1
|
Li N, Zhang Z, Shen L, Song G, Tian J, Liu Q, Ni J. Selenium metabolism and selenoproteins function in brain and encephalopathy. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-023-2621-7. [PMID: 39546178 DOI: 10.1007/s11427-023-2621-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/09/2024] [Indexed: 11/17/2024]
Abstract
Selenium (Se) is an essential trace element of the utmost importance to human health. Its deficiency induces various disorders. Se species can be absorbed by organisms and metabolized to hydrogen selenide for the biosynthesis of selenoproteins, selenonucleic acids, or selenosugars. Se in mammals mainly acts as selenoproteins to exert their biological functions. The brain ranks highest in the specific hierarchy of organs to maintain the level of Se and the expression of selenoproteins under the circumstances of Se deficiency. Dyshomeostasis of Se and dysregulation of selenoproteins result in encephalopathy such as Alzheimer's disease, Parkinson's disease, depression, amyotrophic lateral sclerosis, and multiple sclerosis. This review provides a summary and discussion of Se metabolism, selenoprotein function, and their roles in modulating brain diseases based on the most currently published literature. It focuses on how Se is utilized and transported to the brain, how selenoproteins are biosynthesized and function physiologically in the brain, and how selenoproteins are involved in neurodegenerative diseases. At the end of this review, the perspectives and problems are outlined regarding Se and selenoproteins in the regulation of encephalopathy.
Collapse
Affiliation(s)
- Nan Li
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Zhonghao Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| | - Liming Shen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Guoli Song
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| | - Jing Tian
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China.
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China.
| | - Jiazuan Ni
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518055, China
| |
Collapse
|
2
|
Abd Elkader HTAE, Al-Shami AS, Darwish HS. Perinatal bisphenol A exposure has an age- and dose-dependent association with thyroid allostasis adaptive response, as well as anxiogenic-depressive-like and asocial behaviors in juvenile and adult male rats. Physiol Behav 2024; 288:114732. [PMID: 39510223 DOI: 10.1016/j.physbeh.2024.114732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
Thyroid hormones are essential for brain development, and a shortage throughout the fetal and postnatal periods can result in mood disorders. Perinatal exposure to bisphenol A (BPA) affects thyroid activity and dependent processes indirectly during pregnancy or early postnatal life. This is particularly important because it may cause changes in tissue ontogeny, increasing the risk of developing disorders later in life. The study aimed to investigate the consequences of thyroid hormone deficiency on anxiety, social, and depressive behaviors, as well as disruption in thyroid peroxidase (TPO) gene expression, which influences the NF-κB/Nrf-2/HO-1/iNOS signaling pathway, leading to oxidative stress, inflammation, and DNA fragmentation in perinatal BPA exposure (PND18), and whether these effects can be observed in juvenile (PND60) and adult (PND95) male offspring rats. BPA increased anxiety-like behavior while decreasing sucrose preference and sociability on a choice task between novel conspecific male rats and enhanced immobility on the forced swim test. Perinatal exposure to BPA causes thyroid insult by overproducing ROS, increasing iNOS, and NF-κB levels-these effects, in turn, down-regulate Nrf-2/HO-1 signaling, resulting in DNA fragmentation within thyroid tissues. Furthermore, perinatal BPA exposure for 60 and 95 days resulted in a significant fold decrease in TPO mRNA levels in the thyroid tissues, with an insignificant fold rise in TPO expression levels in BPA 50-60. In conclusion, the present study found that perinatal BPA exposure induced thyroid allostasis-adaptive response by inhibiting the NF-κB/Nrf-2/HO-1/iNOS signaling pathway and altering the transcriptional expression of TPO, where TSH reinforced a possible association with TPO activity, disrupting thyroid hormone synthesis in juvenile rats and gradual deterioration reaching the adult stage.
Collapse
Affiliation(s)
| | - Ahmed S Al-Shami
- Biotechnology Department, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Hanaa Said Darwish
- Zoology, Biological and Geological Sciences Department, Faculty of Education, Alexandria University, Alexandria, Egypt
| |
Collapse
|
3
|
Giatti S, Cioffi L, Diviccaro S, Piazza R, Melcangi RC. Analysis of the finasteride treatment and its withdrawal in the rat hypothalamus and hippocampus at whole-transcriptome level. J Endocrinol Invest 2024; 47:2565-2574. [PMID: 38493246 PMCID: PMC11393021 DOI: 10.1007/s40618-024-02345-y] [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: 11/23/2023] [Accepted: 02/18/2024] [Indexed: 03/18/2024]
Abstract
PURPOSE As reported in patients treated for androgenetic alopecia with finasteride (i.e., a blocker of the enzyme 5 alpha-reductase) and in an animal model, side effects affecting sexual, psychiatric, neurological, and physical domains, may occur during the treatment and persist with drug suspension. The etiopathogenesis of these side effects has been poorly explored. Therefore, we performed a genome-wide analysis of finasteride effects in the brain of adult male rat. METHODS Animals were treated (i.e., for 20 days) with finasteride (1mg/rat/day). 24 h after the last treatment and 1 month after drug suspension, RNA sequencing analysis was performed in hypothalamus and hippocampus. Data were analyzed by differential expression analysis and Gene-Set Enrichment Analyses (GSEA). RESULTS Data obtained after finasteride treatment showed that 186 genes (i.e., 171 up- and 15 downregulated) and 19 (i.e., 17 up- and 2 downregulated) were differentially expressed in the hypothalamus and hippocampus, respectively. Differential expression analysis at the drug withdrawal failed to identify dysregulated genes. Several gene-sets were enriched in these brain areas at both time points. CONCLUSION Some of the genes reported to be differentially expressed (i.e., TTR, DIO2, CLDN1, CLDN2, SLC4A5, KCNE2, CROT, HCRT, MARCKSL1, VGF, IRF2BPL) and GSEA, suggest a potential link with specific side effects previously observed in patients and in the animal model, such as depression, anxiety, disturbance in memory and attention, and sleep disturbance. These data may provide an important background for future experiments aimed at confirming the pathological role of these genes.
Collapse
Affiliation(s)
- S Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - L Cioffi
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - S Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - R Piazza
- Dipartimento di Medicina e Chirurgia, Università di Milano-Bicocca, Milan, Italy
| | - R C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
4
|
Valcárcel-Hernández V, Vancamp P, Butruille L, Remaud S, Guadaño-Ferraz A. Combined deletion of Mct8 and Dio2 impairs SVZ neurogliogenesis and olfactory function in adult mice. Neurobiol Dis 2024; 199:106572. [PMID: 38901782 DOI: 10.1016/j.nbd.2024.106572] [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: 02/15/2024] [Revised: 05/30/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024] Open
Abstract
Within the adult mouse subventricular zone (SVZ), neural stem cells (NSCs) produce neuroblasts and oligodendrocyte precursor cells (OPCs). T3, the active thyroid hormone, influences renewal and commitment of SVZ progenitors. However, how regulators of T3 availability affect these processes is less understood. Using Mct8/Dio2 knockout mice, we investigated the role of MCT8, a TH transporter, and DIO2, the T3-generating enzyme, in regulating adult SVZ-neurogliogenesis. Single-cell RNA-Seq revealed Mct8 expression in various SVZ cell types in WT mice, while Dio2 was enriched in neurons, astrocytes, and quiescent NSCs. The absence of both regulators in the knockout model dysregulated gene expression, increased the neuroblast/OPC ratio and hindered OPC differentiation. Immunostainings demonstrated compromised neuroblast migration reducing their supply to the olfactory bulbs, impairing interneuron differentiation and odor discrimination. These findings underscore the pivotal roles of MCT8 and DIO2 in neuro- and oligodendrogenesis, offering targets for therapeutic avenues in neurodegenerative and demyelinating diseases.
Collapse
Affiliation(s)
- Víctor Valcárcel-Hernández
- Laboratory of Thyroid hormones and CNS, Department of Neurological Diseases and Aging, Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Arturo Duperier 4, 28029 Madrid, Spain; Laboratory of Molecular Physiology and Adaptation, CNRS UMR 7221, Department Adaptations of Life, Muséum National d'Histoire Naturelle, F-75005 Paris, France
| | - Pieter Vancamp
- Laboratory of Molecular Physiology and Adaptation, CNRS UMR 7221, Department Adaptations of Life, Muséum National d'Histoire Naturelle, F-75005 Paris, France
| | - Lucile Butruille
- Laboratory of Molecular Physiology and Adaptation, CNRS UMR 7221, Department Adaptations of Life, Muséum National d'Histoire Naturelle, F-75005 Paris, France
| | - Sylvie Remaud
- Laboratory of Molecular Physiology and Adaptation, CNRS UMR 7221, Department Adaptations of Life, Muséum National d'Histoire Naturelle, F-75005 Paris, France.
| | - Ana Guadaño-Ferraz
- Laboratory of Thyroid hormones and CNS, Department of Neurological Diseases and Aging, Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Arturo Duperier 4, 28029 Madrid, Spain.
| |
Collapse
|
5
|
Mohácsik P, Halmos E, Dorogházi B, Ruska Y, Wittmann G, Bianco AC, Fekete C, Gereben B. The Musashi-1-type 2 deiodinase pathway regulates astrocyte proliferation. J Biol Chem 2024; 300:107477. [PMID: 38879014 PMCID: PMC11301063 DOI: 10.1016/j.jbc.2024.107477] [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: 01/12/2024] [Revised: 05/14/2024] [Accepted: 06/10/2024] [Indexed: 07/14/2024] Open
Abstract
Thyroid hormone (TH) is a critical regulator of cellular function and cell fate. The circulating TH level is relatively stable, while tissue TH action fluctuates according to cell type-specific mechanisms. Here, we focused on identifying mechanisms that regulate TH action through the type 2 deiodinase (D2) in glial cells. Dio2 mRNA has an unusually long 3'UTR where we identified multiple putative MSI1 binding sites for Musashi-1 (MSI1), a highly conserved RNA-binding cell cycle regulator. Binding to these sites was confirmed through electrophoretic mobility shift assay. In H4 glioma cells, shRNA-mediated MSI1 knockdown increased endogenous D2 activity, whereas MSI1 overexpression in HEK293T cells decreased D2 expression. This latter effect could be prevented by the deletion of a 3.6 kb region of the 3'UTR of Dio2 mRNA containing MSI1 binding sites. MSI1 immunoreactivity was observed in 2 mouse Dio2-expressing cell types, that is, cortical astrocytes and hypothalamic tanycytes, establishing the anatomical basis for a potential in vivo interaction of Dio2 mRNA and MSl1. Indeed, increased D2 expression was observed in the cortex of mice lacking MSI1 protein. Furthermore, MSI1 knockdown-induced D2 expression slowed down cell proliferation by 56% in primary cultures of mouse cortical astrocytes, establishing the functionality of the MSI1-D2-T3 pathway. In summary, Dio2 mRNA is a target of MSI1 and the MSI1-D2-T3 pathway is a novel regulatory mechanism of astrocyte proliferation with the potential to regulate the pathogenesis of human glioblastoma.
