation, metabolism, action and therapeutic applications of TA3.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine and Academic Center for Thyroid DiseasesErasmus University Medical Center, Rotterdam, The Netherlands
| | - Robin P Peeters
- Department of Internal Medicine and Academic Center for Thyroid DiseasesErasmus University Medical Center, Rotterdam, The Netherlands
| | - Theo J Visser
- Department of Internal Medicine and Academic Center for Thyroid DiseasesErasmus University Medical Center, Rotterdam, The Netherlands
| | - W Edward Visser
- Department of Internal Medicine and Academic Center for Thyroid DiseasesErasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
27
|
Tolcos M, Petratos S, Hirst JJ, Wong F, Spencer SJ, Azhan A, Emery B, Walker DW. Blocked, delayed, or obstructed: What causes poor white matter development in intrauterine growth restricted infants? Prog Neurobiol 2017; 154:62-77. [PMID: 28392287 DOI: 10.1016/j.pneurobio.2017.03.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 03/17/2017] [Accepted: 03/29/2017] [Indexed: 12/22/2022]
Abstract
Poor white matter development in intrauterine growth restricted (IUGR) babies remains a major, untreated problem in neonatology. New therapies, guided by an understanding of the mechanisms that underlie normal and abnormal oligodendrocyte development and myelin formation, are required. Much of our knowledge of the mechanisms that underlie impaired myelination come from studies in adult demyelinating disease, preterm brain injury, or experimental models of hypoxia-ischemia. However, relatively less is known for IUGR which is surprising because IUGR is a leading cause of perinatal mortality and morbidity, second only to premature birth. IUGR is also a significant risk factor for the later development of cerebral palsy, and is a greater risk compared to some of the more traditionally researched antecedents - asphyxia and inflammation. Recent evidence suggests that the white matter injury and reduced myelination in the brains of some preterm babies is due to impaired maturation of oligodendrocytes thereby resulting in the reduced capacity to synthesize myelin. Therefore, it is not surprising that the hypomyelination observable in the central nervous system of IUGR infants has similarly lead to investigations identifying a delay or blockade in the progress of maturation of oligodendrocytes in these infants. This review will discuss current ideas thought to account for the poor myelination often present in the neonate's brain following IUGR, and discuss novel interventions that are promising as treatments that promote oligodendrocyte maturation, and thereby repair the myelination deficits that otherwise persist into infancy and childhood and lead to neurodevelopmental abnormalities.
Collapse
Affiliation(s)
- Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia.
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
| | - Jonathan J Hirst
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Flora Wong
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia; Monash Newborn and Monash University, Clayton, Victoria, 3168, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Aminath Azhan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
| | - Ben Emery
- Oregon Health and Science University, Portland, OR, 97239-3098, USA
| | - David W Walker
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| |
Collapse
|
28
|
Affiliation(s)
- W Edward Visser
- 1 Department of Internal Medicine, Erasmus Medical Center , Rotterdam, The Netherlands
| | - Heike Heuer
- 2 Leibniz Research Institute for Environmental Medicine , Düsseldorf, Germany
| | - Theo J Visser
- 1 Department of Internal Medicine, Erasmus Medical Center , Rotterdam, The Netherlands
| |
Collapse
|