Collapse
Affiliation(s)
- Petra Mohácsik
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Emese Halmos
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Beáta Dorogházi
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Yvette Ruska
- Laboratory of Integrative Neuroendocrinology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Gábor Wittmann
- Laboratory of Integrative Neuroendocrinology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Antonio C Bianco
- Section of Adult and Pediatric Endocrinology and Metabolism, University of Chicago, Chicago, Illinois, USA
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
6
|
Zare Z, Shafia S, Mohammadi M. Thyroid hormone deficiency affects anxiety-related behaviors and expression of hippocampal glutamate transporters in male congenital hypothyroid rat offspring. Horm Behav 2024; 162:105548. [PMID: 38636205 DOI: 10.1016/j.yhbeh.2024.105548] [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: 08/19/2023] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Thyroid hormones are crucial for brain development and their deficiency during fetal and postnatal periods can lead to mood and cognitive disorders. We aimed to examine the consequences of thyroid hormone deficiency on anxiety-related behaviors and protein expression of hippocampal glutamate transporters in congenital hypothyroid male offspring rats. Possible beneficial effects of treadmill exercise have also been examined. Congenital hypothyroidism was induced by adding propylthiouracil (PTU) to drinking water of pregnant Wistar rats from gestational day 6 until the end of the weaning period (postnatal day 28). Next, following 4 weeks of treadmill exercise (5 days per week), anxiety-related behaviors were examined using elevated plus maze (EPM) and light/dark box tests. Thereafter, protein expression of astrocytic (GLAST and GLT-1) and neuronal (EAAC1) glutamate transporters were measured in the hippocampus by immunoblotting. Hypothyroid rats showed decreased anxiety-like behavior, as measured by longer time spent in the open arms of the EPM and in the light area of the light/dark box, compared to control rats. Hypothyroid rats had significantly higher GLAST and GLT-1 and lower EAAC1 protein levels in the hippocampus than did the euthyroid rats. Following exercise, anxiety levels decreased in the euthyroid group while protein expression of EAAC1 increased and returned to normal levels in the hypothyroid group. Our findings indicate that thyroid hormone deficiency was associated with alterations in protein expression of glutamate transporters in the hippocampus. Up-regulation of hippocampal GLAST and GLT-1 could be at least one of the mechanisms associated with the anxiolytic effects of congenital hypothyroidism.
Collapse
Affiliation(s)
- Zohreh Zare
- Department of Anatomical Sciences, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sakineh Shafia
- Department of Physiology, Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Moslem Mohammadi
- Department of Physiology, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
7
|
Hindrichs C, Walk T, Melching-Kollmuss S, Landsiedel R, Kamp H, Funk-Weyer D. A Novel and Fast Online-SPE-LC-MS/MS Method to Quantify Thyroid Hormone Metabolites in Rat Plasma. Chem Res Toxicol 2024; 37:33-41. [PMID: 38078760 DOI: 10.1021/acs.chemrestox.3c00152] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Since the focus in regulatory toxicology has drifted toward the identification of endocrine disruptors, the improvement in determination of alterations in the thyroid hormone system has become more important. THs are involved in several molecular processes important for a proper pre- and postnatal development so that disturbances can inter alia lead to incorrect brain maturation and/or disturbed metabolic processes (thermogenesis or lipolysis). In this publication, a new automated online solid-phase extraction (SPE)-liquid chromatography (LC)-tandem mass spectrometry (MS/MS, xLC-MS/MS) is introduced which simultaneously analyzes total T4, T3, rT3, T2, and T1. Method validation parameters are presented, and the method was positively verified by analyzing control and PTU-treated rat plasma samples (time points day 7, 14, and 28) for their total TH content. The obtained results were compared to published results by using a radioimmunoassay method. The automated SPE system ensures a consistent unified sample preparation, and this method overall showed sufficient specificity and accuracy to detect the given analytes in rat plasma. For the preparation of 50 μL of rat plasma, the following LOQs were established: 0.020 nM for T1, 0.029 nM for T2, 0.023 nM for rT3 and T3, and 3.22 nM for T4. This method is suitable to assess the identification of mechanisms leading to adverse effects, such as disturbed TH metabolism and regulation.
Collapse
Affiliation(s)
- Christiane Hindrichs
- BASF Metabolome Solutions GmbH, 10589 Berlin, Germany
- Rheinland-Pfälzischen Technischen Universität Kaiserslautern-Landau, 67663 Kaiserslautern, Germany
| | - Tilmann Walk
- BASF Metabolome Solutions GmbH, 10589 Berlin, Germany
| | | | - Robert Landsiedel
- Experimental Toxicology and Ecology 10, BASF SE, 67056 Ludwigshafen am Rhein, Germany
- Free University of Berlin, Pharmacy, Pharmacology and Toxicology, 14195 Berlin, Germany
| | - Hennicke Kamp
- BASF Metabolome Solutions GmbH, 10589 Berlin, Germany
| | - Dorothee Funk-Weyer
- Experimental Toxicology and Ecology 10, BASF SE, 67056 Ludwigshafen am Rhein, Germany
| |
Collapse
|
8
|
Haigis AC, Vergauwen L, LaLone CA, Villeneuve DL, O'Brien JM, Knapen D. Cross-species applicability of an adverse outcome pathway network for thyroid hormone system disruption. Toxicol Sci 2023; 195:1-27. [PMID: 37405877 DOI: 10.1093/toxsci/kfad063] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023] Open
Abstract
Thyroid hormone system disrupting compounds are considered potential threats for human and environmental health. Multiple adverse outcome pathways (AOPs) for thyroid hormone system disruption (THSD) are being developed in different taxa. Combining these AOPs results in a cross-species AOP network for THSD which may provide an evidence-based foundation for extrapolating THSD data across vertebrate species and bridging the gap between human and environmental health. This review aimed to advance the description of the taxonomic domain of applicability (tDOA) in the network to improve its utility for cross-species extrapolation. We focused on the molecular initiating events (MIEs) and adverse outcomes (AOs) and evaluated both their plausible domain of applicability (taxa they are likely applicable to) and empirical domain of applicability (where evidence for applicability to various taxa exists) in a THSD context. The evaluation showed that all MIEs in the AOP network are applicable to mammals. With some exceptions, there was evidence of structural conservation across vertebrate taxa and especially for fish and amphibians, and to a lesser extent for birds, empirical evidence was found. Current evidence supports the applicability of impaired neurodevelopment, neurosensory development (eg, vision) and reproduction across vertebrate taxa. The results of this tDOA evaluation are summarized in a conceptual AOP network that helps prioritize (parts of) AOPs for a more detailed evaluation. In conclusion, this review advances the tDOA description of an existing THSD AOP network and serves as a catalog summarizing plausible and empirical evidence on which future cross-species AOP development and tDOA assessment could build.
Collapse
Affiliation(s)
- Ann-Cathrin Haigis
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Lucia Vergauwen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Carlie A LaLone
- Great Lakes Toxicology and Ecology Division, United States Environmental Protection Agency, Duluth, Minnesota 55804, USA
| | - Daniel L Villeneuve
- Great Lakes Toxicology and Ecology Division, United States Environmental Protection Agency, Duluth, Minnesota 55804, USA
| | - Jason M O'Brien
- Ecotoxicology and Wildlife Health Division, Environment and Climate Change Canada, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Dries Knapen
- Zebrafishlab, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
9
|
Salas-Lucia F, Fekete C, Sinkó R, Egri P, Rada K, Ruska Y, Gereben B, Bianco AC. Axonal T3 uptake and transport can trigger thyroid hormone signaling in the brain. eLife 2023; 12:e82683. [PMID: 37204837 PMCID: PMC10241515 DOI: 10.7554/elife.82683] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 05/18/2023] [Indexed: 05/20/2023] Open
Abstract
The development of the brain, as well as mood and cognitive functions, are affected by thyroid hormone (TH) signaling. Neurons are the critical cellular target for TH action, with T3 regulating the expression of important neuronal gene sets. However, the steps involved in T3 signaling remain poorly known given that neurons express high levels of type 3 deiodinase (D3), which inactivates both T4 and T3. To investigate this mechanism, we used a compartmentalized microfluid device and identified a novel neuronal pathway of T3 transport and action that involves axonal T3 uptake into clathrin-dependent, endosomal/non-degradative lysosomes (NDLs). NDLs-containing T3 are retrogradely transported via microtubules, delivering T3 to the cell nucleus, and doubling the expression of a T3-responsive reporter gene. The NDLs also contain the monocarboxylate transporter 8 (Mct8) and D3, which transport and inactivate T3, respectively. Notwithstanding, T3 gets away from degradation because D3's active center is in the cytosol. Moreover, we used a unique mouse system to show that T3 implanted in specific brain areas can trigger selective signaling in distant locations, as far as the contralateral hemisphere. These findings provide a pathway for L-T3 to reach neurons and resolve the paradox of T3 signaling in the brain amid high D3 activity.
Collapse
Affiliation(s)
- Federico Salas-Lucia
- Section of Adult and Pediatric Endocrinology and Metabolism, University of ChicagoChicagoUnited States
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental MedicineBudapestHungary
| | - Richárd Sinkó
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
- János Szentágothai PhD School of Neurosciences, Semmelweis UniversityBudapestHungary
| | - Péter Egri
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
| | - Kristóf Rada
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
| | - Yvette Ruska
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental MedicineBudapestHungary
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
| | - Antonio C Bianco
- Section of Adult and Pediatric Endocrinology and Metabolism, University of ChicagoChicagoUnited States
| |
Collapse
|
10
|
Deng Y, Han Y, Gao S, Dong W, Yu Y. The Physiological Functions and Polymorphisms of Type II Deiodinase. Endocrinol Metab (Seoul) 2023; 38:190-202. [PMID: 37150515 PMCID: PMC10164501 DOI: 10.3803/enm.2022.1599] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/21/2023] [Indexed: 05/09/2023] Open
Abstract
Type II deiodinase (DIO2) is thought to provide triiodothyronine (T3) to the nucleus to meet intracellular needs by deiodinating the prohormone thyroxine. DIO2 is expressed widely in many tissues and plays an important role in a variety of physiological processes, such as controlling T3 content in developing tissues (e.g., bone, muscles, and skin) and the adult brain, and regulating adaptive thermogenesis in brown adipose tissue (BAT). However, the identification and cloning of DIO2 have been challenging. In recent years, several clinical investigations have focused on the Thr92Ala polymorphism, which is closely correlated with clinical syndromes such as type 2 diabetes, obesity, hypertension, and osteoarthritis. Thr92Ala-DIO2 was also found to be related to bone and neurodegenerative diseases and tumors. However, relatively few reviews have synthesized research on individual deiodinases, especially DIO2, in the past 5 years. This review summarizes current knowledge regarding the physiological functions of DIO2 in thyroid hormone signaling and adaptive thermogenesis in BAT and the brain, as well as the associations between Thr92Ala-DIO2 and bone and neurodegenerative diseases and tumors. This discussion is expected to provide insights into the physiological functions of DIO2 and the clinical syndromes associated with Thr92Ala-DIO2.
Collapse
Affiliation(s)
- Yan Deng
- Department of Histology and Embryology, School of Basic Medical Sciences, Southwest Medical University, China
| | - Yi Han
- Department of Histology and Embryology, School of Basic Medical Sciences, Southwest Medical University, China
| | - Sheng Gao
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yang Yu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southwest Medical University, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Abstract
In this article, starting with the recognition that iodine is essential for normal thyroid function and is a component of thyroid hormone (TH) molecules, we discuss the many seminal observations and discoveries that have led to identification of various pathways of TH metabolism and their potential roles in TH economy and action. We then recount evidence that TH metabolism participates in maintaining the appropriate content of active hormone in a TH-responsive tissue or cell. Thus, metabolism of the TH is not merely a means by which it is degraded and eliminated from the body, but an essential component of an intricate system by which the thyroid exerts its multiple regulatory effects on almost all organs and tissues. The article ends with a summary of the current concepts and some outstanding questions that are awaiting answers.
Collapse
Affiliation(s)
- Valerie Anne Galton
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Arturo Hernandez
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| |
Collapse
|
12
|
Murolo M, Di Vincenzo O, Cicatiello AG, Scalfi L, Dentice M. Cardiovascular and Neuronal Consequences of Thyroid Hormones Alterations in the Ischemic Stroke. Metabolites 2022; 13:metabo13010022. [PMID: 36676947 PMCID: PMC9863748 DOI: 10.3390/metabo13010022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Ischemic stroke is one of the leading global causes of neurological morbidity and decease. Its etiology depends on multiple events such as cardiac embolism, brain capillaries occlusion and atherosclerosis, which ultimately culminate in blood flow interruption, incurring hypoxia and nutrient deprivation. Thyroid hormones (THs) are pleiotropic modulators of several metabolic pathways, and critically influence different aspects of tissues development. The brain is a key TH target tissue and both hypo- and hyperthyroidism, during embryonic and adult life, are associated with deranged neuronal formation and cognitive functions. Accordingly, increasing pieces of evidence are drawing attention on the consistent relationship between the THs status and the acute cerebral and cardiac diseases. However, the concrete contribution of THs systemic or local alteration to the pathology outcome still needs to be fully addressed. In this review, we aim to summarize the multiple influences that THs exert on the brain and heart patho-physiology, to deepen the reasons for the harmful effects of hypo- and hyperthyroidism on these organs and to provide insights on the intricate relationship between the THs variations and the pathological alterations that take place after the ischemic injury.
Collapse
Affiliation(s)
- Melania Murolo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
- Correspondence:
| | - Olivia Di Vincenzo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy
| | | | - Luca Scalfi
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
- CEINGE-Biotecnologie Avanzate S.c.a.r.l., 80131 Naples, Italy
| |
Collapse
|
13
|
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
|
14
|
Jehan C, Cartier D, Bucharles C, Anouar Y, Lihrmann I. Emerging roles of ER-resident selenoproteins in brain physiology and physiopathology. Redox Biol 2022; 55:102412. [PMID: 35917681 PMCID: PMC9344019 DOI: 10.1016/j.redox.2022.102412] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/05/2022] [Accepted: 07/14/2022] [Indexed: 12/23/2022] Open
Abstract
The brain has a very high oxygen consumption rate and is particularly sensitive to oxidative stress. It is also the last organ to suffer from a loss of selenium (Se) in case of deficiency. Se is a crucial trace element present in the form of selenocysteine, the 21st proteinogenic amino acid present in selenoproteins, an essential protein family in the brain that participates in redox signaling. Among the most abundant selenoproteins in the brain are glutathione peroxidase 4 (GPX4), which reduces lipid peroxides and prevents ferroptosis, and selenoproteins W, I, F, K, M, O and T. Remarkably, more than half of them are proteins present in the ER and recent studies have shown their involvement in the maintenance of ER homeostasis, glycoprotein folding and quality control, redox balance, ER stress response signaling pathways and Ca2+ homeostasis. However, their molecular functions remain mostly undetermined. The ER is a highly specialized organelle in neurons that maintains the physical continuity of axons over long distances through its continuous distribution from the cell body to the nerve terminals. Alteration of this continuity can lead to degeneration of distal axons and subsequent neuronal death. Elucidation of the function of ER-resident selenoproteins in neuronal pathophysiology may therefore become a new perspective for understanding the pathophysiology of neurological diseases. Here we summarize what is currently known about each of their molecular functions and their impact on the nervous system during development and stress.
Collapse
Affiliation(s)
- Cédric Jehan
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Dorthe Cartier
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Christine Bucharles
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Youssef Anouar
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Isabelle Lihrmann
- Rouen-Normandie University, UNIROUEN, Inserm, U1239, Neuroendocrine, Endocrine and Germinal Differenciation and Communication Laboratory, Mont-Saint-Aignan Cedex, France; Institute for Research and Innovation in Biomedicine, Rouen, France.
| |
Collapse
|
15
|
Gao Y, Zhao L, Son JS, Liu X, Chen Y, Deavila JM, Zhu MJ, Murdoch GK, Du M. Maternal Exercise Before and During Pregnancy Facilitates Embryonic Myogenesis by Enhancing Thyroid Hormone Signaling. Thyroid 2022; 32:581-593. [PMID: 35286177 PMCID: PMC9145266 DOI: 10.1089/thy.2021.0639] [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: 11/13/2022]
Abstract
Background: Maternal exercise (ME) improves fetal and offspring muscle development, but mechanisms remain to be established. Since the thyroid hormone (TH) is critical for cell differentiation during embryonic development, we hypothesized that ME elevates TH receptor (THR) signaling in embryos, which promotes embryonic myogenesis. Methods: Female mice were exercised daily on a treadmill or received a daily TH, triiodothyronine (T3) injection. Embryos (embryonic day 12.5 [E12.5]) and P19 cells were used for studying effects of TH on embryonic myogenesis. TH levels in serum and embryos after ME or T3I were analyzed. Expression of TH signaling related genes and myogenic genes was assessed. THRα binding to the promoters of myogenic genes was investigated by chromatin immunoprecipitation-qantitative polymerase chain reaction (ChIP-qPCR). A CRISPR/CAS9 plasmid was utilized to knock out THRα in P19 cells. Results: ME elevated TH levels in both maternal circulation and embryos, which were correlated with enhanced TH signaling and myogenesis. At E12.5, both myogenic determinants (Pax3, Pax7) and myogenic regulatory factors (Myf5, Myod) were upregulated in ME embryos. ME increased THRα content and elevated messenger RNA (mRNA) expression of TH transporter Slc16a2 and deiodinase Dio2. In addition, the THRα binding to the promoters of Pax3/7 was increased. In P19 embryoid bodies, T3 promoted myogenic differentiation, which was abolished by ablating THRα. Furthermore, maternal daily injection of T3 at a level matching exercised mothers promoted embryonic myogenesis. Conclusions: ME promotes TH delivery to the embryos and enhances embryonic myogenesis, which is partially mediated by enhanced TH signaling in ME embryos.
Collapse
Affiliation(s)
- Yao Gao
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Liang Zhao
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Jun Seok Son
- Laboratory of Perinatal Kinesioepigenetics, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xiangdong Liu
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Yanting Chen
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Jeanene Marie Deavila
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Mei-Jun Zhu
- Food Microbiology and Nutrigenomics Laboratory, School of Food Science, Washington State University, Pullman, Washington, USA
| | - Gordon K. Murdoch
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, Washington, USA
| | - Min Du
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, Washington, USA
- Address correspondence to: Min Du, PhD, Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
16
|
Zamar A, Lulsegged A, Kouimtsidis C. A new approach for the treatment of subthreshold bipolar disorders: Targeted high dose levothyroxine and repetitive transcranial magnetic stimulation for mitochondrial treatment. Front Psychiatry 2022; 13:976544. [PMID: 36311500 PMCID: PMC9606762 DOI: 10.3389/fpsyt.2022.976544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/16/2022] [Indexed: 11/18/2022] Open
Abstract
Bipolar spectrum disorder includes Bipolar I, Bipolar II and subthreshold bipolar disorders (BD). The condition is highly prevalent, disabling and associated with high mortality. Failure of diagnosis is high. Subthreshold presentations present as 4 or more changes in polarity, are generally less responsive to standard treatment and as a result, drug combinations are often needed. High Dose Levothyroxine (HDT) has been reported to be safe and effective with this condition. Treatment response has been associated with mutations in thyroid activating enzymes and intra cerebral transporter protein carrier. Repetitive Transcranial Magnetic Stimulation (rTMS) has been shown to be effective in bipolar depression and has been proved to have neuroplastic effect. Present authors had reported clinical evidence of safe and effective use of a combination treatment protocol. Potential mechanisms of action of the combined treatment protocol and the role of mitochondria function are discussed.
Collapse
Affiliation(s)
- Andy Zamar
- Consultant Psychiatrist, The London Psychiatry Centre, London, United Kingdom
| | - Abbi Lulsegged
- Consultant Endocrinologist Health 121 Ltd., London, United Kingdom
| | - Christos Kouimtsidis
- Consultant Psychiatrist, The London Psychiatry Centre, London, United Kingdom.,Honorary Senior Lecturer Imperial College London, London, United Kingdom
| |
Collapse
|
17
|
Sabatino L, Vassalle C, Del Seppia C, Iervasi G. Deiodinases and the Three Types of Thyroid Hormone Deiodination Reactions. Endocrinol Metab (Seoul) 2021; 36:952-964. [PMID: 34674502 PMCID: PMC8566136 DOI: 10.3803/enm.2021.1198] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/01/2021] [Accepted: 09/15/2021] [Indexed: 12/27/2022] Open
Abstract
Thyroid hormone (TH) signaling is strictly regulated by iodothyronine deiodinase activity, which both preserves the circulating levels of the biologically active triiodothyronine (T3) and regulates TH homeostasis at the local level, in a cell- and time-dependent manner. Three deiodinases have been identified-namely iodothyronine deiodinase 1 (DIO1), DIO2, and DIO3-that differ in their catalytic properties and tissue distribution. The deiodinases represent a dynamic system that changes in the different stages of life according to their functions and roles in various cell types and tissues. Deiodinase activity at the tissue level permits cell-targeted fine regulation of TH homeostasis, mediating the activation (DIO1 and DIO2) and inactivation (DIO3) of THs. Deiodinase homeostasis is the driving force that leads T3-target cells towards customized TH signaling, which takes into account both the hormonal circulating levels and the tissue-specific response. This review analyzes the complex role of deiodinases in physiological and pathological contexts, exploring new challenges and opportunities deriving from a deeper knowledge of the dynamics underlying their roles and functions.
Collapse
Affiliation(s)
- Laura Sabatino
- Institute of Clinical Physiology, National Research Council of Italy (CNR), Pisa,
Italy
| | | | - Cristina Del Seppia
- Institute of Clinical Physiology, National Research Council of Italy (CNR), Pisa,
Italy
| | - Giorgio Iervasi
- Institute of Clinical Physiology, National Research Council of Italy (CNR), Pisa,
Italy
| |
Collapse
|
18
|
Hernandez A, Martinez ME, Ng L, Forrest D. Thyroid Hormone Deiodinases: Dynamic Switches in Developmental Transitions. Endocrinology 2021; 162:bqab091. [PMID: 33963379 PMCID: PMC8248586 DOI: 10.1210/endocr/bqab091] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Indexed: 12/15/2022]
Abstract
Thyroid hormones exert pleiotropic, essential actions in mammalian, including human, development. These actions depend on provision of thyroid hormones in the circulation but also to a remarkable extent on deiodinase enzymes in target tissues that amplify or deplete the local concentration of the primary active form of the hormone T3 (3,5,3'-triiodothyronine), the high affinity ligand for thyroid hormone receptors. Genetic analyses in mice have revealed key roles for activating (DIO2) and inactivating (DIO3) deiodinases in cell differentiation fates and tissue maturation, ultimately promoting neonatal viability, growth, fertility, brain development, and behavior, as well as metabolic, endocrine, and sensory functions. An emerging paradigm is how the opposing activities of DIO2 and DIO3 are coordinated, providing a dynamic switch that controls the developmental timing of a tissue response, often during neonatal and maturational transitions. A second paradigm is how cell to cell communication within a tissue determines the response to T3. Deiodinases in specific cell types, often strategically located near to blood vessels that convey thyroid hormones into the tissue, can regulate neighboring cell types, suggesting a paracrine-like layer of control of T3 action. We discuss deiodinases as switches for developmental transitions and their potential to influence tissue dysfunction in human thyroid disorders.
Collapse
Affiliation(s)
- Arturo Hernandez
- Department of Molecular Medicine, Maine Medical Center Research Institute, Maine Health, Scarborough, Maine 04074, USA
- Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine 04469, USA
| | - M Elena Martinez
- Department of Molecular Medicine, Maine Medical Center Research Institute, Maine Health, Scarborough, Maine 04074, USA
| | - Lily Ng
- National Institute of Diabetes and Digestive and Kidney Diseases, Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Douglas Forrest
- National Institute of Diabetes and Digestive and Kidney Diseases, Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
19
|
Ishii S, Amano I, Koibuchi N. The Role of Thyroid Hormone in the Regulation of Cerebellar Development. Endocrinol Metab (Seoul) 2021; 36:703-716. [PMID: 34365775 PMCID: PMC8419606 DOI: 10.3803/enm.2021.1150] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022] Open
Abstract
The proper organized expression of specific genes in time and space is responsible for the organogenesis of the central nervous system including the cerebellum. The epigenetic regulation of gene expression is tightly regulated by an intrinsic intracellular genetic program, local stimuli such as synaptic inputs and trophic factors, and peripheral stimuli from outside of the brain including hormones. Some hormone receptors are expressed in the cerebellum. Thyroid hormones (THs), among numerous circulating hormones, are well-known major regulators of cerebellar development. In both rodents and human, hypothyroidism during the postnatal developmental period results in abnormal morphogenesis or altered function. THs bind to the thyroid hormone receptors (TRs) in the nuclei and with the help of transcriptional cofactors regulate the transcription of target genes. Gene regulation by TR induces cell proliferation, migration, and differentiation, which are necessary for brain development and plasticity. Thus, the lack of TH action mediators may directly cause aberrant cerebellar development. Various kinds of animal models have been established in a bid to study the mechanism of TH action in the cerebellum. Interestingly, the phenotypes differ greatly depending on the models. Herein we summarize the actions of TH and TR particularly in the developing cerebellum.
Collapse
Affiliation(s)
- Sumiyasu Ishii
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Izuki Amano
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
20
|
Galton VA, Martinez ME, Dragon JA, St Germain DL, Hernandez A. The Intrinsic Activity of Thyroxine Is Critical for Survival and Growth and Regulates Gene Expression in Neonatal Liver. Thyroid 2021; 31:528-541. [PMID: 32791885 PMCID: PMC7994419 DOI: 10.1089/thy.2020.0508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: Thyroxine (T4) is generally considered to be a prohormone that requires conversion to triiodothyronine (T3) to exert biological activity. Although evidence suggests that T4 has intrinsic activity, it is questionable if this activity has any physiological relevance. Methods: To answer this question, triple knockout (KO) mice (Triples) that cannot express the types 1 (D1) and 2 (D2) deiodinase and the Pax8 genes were generated. Thus, they lack a thyroid and cannot convert T4 to T3. Triples were injected on alternate days with either vehicle or physiological doses of T4, T3, or T3+T4 from postnatal days 2-14. They were euthanized at P15, and RNA-seq was employed to profile gene expression in the liver. In another experiment, Pax8KO mice were injected with T3, T4, or T4+T3, and growth rate and survival to P84 were determined. Results: The growth retardation of Triples was not improved by either T3 or T4 alone but was significantly improved by T4+T3. In the liver, T4 significantly regulated the expression of genes that were also regulated by T3, but the proportion of genes that were negatively regulated was higher in mice treated with T4 than in mice treated with T3. Treatment with T4+T3 identified genes that were regulated synergistically by T3 and T4, and genes that were regulated only by T4+T3. Analysis of these genes revealed enrichment in mechanisms related to cell proliferation and cholesterol physiology, suggesting a unique contribution of T4 to these biological functions. Pax8KO mice all survived to P84 when injected with T4 or T4+T3. However, survival rate with T3 was only 50% and 10% at 3.5 and 12 weeks of life, respectively. Conclusions: T4 has intrinsic activity in vivo and is critical for survival and growth. At a physiological level, T4 per se can upregulate or downregulate many T3 target genes in the neonatal liver. While most of these genes are also regulated by T3, subsets respond exclusively to T4 or demonstrate enhanced or normalized expression only in the presence of both hormones. These studies demonstrate for the first time a complex dependency on both T4 and T3 for normal mammalian growth and development.
Collapse
Affiliation(s)
- Valerie Anne Galton
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | | | - Julie A. Dragon
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | | | - Arturo Hernandez
- Maine Medical Center Research Institute, MaineHealth, Scarborough, Maine, USA
- Department of Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| |
Collapse
|
21
|
Gilbert ME, O'Shaughnessy KL, Axelstad M. Regulation of Thyroid-disrupting Chemicals to Protect the Developing Brain. Endocrinology 2020; 161:bqaa106. [PMID: 32615585 PMCID: PMC8650774 DOI: 10.1210/endocr/bqaa106] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022]
Abstract
Synthetic chemicals with endocrine disrupting properties are pervasive in the environment and are present in the bodies of humans and wildlife. As thyroid hormones (THs) control normal brain development, and maternal hypothyroxinemia is associated with neurological impairments in children, chemicals that interfere with TH signaling are of considerable concern for children's health. However, identifying thyroid-disrupting chemicals (TDCs) in vivo is largely based on measuring serum tetraiodothyronine in rats, which may be inadequate to assess TDCs with disparate mechanisms of action and insufficient to evaluate the potential neurotoxicity of TDCs. In this review 2 neurodevelopmental processes that are dependent on TH action are highlighted, neuronal migration and maturation of gamma amino butyric acid-ergic interneurons. We discuss how interruption of these processes by TDCs may contribute to abnormal brain circuitry following developmental TH insufficiency. Finally, we identify issues in evaluating the developmental neurotoxicity of TDCs and the strengths and limitations of current approaches designed to regulate them. It is clear that an enhanced understanding of how THs affect brain development will lead to refined toxicity testing, reducing uncertainty and improving our ability to protect children's health.
Collapse
Affiliation(s)
- Mary E Gilbert
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Katherine L O'Shaughnessy
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Marta Axelstad
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
22
|
Leonardi A, Evke S, Lee M, Melendez JA, Begley TJ. Epitranscriptomic systems regulate the translation of reactive oxygen species detoxifying and disease linked selenoproteins. Free Radic Biol Med 2019; 143:573-593. [PMID: 31476365 PMCID: PMC7650020 DOI: 10.1016/j.freeradbiomed.2019.08.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Here we highlight the role of epitranscriptomic systems in post-transcriptional regulation, with a specific focus on RNA modifying writers required for the incorporation of the 21st amino acid selenocysteine during translation, and the pathologies linked to epitranscriptomic and selenoprotein defects. Epitranscriptomic marks in the form of enzyme-catalyzed modifications to RNA have been shown to be important signals regulating translation, with defects linked to altered development, intellectual impairment, and cancer. Modifications to rRNA, mRNA and tRNA can affect their structure and function, while the levels of these dynamic tRNA-specific epitranscriptomic marks are stress-regulated to control translation. The tRNA for selenocysteine contains five distinct epitranscriptomic marks and the ALKBH8 writer for the wobble uridine (U) has been shown to be vital for the translation of the glutathione peroxidase (GPX) and thioredoxin reductase (TRXR) family of selenoproteins. The reactive oxygen species (ROS) detoxifying selenocysteine containing proteins are a prime examples of how specialized translation can be regulated by specific tRNA modifications working in conjunction with distinct codon usage patterns, RNA binding proteins and specific 3' untranslated region (UTR) signals. We highlight the important role of selenoproteins in detoxifying ROS and provide details on how epitranscriptomic marks and selenoproteins can play key roles in and maintaining mitochondrial function and preventing disease.
Collapse
Affiliation(s)
- Andrea Leonardi
- Colleges of Nanoscale Science and Engineering, University at Albany, State University of New York, Albany, NY, USA
| | - Sara Evke
- Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY, USA
| | - May Lee
- Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY, USA
| | - J Andres Melendez
- Colleges of Nanoscale Science and Engineering, State University of New York Polytechnic Institute, Albany, NY, USA.
| | - Thomas J Begley
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA; RNA Institute, University at Albany, State University of New York, Albany, NY, USA.
| |
Collapse
|
23
|
Bárez-López S, Grijota-Martínez C, Ausó E, Fernández-de Frutos M, Montero-Pedrazuela A, Guadaño-Ferraz A. Adult Mice Lacking Mct8 and Dio2 Proteins Present Alterations in Peripheral Thyroid Hormone Levels and Severe Brain and Motor Skill Impairments. Thyroid 2019; 29:1669-1682. [PMID: 31359845 DOI: 10.1089/thy.2019.0068] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Mutations in the thyroid hormone (TH) transporter monocarboxylate transporter 8 (MCT8) lead to peripheral hyperthyroidism and profound psychomotor alterations in humans. Mice lacking Mct8 present peripheral hyperthyroidism but no gross neurological abnormalities due to brain compensatory mechanisms involving the enzyme deiodinase type 2 (Dio2). Methods: Here we have analyzed the endocrine and neurologic phenotype of mice lacking both Mct8 and Dio2 at three and six months of age. Thyroxine (T4) and 3,5,3' triiodothyronine (T3) levels/content were measured by specific radioimmunoassays; motor skill performance was evaluated by the footprint, rotarod, four limb hanging wire, and balance beam tests; and brain histological analysis was performed by immunostaining for neurofilament and parvalbumin. Results: We have found that this mouse model presents peripheral hyperthyroidism and brain hypothyroidism. Interestingly, the severity of the brain hypothyroidism seems permanent and varies across regions, with the striatum being a particularly affected area. We have also found brain alterations at the histological level compatible with TH deficiency and impaired motor skills. Conclusions: These findings indicate the potential of Mct8/Dio2-deficient mice to represent a model for human MCT8 deficiency, to understand the mechanisms underlying its pathophysiology, and ultimately design therapeutic interventions for human patients.
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
- Unit 708, Center for Biomedical Research On Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Grijota-Martínez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Unit 708, Center for Biomedical Research On Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
| | - Eva Ausó
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Departamento de Óptica, Farmacología y Anatomía, Universidad de Alicante, Alicante, Spain
| | - Mario Fernández-de Frutos
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Montero-Pedrazuela
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Guadaño-Ferraz
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Unit 708, Center for Biomedical Research On Rare Diseases (Ciberer), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
24
|
Noyes PD, Friedman KP, Browne P, Haselman JT, Gilbert ME, Hornung MW, Barone S, Crofton KM, Laws SC, Stoker TE, Simmons SO, Tietge JE, Degitz SJ. Evaluating Chemicals for Thyroid Disruption: Opportunities and Challenges with in Vitro Testing and Adverse Outcome Pathway Approaches. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:95001. [PMID: 31487205 PMCID: PMC6791490 DOI: 10.1289/ehp5297] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/01/2019] [Accepted: 08/13/2019] [Indexed: 05/19/2023]
Abstract
BACKGROUND Extensive clinical and experimental research documents the potential for chemical disruption of thyroid hormone (TH) signaling through multiple molecular targets. Perturbation of TH signaling can lead to abnormal brain development, cognitive impairments, and other adverse outcomes in humans and wildlife. To increase chemical safety screening efficiency and reduce vertebrate animal testing, in vitro assays that identify chemical interactions with molecular targets of the thyroid system have been developed and implemented. OBJECTIVES We present an adverse outcome pathway (AOP) network to link data derived from in vitro assays that measure chemical interactions with thyroid molecular targets to downstream events and adverse outcomes traditionally derived from in vivo testing. We examine the role of new in vitro technologies, in the context of the AOP network, in facilitating consideration of several important regulatory and biological challenges in characterizing chemicals that exert effects through a thyroid mechanism. DISCUSSION There is a substantial body of knowledge describing chemical effects on molecular and physiological regulation of TH signaling and associated adverse outcomes. Until recently, few alternative nonanimal assays were available to interrogate chemical effects on TH signaling. With the development of these new tools, screening large libraries of chemicals for interactions with molecular targets of the thyroid is now possible. Measuring early chemical interactions with targets in the thyroid pathway provides a means of linking adverse outcomes, which may be influenced by many biological processes, to a thyroid mechanism. However, the use of in vitro assays beyond chemical screening is complicated by continuing limits in our knowledge of TH signaling in important life stages and tissues, such as during fetal brain development. Nonetheless, the thyroid AOP network provides an ideal tool for defining causal linkages of a chemical exerting thyroid-dependent effects and identifying research needs to quantify these effects in support of regulatory decision making. https://doi.org/10.1289/EHP5297.
Collapse
Affiliation(s)
- Pamela D Noyes
- National Center for Environmental Assessment, Office of Research and Development (ORD), U.S. Environmental Protection Agency (EPA), Washington, DC, USA
| | - Katie Paul Friedman
- National Center for Computational Toxicology, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Patience Browne
- Environment Health and Safety Division, Environment Directorate, Organisation for Economic Co-operation and Development (OECD), Paris, France
| | - Jonathan T Haselman
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| | - Mary E Gilbert
- Toxicity Assessment Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Michael W Hornung
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| | - Stan Barone
- Office of Pollution Prevention and Toxics, Office of Chemical Safety and Pollution Prevention, U.S. EPA, Washington, DC, USA
| | - Kevin M Crofton
- National Center for Computational Toxicology, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Susan C Laws
- Toxicity Assessment Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Tammy E Stoker
- Toxicity Assessment Division, NHEERL, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Steven O Simmons
- National Center for Computational Toxicology, ORD, U.S. EPA, Research Triangle Park, North Carolina, USA
| | - Joseph E Tietge
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| | - Sigmund J Degitz
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory (NHEERL), ORD, U.S. EPA, Duluth, Minnesota, USA
| |
Collapse
|
25
|
Bianco AC, Dumitrescu A, Gereben B, Ribeiro MO, Fonseca TL, Fernandes GW, Bocco BMLC. Paradigms of Dynamic Control of Thyroid Hormone Signaling. Endocr Rev 2019; 40:1000-1047. [PMID: 31033998 PMCID: PMC6596318 DOI: 10.1210/er.2018-00275] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/15/2019] [Indexed: 12/17/2022]
Abstract
Thyroid hormone (TH) molecules enter cells via membrane transporters and, depending on the cell type, can be activated (i.e., T4 to T3 conversion) or inactivated (i.e., T3 to 3,3'-diiodo-l-thyronine or T4 to reverse T3 conversion). These reactions are catalyzed by the deiodinases. The biologically active hormone, T3, eventually binds to intracellular TH receptors (TRs), TRα and TRβ, and initiate TH signaling, that is, regulation of target genes and other metabolic pathways. At least three families of transmembrane transporters, MCT, OATP, and LAT, facilitate the entry of TH into cells, which follow the gradient of free hormone between the extracellular fluid and the cytoplasm. Inactivation or marked downregulation of TH transporters can dampen TH signaling. At the same time, dynamic modifications in the expression or activity of TRs and transcriptional coregulators can affect positively or negatively the intensity of TH signaling. However, the deiodinases are the element that provides greatest amplitude in dynamic control of TH signaling. Cells that express the activating deiodinase DIO2 can rapidly enhance TH signaling due to intracellular buildup of T3. In contrast, TH signaling is dampened in cells that express the inactivating deiodinase DIO3. This explains how THs can regulate pathways in development, metabolism, and growth, despite rather stable levels in the circulation. As a consequence, TH signaling is unique for each cell (tissue or organ), depending on circulating TH levels and on the exclusive blend of transporters, deiodinases, and TRs present in each cell. In this review we explore the key mechanisms underlying customization of TH signaling during development, in health and in disease states.
Collapse
Affiliation(s)
- Antonio C Bianco
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| | - Alexandra Dumitrescu
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Miriam O Ribeiro
- Developmental Disorders Program, Center of Biologic Sciences and Health, Mackenzie Presbyterian University, São Paulo, São Paulo, Brazil
| | - Tatiana L Fonseca
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| | - Gustavo W Fernandes
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| | - Barbara M L C Bocco
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| |
Collapse
|
26
|
de Souza JS, Ferreira DR, Herai R, Carromeu C, Torres LB, Araujo BHS, Cugola F, Maciel RMB, Muotri AR, Giannocco G. Altered Gene Expression of Thyroid Hormone Transporters and Deiodinases in iPS MeCP2-Knockout Cells-Derived Neurons. Mol Neurobiol 2019; 56:8277-8295. [DOI: 10.1007/s12035-019-01645-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022]
|
27
|
López-Espíndola D, García-Aldea Á, Gómez de la Riva I, Rodríguez-García AM, Salvatore D, Visser TJ, Bernal J, Guadaño-Ferraz A. Thyroid hormone availability in the human fetal brain: novel entry pathways and role of radial glia. Brain Struct Funct 2019; 224:2103-2119. [PMID: 31165302 DOI: 10.1007/s00429-019-01896-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/27/2019] [Indexed: 01/13/2023]
Abstract
Thyroid hormones (TH) are crucial for brain development; their deficiency during neurodevelopment impairs neural cell differentiation and causes irreversible neurological alterations. Understanding TH action, and in particular the mechanisms regulating TH availability in the prenatal human brain is essential to design therapeutic strategies for neurological diseases due to impaired TH signaling during neurodevelopment. We aimed at the identification of cells involved in the regulation of TH availability in the human brain at fetal stages. To this end, we studied the distribution of the TH transporters monocarboxylate transporter 8 (MCT8) and organic anion-transporting polypeptide 1C1 (OATP1C1), as well as the TH-metabolizing enzymes types 2 and 3 deiodinases (DIO2 and DIO3). Paraffin-embedded human brain sections obtained from necropsies of thirteen fetuses from 14 to 38 gestational weeks were analyzed by immunohistochemistry and in situ hybridization. We found these proteins localized along radial glial cells, in brain barriers, in Cajal-Retzius cells, in migrating fibers of the brainstem and in some neurons and glial cells with particular and complex spatiotemporal patterns. Our findings point to an important role of radial glia in controlling TH delivery and metabolism and suggest two additional novel pathways for TH availability in the prenatal human brain: the outer, and the inner cerebrospinal fluid-brain barriers. Based on our data we propose a model of TH availability for neural cells in the human prenatal brain in which several cell types have the ability to autonomously control the required TH content.
Collapse
Affiliation(s)
- Daniela López-Espíndola
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28029, Madrid, Spain
- Escuela de Tecnología Médica and Centro de Investigaciones Biomédicas (CIB), Universidad de Valparaíso, Angamos 655, Reñaca, Viña del Mar, Chile
| | - Ángel García-Aldea
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28029, Madrid, Spain
| | | | | | - Domenico Salvatore
- Department of Public Health, University of Naples "Federico II", 80133, Naples, Italy
- CEINGE-Biotecnologie Avanzate s.c.a.r.l, 80145, Naples, Italy
| | - Theo J Visser
- Department of Internal Medicine, Erasmus Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - 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), 28029, Madrid, Spain.
- Center for Biomedical Research on Rare Diseases (CIBERER), U708, 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), 28029, Madrid, Spain.
- Center for Biomedical Research on Rare Diseases (CIBERER), U708, Madrid, Spain.
| |
Collapse
|
28
|
Zevenbergen C, Groeneweg S, Swagemakers SMA, de Jong A, Medici-Van den Herik E, Rispens M, Klootwijk W, Medici M, de Rijke YB, Meima ME, Larsen PR, Chavatte L, Venter D, Peeters RP, Van der Spek PJ, Visser WE. Functional Analysis of Genetic Variation in the SECIS Element of Thyroid Hormone Activating Type 2 Deiodinase. J Clin Endocrinol Metab 2019; 104:1369-1377. [PMID: 30423129 DOI: 10.1210/jc.2018-01605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/08/2018] [Indexed: 01/05/2023]
Abstract
CONTEXT Thyroid hormone is important for normal brain development. The type 2 deiodinase (D2) controls thyroid hormone action in the brain by activating T4 to T3. The enzymatic activity of D2 depends on the incorporation of selenocysteine for which the selenocysteine-insertion sequence (SECIS) element located in the 3' untranslated region is indispensable. We hypothesized that mutations in the SECIS element could affect D2 function, resulting in a neurocognitive phenotype. OBJECTIVE To identify mutations in the SECIS element of DIO2 in patients with intellectual disability and to test their functional consequences. DESIGN, SETTING, AND PATIENTS The SECIS element of DIO2 was sequenced in 387 patients with unexplained intellectual disability using a predefined pattern of thyroid function tests. SECIS element read-through in wild-type or mutant D2 was quantified by a luciferase reporter system in transfected cells. Functional consequences were assessed by quantifying D2 activity in cell lysate or intact cell metabolism studies. RESULTS Sequence analysis revealed 2 heterozygous mutations: c.5703C>T and c.5730A>T, which were also present in the unaffected family members. The functional evaluation showed that both mutations did not affect D2 enzyme activity in cell lysates or intact cells, although the 5730A>T mutation decreased SECIS element read-through by 75%. In the patient harboring the c.5730A>T variant, whole genome sequencing revealed a pathogenic deletion of the STXBP1 gene. CONCLUSIONS We report on two families with mutations in the SECIS element of D2. Although functional analysis showed that nucleotide 5730 is important for normal SECIS element read-through, the two variants did not segregate with a distinct phenotype.
Collapse
Affiliation(s)
- Chantal Zevenbergen
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Stefan Groeneweg
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Sigrid M A Swagemakers
- Department of Bioinformatics, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Pathology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | | | - Evita Medici-Van den Herik
- Department of Child Neurology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | | | - Wim Klootwijk
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Marco Medici
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Marcel E Meima
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - P Reed Larsen
- Department of Internal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Laurent Chavatte
- Centre International de Recherche en Infectiologie, CIRI, INSERM U1111, CNRS/ENS/UCBL1 UMR5308, Lyon, France
| | - Deon Venter
- Department of Pathology, Mater Health Services, South Brisbane, Queensland, Australia
| | - Robin P Peeters
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Peter J Van der Spek
- Department of Bioinformatics, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Pathology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
29
|
Carmody C, Ogawa-Wong AN, Martin C, Luongo C, Zuidwijk M, Sager B, Petersen T, Roginski Guetter A, Janssen R, Wu EY, Bogaards S, Neumann NM, Hau K, Marsili A, Boelen A, Silva JE, Dentice M, Salvatore D, Wagers AJ, Larsen PR, Simonides WS, Zavacki AM. A Global Loss of Dio2 Leads to Unexpected Changes in Function and Fiber Types of Slow Skeletal Muscle in Male Mice. Endocrinology 2019; 160:1205-1222. [PMID: 30951174 PMCID: PMC6482039 DOI: 10.1210/en.2019-00088] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022]
Abstract
The type 2 iodothyronine-deiodinase (D2) enzyme converts T4 to T3, and mice deficient in this enzyme [D2 knockout (D2KO) mice] have decreased T3 derived from T4 in skeletal muscle despite normal circulating T3 levels. Because slow skeletal muscle is particularly susceptible to changes in T3 levels, we expected D2 inactivation to result in more pronounced slow-muscle characteristics in the soleus muscle, mirroring hypothyroidism. However, ex vivo studies of D2KO soleus revealed higher rates of twitch contraction and relaxation and reduced resistance to fatigue. Immunostaining of D2KO soleus showed that these properties were associated with changes in muscle fiber type composition, including a marked increase in the number of fast, glycolytic type IIB fibers. D2KO soleus muscle fibers had a larger cross-sectional area, and this correlated with increased myonuclear accretion in myotubes formed from D2KO skeletal muscle precursor cells differentiated in vitro. Consistent with our functional findings, D2KO soleus gene expression was markedly different from that in hypothyroid wild-type (WT) mice. Comparison of gene expression between euthyroid WT and D2KO mice indicated that PGC-1α, a T3-dependent regulator of slow muscle fiber type, was decreased by ∼50% in D2KO soleus. Disruption of Dio2 in the C2C12 myoblast cell line led to a significant decrease in PGC-1α expression and a faster muscle phenotype upon differentiation. These results indicate that D2 loss leads to significant changes in soleus contractile function and fiber type composition that are inconsistent with local hypothyroidism and suggest that reduced levels of PCG-1α may contribute to the observed phenotypical changes.
Collapse
Affiliation(s)
| | | | | | - Cristina Luongo
- Brigham and Women’s Hospital, Boston, Massachusetts
- University of Naples “Federico II,” Napoli, Italy
| | - Marian Zuidwijk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | | | | | - Rob Janssen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Sylvia Bogaards
- Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Kaman Hau
- Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Anita Boelen
- Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - J Enrique Silva
- Baystate Medical Center, Springfield, Massachusetts
- Tufts University School of Medicine, Boston, Massachusetts
| | | | | | | | | | | | | |
Collapse
|
30
|
Stepien BK, Huttner WB. Transport, Metabolism, and Function of Thyroid Hormones in the Developing Mammalian Brain. Front Endocrinol (Lausanne) 2019; 10:209. [PMID: 31001205 PMCID: PMC6456649 DOI: 10.3389/fendo.2019.00209] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022] Open
Abstract
Ever since the discovery of thyroid hormone deficiency as the primary cause of cretinism in the second half of the 19th century, the crucial role of thyroid hormone (TH) signaling in embryonic brain development has been established. However, the biological understanding of TH function in brain formation is far from complete, despite advances in treating thyroid function deficiency disorders. The pleiotropic nature of TH action makes it difficult to identify and study discrete roles of TH in various aspect of embryogenesis, including neurogenesis and brain maturation. These challenges notwithstanding, enormous progress has been achieved in understanding TH production and its regulation, their conversions and routes of entry into the developing mammalian brain. The endocrine environment has to adjust when an embryo ceases to rely solely on maternal source of hormones as its own thyroid gland develops and starts to produce endogenous TH. A number of mechanisms are in place to secure the proper delivery and action of TH with placenta, blood-brain interface, and choroid plexus as barriers of entry that need to selectively transport and modify these hormones thus controlling their active levels. Additionally, target cells also possess mechanisms to import, modify and bind TH to further fine-tune their action. A complex picture of a tightly regulated network of transport proteins, modifying enzymes, and receptors has emerged from the past studies. TH have been implicated in multiple processes related to brain formation in mammals-neuronal progenitor proliferation, neuronal migration, functional maturation, and survival-with their exact roles changing over developmental time. Given the plethora of effects thyroid hormones exert on various cell types at different developmental periods, the precise spatiotemporal regulation of their action is of crucial importance. In this review we summarize the current knowledge about TH delivery, conversions, and function in the developing mammalian brain. We also discuss their potential role in vertebrate brain evolution and offer future directions for research aimed at elucidating TH signaling in nervous system development.
Collapse
|
31
|
Domingues JT, Wajima CS, Cesconetto PA, Parisotto EB, Winkelmann-Duarte E, Santos KD, Saleh N, Filippin-Monteiro FB, Razzera G, Mena Barreto Silva FR, Pessoa-Pureur R, Zamoner A. Experimentally-induced maternal hypothyroidism alters enzyme activities and the sensorimotor cortex of the offspring rats. Mol Cell Endocrinol 2018; 478:62-76. [PMID: 30031104 DOI: 10.1016/j.mce.2018.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 01/19/2023]
Abstract
In this study, we used an experimental model of congenital hypothyroidism to show that deficient thyroid hormones (TH) disrupt different neurochemical, morphological and functional aspects in the cerebral cortex of 15-day-old offspring. Our results showing decreased glutamine synthetase (GS) activity and Ca2+ overload in the cerebral cortex of hypothyroid pups suggest misregulated glutamate metabolism associated with developmentally induced TH deficiency. The 14C-MeAIB accumulation indicates upregulated System A activity and glutamine uptake by neurons. Energy metabolism in hypothyroid cortical slices was preserved, as demonstrated by unaltered glucose metabolism. We also found upregulated acetylcholinesterase activity, depleting acetylcholine from the synaptic cleft, pointing to disrupted cholinergic system. Increased reactive oxygen species (ROS) generation, lipid peroxidation, glutathione (GSH) depletion, which were associated with glutathione peroxidase, superoxide dismutase and gamma-glutamyltransferase downregulation suggest redox imbalance. Disrupted astrocyte cytoskeleton was evidenced by downregulated and hyperphosphorylated glial fibrillary acidic protein (GFAP). Morphological and structural characterization of the sensorimotor cerebral cortex (SCC) showed unaltered thickness of the SCC. However, decreased size of neurons on the layers II & III and IV in the right SCC and increased NeuN positive neurons in specific SCC layers, suggest that they are differently affected by the low TH levels during neurodevelopment. Hypothyroid pups presented increased number of foot-faults in the gridwalk test indicating affected motor functions. Taken together, our results show that congenital hypothyroidism disrupts glutamatergic and cholinergic neurotransmission, Ca2+ equilibrium, redox balance, cytoskeleton integrity, morphological and functional aspects in the cerebral cortex of young rats.
Collapse
Affiliation(s)
- Juliana Tonietto Domingues
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Carolinne Sayury Wajima
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Patricia Acordi Cesconetto
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Eduardo Benedetti Parisotto
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Elisa Winkelmann-Duarte
- Departamento de Ciências Morfológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Karin Dos Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Najla Saleh
- Departamento de Análises Clínicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Fabíola Branco Filippin-Monteiro
- Departamento de Análises Clínicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Guilherme Razzera
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Regina Pessoa-Pureur
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ariane Zamoner
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
32
|
Domingues JT, Cattani D, Cesconetto PA, Nascimento de Almeida BA, Pierozan P, Dos Santos K, Razzera G, Mena Barreto Silva FR, Pessoa-Pureur R, Zamoner A. Reverse T 3 interacts with αvβ3 integrin receptor and restores enzyme activities in the hippocampus of hypothyroid developing rats: Insight on signaling mechanisms. Mol Cell Endocrinol 2018; 470:281-294. [PMID: 29155306 DOI: 10.1016/j.mce.2017.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/10/2017] [Accepted: 11/15/2017] [Indexed: 01/18/2023]
Abstract
In the present study we provide evidence that 3,3',5'-triiodothyronine (reverse T3, rT3) restores neurochemical parameters induced by congenital hypothyroidism in rat hippocampus. Congenital hypothyroidism was induced by adding 0.05% propylthiouracil in the drinking water from gestation day 8 and continually up to lactation day 15. In the in vivo rT3 exposure, hypothyroid 12-day old pups were daily injected with rT3 (50 ng/kg body weight) or saline until day 14. In the ex vivo rT3 treatment, hippocampal slices from 15-day-old hypothyroid pups were incubated for 30 min with or without rT3 (1 nM). We found that ex vivo and/or in vivo exposure to rT3 failed in restoring the decreased 14C-glutamate uptake; however, restored the phosphorylation of glial fibrillary acidic protein (GFAP), 45Ca2+ influx, aspartate transaminase (AST), glutamine synthetase (GS) and gamma-glutamate transferase (GGT) activities, as well as glutathione (GSH) levels in hypothyroid hippocampus. In addition, rT3 improved 14C-2-deoxy-D-glucose uptake and lactate dehydrogenase (LDH) activity. Receptor agonists/antagonists (RGD peptide and AP-5), kinase inhibitors of p38MAPK, ERK1/2, CaMKII, PKA (SB239063, PD98059, KN93 and H89, respectively), L-type voltage-dependent calcium channel blocker (nifedipine) and intracellular calcium chelator (BAPTA-AM) were used to determine the mechanisms of the nongenomic rT3 action on GGT activity. Using molecular docking analysis, we found rT3 interaction with αvβ3 integrin receptors, nongenomically activating signaling pathways (PKA, CaMKII, p38MAPK) that restored GGT activity. We provide evidence that rT3 is an active TH metabolite and our results represent an important contribution to elucidate the nonclassical mechanism of action of this metabolite in hypothyroidism.
Collapse
Affiliation(s)
- Juliana Tonietto Domingues
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Daiane Cattani
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Patricia Acordi Cesconetto
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Paula Pierozan
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Karin Dos Santos
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Guilherme Razzera
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Regina Pessoa-Pureur
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ariane Zamoner
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Programa de Pós-Graduação em Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
33
|
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: 2.7] [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
|
34
|
van der Spek AH, Surovtseva OV, Jim KK, van Oudenaren A, Brouwer MC, Vandenbroucke-Grauls CMJE, Leenen PJM, van de Beek D, Hernandez A, Fliers E, Boelen A. Regulation of Intracellular Triiodothyronine Is Essential for Optimal Macrophage Function. Endocrinology 2018; 159:2241-2252. [PMID: 29648626 PMCID: PMC5920313 DOI: 10.1210/en.2018-00053] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/30/2018] [Indexed: 01/16/2023]
Abstract
Innate immune cells, including macrophages, have recently been identified as target cells for thyroid hormone. We hypothesized that optimal intracellular concentrations of the active thyroid hormone triiodothyronine (T3) are essential for proinflammatory macrophage function. T3 is generated intracellularly by type 2 deiodinase (D2) and acts via the nuclear thyroid hormone receptor (TR). In zebrafish embryos, D2 knockdown increased mortality during pneumococcal meningitis. Primary murine D2 knockout macrophages exhibited impaired phagocytosis and partially reduced cytokine response to stimulation with bacterial endotoxin. These effects are presumably due to reduced intracellular T3 availability. Knockdown of the main TR in macrophages, TRα, impaired polarization into proinflammatory macrophages and amplified polarization into immunomodulatory macrophages. Intracellular T3 availability and action appear to play a crucial role in macrophage function. Our data suggest that low intracellular T3 action has an anti-inflammatory effect, possibly due to an effect on macrophage polarization mediated via the TRα. This study provides important insights into the link between the endocrine and innate immune system.
Collapse
Affiliation(s)
- Anne H van der Spek
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, AZ Amsterdam, Netherlands
| | - Olga V Surovtseva
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, AZ Amsterdam, Netherlands
| | - Kin Ki Jim
- Department of Neurology, Academic Medical Center, University of Amsterdam, Amsterdam Neuroscience, AZ Amsterdam, Netherlands
- Department of Medical Microbiology and Infection Control, VU University Medical Center, HV Amsterdam, Netherlands
| | - Adri van Oudenaren
- Department of Immunology, Erasmus University Medical Center, CE Rotterdam, Netherlands
| | - Matthijs C Brouwer
- Department of Neurology, Academic Medical Center, University of Amsterdam, Amsterdam Neuroscience, AZ Amsterdam, Netherlands
| | | | - Pieter J M Leenen
- Department of Immunology, Erasmus University Medical Center, CE Rotterdam, Netherlands
| | - Diederik van de Beek
- Department of Neurology, Academic Medical Center, University of Amsterdam, Amsterdam Neuroscience, AZ Amsterdam, Netherlands
| | - Arturo Hernandez
- Department of Molecular Medicine, Maine Medical Center Research Institute, Maine Medical Center, Scarborough, Maine
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, AZ Amsterdam, Netherlands
| | - Anita Boelen
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, AZ Amsterdam, Netherlands
| |
Collapse
|
35
|
Gouveia CHA, Miranda-Rodrigues M, Martins GM, Neofiti-Papi B. Thyroid Hormone and Skeletal Development. VITAMINS AND HORMONES 2018; 106:383-472. [PMID: 29407443 DOI: 10.1016/bs.vh.2017.06.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Thyroid hormone (TH) is essential for skeletal development from the late fetal life to the onset of puberty. During this large window of actions, TH has key roles in endochondral and intramembranous ossifications and in the longitudinal bone growth. There is evidence that TH acts directly in skeletal cells but also indirectly, specially via the growth hormone/insulin-like growth factor-1 axis, to control the linear skeletal growth and maturation. The presence of receptors, plasma membrane transporters, and activating and inactivating enzymes of TH in skeletal cells suggests that direct actions of TH in these cells are crucial for skeletal development, which has been confirmed by several in vitro and in vivo studies, including mouse genetic studies, and clinical studies in patients with resistance to thyroid hormone due to dominant-negative mutations in TH receptors. This review examines progress made on understanding the mechanisms by which TH regulates the skeletal development.
Collapse
Affiliation(s)
- Cecilia H A Gouveia
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil.
| | | | - Gisele M Martins
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil; Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Bianca Neofiti-Papi
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Experimental Pathophysiology Program, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
36
|
Richard S, Flamant F. Regulation of T3 Availability in the Developing Brain: The Mouse Genetics Contribution. Front Endocrinol (Lausanne) 2018; 9:265. [PMID: 29892264 PMCID: PMC5985302 DOI: 10.3389/fendo.2018.00265] [Citation(s) in RCA: 14] [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: 12/22/2017] [Accepted: 05/07/2018] [Indexed: 12/11/2022] Open
Abstract
Alterations in maternal thyroid physiology may have deleterious consequences on the development of the fetal brain, but the underlying mechanisms remain elusive, hampering the development of appropriate therapeutic strategies. The present review sums up the contribution of genetically modified mouse models to this field. In particular, knocking out genes involved in thyroid hormone (TH) deiodination, transport, and storage has significantly improved the picture that we have of the economy of TH in the fetal brain and the underlying genetic program. These data pave the way for future studies to bridge the gap in knowledge between thyroid physiology and brain development.
Collapse
|
37
|
Abstract
Thyroid hormone signaling is customized in a time and cell-specific manner by the deiodinases, homodimeric thioredoxin fold containing selenoproteins. This ensures adequate T3 action in developing tissues, healthy adults and many disease states. D2 activates thyroid hormone by converting the pro-hormone T4 to T3, the biologically active thyroid hormone. D2 expression is tightly regulated by transcriptional mechanisms triggered by endogenous as well as environmental cues. There is also an on/off switch mechanism that controls D2 activity that is triggered by catalysis and functions via D2 ubiquitination/deubiquitination. D3 terminates thyroid hormone action by inactivation of both T4 and T3 molecules. Deiodinases play a role in thyroid hormone homeostasis, development, growth and metabolic control by affecting the intracellular levels of T3 and thus gene expression on a cell-specific basis. In many cases, tight control of these pathways by T3 is achieved with coordinated reciprocal changes in D2-mediated thyroid hormone activation D3-mediated thyroid hormone inactivation.
Collapse
|
38
|
Gothié JD, Demeneix B, Remaud S. Comparative approaches to understanding thyroid hormone regulation of neurogenesis. Mol Cell Endocrinol 2017; 459:104-115. [PMID: 28545819 DOI: 10.1016/j.mce.2017.05.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/11/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
Abstract
Thyroid hormone (TH) signalling, an evolutionary conserved pathway, is crucial for brain function and cognition throughout life, from early development to ageing. In humans, TH deficiency during pregnancy alters offspring brain development, increasing the risk of cognitive disorders. How TH regulates neurogenesis and subsequent behaviour and cognitive functions remains a major research challenge. Cellular and molecular mechanisms underlying TH signalling on proliferation, survival, determination, migration, differentiation and maturation have been studied in mammalian animal models for over a century. However, recent data show that THs also influence embryonic and adult neurogenesis throughout vertebrates (from mammals to teleosts). These latest observations raise the question of how TH availability is controlled during neurogenesis and particularly in specific neural stem cell populations. This review deals with the role of TH in regulating neurogenesis in the developing and the adult brain across different vertebrate species. Such evo-devo approaches can shed new light on (i) the evolution of the nervous system and (ii) the evolutionary control of neurogenesis by TH across animal phyla. We also discuss the role of thyroid disruptors on brain development in an evolutionary context.
Collapse
Affiliation(s)
- Jean-David Gothié
- CNRS, UMR 7221, Muséum National d'Histoire Naturelle, F-75005 Paris France
| | - Barbara Demeneix
- CNRS, UMR 7221, Muséum National d'Histoire Naturelle, F-75005 Paris France.
| | - Sylvie Remaud
- CNRS, UMR 7221, Muséum National d'Histoire Naturelle, F-75005 Paris France.
| |
Collapse
|
39
|
Wirth EK, Meyer F. Neuronal effects of thyroid hormone metabolites. Mol Cell Endocrinol 2017; 458:136-142. [PMID: 28088465 DOI: 10.1016/j.mce.2017.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/06/2017] [Accepted: 01/07/2017] [Indexed: 12/19/2022]
Abstract
Thyroid hormones and their metabolites are active regulators of gene expression, mitochondrial function and various other physiological actions in different organs and tissues. These actions are mediated by a spatio-temporal regulation of thyroid hormones and metabolites within a target cell. This spatio-temporal resolution as well as classical and non-classical actions of thyroid hormones and metabolites is accomplished and regulated on multiple levels as uptake, local activation and signaling of thyroid hormones. In this review, we will give an overview of the systems involved in regulating the presence and activity of thyroid hormones and their metabolites within the brain, specifically in neurons. While a wealth of data on thyroxin (T4) and 3,5,3'-triiodothyronine (T3) in the brain has been generated, research into the presence of action of other thyroid hormone metabolites is still sparse and requires further investigations.
Collapse
Affiliation(s)
- Eva K Wirth
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Franziska Meyer
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
40
|
Galton VA. The ups and downs of the thyroxine pro-hormone hypothesis. Mol Cell Endocrinol 2017; 458:105-111. [PMID: 28130114 DOI: 10.1016/j.mce.2017.01.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/20/2016] [Accepted: 01/19/2017] [Indexed: 12/25/2022]
Abstract
Thyroxine (T4) is the major thyroid hormone in the thyroid gland and the circulation. However, it is widely accepted on the basis of abundant evidence that 3,5,3'-triiodothyronine (T3) is responsible for most, if not all, of the physiological effects of TH in extrathyroidal tissues, and T4 functions as the pro-hormone. Whether T4 has any intrinsic activity per se or is merely a pro-hormone that must be converted to T3 in order to exert any TH action has yet to be resolved. Although there are some physiological actions of T4 that are mediated by receptors at the cell membrane (non-genomic effects), the vast majority of the physiological effects of the THs identified to date involve the binding of T3 to specific nuclear receptors to regulate gene expression (genomic effects). This review examines how the role of T4 in genomic TH action has been viewed and debated during the hundred years since it was first isolated in 1914.
Collapse
Affiliation(s)
- Valerie Anne Galton
- Department of Physiology and Neurobiology, The Geisel School of Medicine at Dartmouth, 1 Medical Center Drive, Lebanon, NH 03756, USA.
| |
Collapse
|
41
|
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: 28] [Impact Index Per Article: 4.0] [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
|
42
|
Leitch VD, Di Cosmo C, Liao XH, O’Boy S, Galliford TM, Evans H, Croucher PI, Boyde A, Dumitrescu A, Weiss RE, Refetoff S, Williams GR, Bassett JHD. An Essential Physiological Role for MCT8 in Bone in Male Mice. Endocrinology 2017; 158:3055-3066. [PMID: 28637283 PMCID: PMC5659673 DOI: 10.1210/en.2017-00399] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/12/2017] [Indexed: 11/19/2022]
Abstract
T3 is an important regulator of skeletal development and adult bone maintenance. Thyroid hormone action requires efficient transport of T4 and T3 into target cells. We hypothesized that monocarboxylate transporter (MCT) 8, encoded by Mct8 on the X-chromosome, is an essential thyroid hormone transporter in bone. To test this hypothesis, we determined the juvenile and adult skeletal phenotypes of male Mct8 knockout mice (Mct8KO) and Mct8D1D2KO compound mutants, which additionally lack the ability to convert the prohormone T4 to the active hormone T3. Prenatal skeletal development was normal in both Mct8KO and Mct8D1D2KO mice, whereas postnatal endochondral ossification and linear growth were delayed in both Mct8KO and Mct8D1D2KO mice. Furthermore, bone mass and mineralization were decreased in adult Mct8KO and Mct8D1D2KO mice, and compound mutants also had reduced bone strength. Delayed bone development and maturation in Mct8KO and Mct8D1D2KO mice is consistent with decreased thyroid hormone action in growth plate chondrocytes despite elevated serum T3 concentrations, whereas low bone mass and osteoporosis reflects increased thyroid hormone action in adult bone due to elevated systemic T3 levels. These studies identify an essential physiological requirement for MCT8 in chondrocytes, and demonstrate a role for additional transporters in other skeletal cells during adult bone maintenance.
Collapse
Affiliation(s)
- Victoria D. Leitch
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| | - Caterina Di Cosmo
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637
| | - Sam O’Boy
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| | - Thomas M. Galliford
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| | - Holly Evans
- Sheffield Myeloma Research Team, University of Sheffield, Sheffield S10 2RX, United Kingdom
| | - Peter I. Croucher
- The Garvan Institute of Medical Research and St. Vincent’s Clinical School, University of New South Wales Medicine, Sydney, New South Wales 2010, Australia
| | - Alan Boyde
- Queen Mary University of London, Oral Growth and Development, Bart’s and The London School of Medicine and Dentistry, London E1 4NS, United Kingdom
| | | | - Roy E. Weiss
- Department of Medicine, University of Miami, Miami, Florida 33136
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637
- Department of Pediatrics, The University of Chicago, Chicago, Illinois 60637
- Committee on Genetics, The University of Chicago, Chicago, Illinois 60637
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| | - J. H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London W12 0NN, United Kingdom
| |
Collapse
|
43
|
Bárez-López S, Guadaño-Ferraz A. Thyroid Hormone Availability and Action during Brain Development in Rodents. Front Cell Neurosci 2017; 11:240. [PMID: 28855863 PMCID: PMC5557746 DOI: 10.3389/fncel.2017.00240] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/31/2017] [Indexed: 12/30/2022] Open
Abstract
Thyroid hormones (THs) play an essential role in the development of all vertebrates; in particular adequate TH content is crucial for proper neurodevelopment. TH availability and action in the brain are precisely regulated by several mechanisms, including the secretion of THs by the thyroid gland, the transport of THs to the brain and neural cells, THs activation and inactivation by the metabolic enzymes deiodinases and, in the fetus, transplacental passage of maternal THs. Although these mechanisms have been extensively studied in rats, in the last decade, models of genetically modified mice have been more frequently used to understand the role of the main proteins involved in TH signaling in health and disease. Despite this, there is little knowledge about the mechanisms underlying THs availability in the mouse brain. This mini-review article gathers information from findings in rats, and the latest findings in mice regarding the ontogeny of TH action and the sources of THs to the brain, with special focus on neurodevelopmental stages. Unraveling TH economy and action in the mouse brain may help to better understand the physiology and pathophysiology of TH signaling in brain and may contribute to addressing the neurological alterations due to hypo and hyperthyroidism and TH resistance syndromes.
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 IIIMadrid, 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 IIIMadrid, Spain
| |
Collapse
|
44
|
Stohn JP, Martinez ME, Zafer M, López-Espíndola D, Keyes LM, Hernandez A. Increased aggression and lack of maternal behavior in Dio3-deficient mice are associated with abnormalities in oxytocin and vasopressin systems. GENES BRAIN AND BEHAVIOR 2017; 17:23-35. [PMID: 28715127 DOI: 10.1111/gbb.12400] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 12/17/2022]
Abstract
Thyroid hormones regulate many aspects of brain development and function, and alterations in the levels of thyroid hormone action lead to abnormal anxiety- and depression-like behaviors. A complement of factors in the brain function independently of circulating levels of hormone to strictly controlled local thyroid hormone signaling. A critical factor is the type 3 deiodinase (DIO3), which is located in neurons and protects the brain from excessive thyroid hormone. Here, we examined whether a local increase in brain thyroid hormone action secondary to DIO3 deficiency is of consequence for social behaviors. Although we did not observe alterations in sociability, Dio3-/- mice of both sexes exhibited a significant increase in aggression-related behaviors and mild deficits in olfactory function. In addition, 85% of Dio3-/- dams manifested no pup-retrieval behavior and increased aggression toward the newborns. The abnormal social behaviors of Dio3-/- mice were associated with sexually dimorphic alterations in the physiology of oxytocin (OXT) and arginine vasopressin (AVP), 2 neuropeptides with important roles in determining social interactions. These alterations included low adult serum levels of OXT and AVP, and an abnormal expression of Oxt, Avp and their receptors in the neonatal and adult hypothalamus. Our results demonstrate that DIO3 is essential for normal aggression and maternal behaviors, and indicate that abnormal local regulation of thyroid hormone action in the brain may contribute to the social deficits associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- J P Stohn
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, ME, USA
| | - M E Martinez
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, ME, USA
| | - M Zafer
- Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - D López-Espíndola
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, ME, USA
| | - L M Keyes
- Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - A Hernandez
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, ME, USA
| |
Collapse
|
45
|
Gil-Ibáñez P, Belinchón MM, Morte B, Obregón MJ, Bernal J. Is the Intrinsic Genomic Activity of Thyroxine Relevant In Vivo? Effects on Gene Expression in Primary Cerebrocortical and Neuroblastoma Cells. Thyroid 2017; 27:1092-1098. [PMID: 28605984 DOI: 10.1089/thy.2017.0024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND The possibility that the intrinsic genomic activity of thyroxine (T4) is of physiological relevance has been frequently hypothesized. It might explain gene expression patterns in the brain found in type 2-deiodinase (Dio2)-deficient mice. These mice display normal expression of most thyroid hormone-dependent genes, despite decreased brain triiodothyronine (T3). METHODS The relative effects of T4 and T3 on gene expression were analyzed in mouse neuro-2a (N2a) cells stably expressing the thyroid hormone receptor α1, and in primary mouse cerebrocortical cells enriched in astrocytes or in neurons. Cortical cells were derived from Dio2-deficient mice to prevent conversion of T4 to T3. T4 and T3 were measured in the media at the beginning and end of incubation, and T4 and T3 antibodies were used to block T4 and T3 action. RESULTS In all cell types, T4 had intrinsic genomic activity. In N2a cells, T4 activity was higher on negative regulation (1/5th of T3 activity) than on positive regulation (1/40th of T3 activity). T4 activity on positive regulation was dependent on the cell context, and was higher in primary cells than in N2a cells. CONCLUSION T4 has intrinsic genomic activity. Positive regulation depends on the cell context, and primary cells appear much more sensitive than neuroblastoma cells. In all cells, negative regulation is more sensitive to T4 than positive regulation. These properties may explain the mostly normal gene expression in the brain of Dio2-deficient mice.
Collapse
Affiliation(s)
- Pilar Gil-Ibáñez
- 1 Instituto de Investigaciones Biomédicas , Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- 2 Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III , Madrid, Spain
| | - Mónica M Belinchón
- 1 Instituto de Investigaciones Biomédicas , Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- 2 Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III , Madrid, Spain
| | - Beatriz Morte
- 2 Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III , Madrid, Spain
| | - Maria Jesus Obregón
- 1 Instituto de Investigaciones Biomédicas , Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Bernal
- 1 Instituto de Investigaciones Biomédicas , Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- 2 Center for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III , Madrid, Spain
| |
Collapse
|
46
|
Young Cho Y, Jeong Kim H, Won Jang H, Hyuk Kim T, Ki CS, Wook Kim S, Hoon Chung J. The relationship of 19 functional polymorphisms in iodothyronine deiodinase and psychological well-being in hypothyroid patients. Endocrine 2017; 57:115-124. [PMID: 28466400 DOI: 10.1007/s12020-017-1307-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/17/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Levothyroxine supplementation is insufficient for the management of one tenth of patients with hypothyroidism. Iodothyronine deiodinases have been suggested to play a role in residual hypothyroid symptoms of these patients by controlling local thyroid hormone homeostasis. Previous research has suggested a relationship between commonly inherited variations in type 2 iodothyronine deiodinase and impaired well-being. We evaluated the prevalence of iodothyronine deiodinase genotypes and their association with psychological well-being in the Korean hypothyroid population. METHODS A prospective observational study. We enrolled 196 hypothyroid subjects (136 chronic autoimmune thyroiditis and 60 thyroid cancer) and assessed baseline well-being using six validated questionnaires. Genotyping was conducted for 19 single nucleotide polymorphisms in type 1, 2, and 3 iodothyronine deiodinase using Sequenom MassARRAY matrix-assisted laser desorption/ionization time-of-flight mass spectrometry in all patients. RESULTS Frequencies of iodothyronine deiodinase genotypes and well-being scores were not different in hypothyroid subjects according to their disease types. Minor genotypes of a few iodothyronine deiodinase 1 variants (rs11206244, rs2294512, and rs4926616) were associated with reduced psychological well-being. However, iodothyronine deiodinase 2 and 3 variants had no effect on baseline well-being. CONCLUSION Minor variations in iodothyronine deiodinase 1 were associated with decreased well-being in the Korean hypothyroid population, whereas iodothyronine deiodinase 2 and 3 were not. Due to controversial results among different ethnicities, further studies to clarify the effects of iodothyronine deiodinase polymorphisms on psychological well-being are warranted in hypothyroid individuals.
Collapse
Affiliation(s)
- Yoon Young Cho
- Division of Endocrinology and Metabolism, Department of Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
- Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Hye Jeong Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Hye Won Jang
- Department of Medical Education, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Hyuk Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Thyroid Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chang-Seok Ki
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sun Wook Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Thyroid Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Hoon Chung
- Division of Endocrinology and Metabolism, Department of Medicine, Thyroid Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
47
|
Ignacio DL, Silvestre DHS, Anne-Palmer E, Bocco BMLC, Fonseca TL, Ribeiro MO, Gereben B, Bianco AC, Werneck-de-Castro JP. Early Developmental Disruption of Type 2 Deiodinase Pathway in Mouse Skeletal Muscle Does Not Impair Muscle Function. Thyroid 2017; 27:577-586. [PMID: 27967605 PMCID: PMC5385430 DOI: 10.1089/thy.2016.0392] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Myogenesis is positively regulated by thyroid hormone (triiodothyronine [T3]), which is amplified by the type 2 deiodinase (D2) activation of thyroxine to T3. Global inactivation of the Dio2 gene impairs skeletal muscle (SKM) differentiation and regeneration in response to muscle injury. Given that newborn and adult mice with late developmental SKM Dio2 disruption do not develop a significant phenotype, it was hypothesized that D2 plays an early role in this process. METHODS This was tested in mice with SKM disruption of Dio2 driven by two early developmental promoters: MYF5 and MYOD. RESULTS MYF5 myoblasts in culture differentiate normally into myotubes, despite loss of almost all D2 activity. Dio2 mRNA levels in developing SKM obtained from MYF5-D2KO embryos (E18.5) were about 54% of control littermates, but the expression of the T3-responsive genes Myh1 and 7 and Atp2a1 and 2 were not affected. In MYF5-D2KO and MYOD-D2KO neonatal hind-limb muscle, the expression of Myh1 and 7 and Atp2a2 remained unaffected, despite 60-70% loss in D2 activity and/or mRNA. Only in MYOD-D2KO neonatal muscle was there a 40% reduction in Atp2a1 mRNA. Postnatal growth of both mouse models and SKM function as assessed by exercise capacity and measurement of muscle strength were normal. Furthermore, an analysis of the adult soleus revealed no changes in the expression of T3-responsive genes, except for an about 18% increase in MYOD-D2KO SOL Myh7 mRNA. CONCLUSION Two mouse models of early developmental disruption of Dio2 in myocyte precursor exhibit no significant SKM phenotype.
Collapse
Affiliation(s)
- Daniele L Ignacio
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
- 2 Biophysics Institute and School of Physical Education and Sports, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
| | - Diego H S Silvestre
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
- 2 Biophysics Institute and School of Physical Education and Sports, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
- 3 Nutrition Institute Josué de Castro, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
| | - Elena Anne-Palmer
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
| | - Barbara M L C Bocco
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
- 4 Department of Translational Medicine, Federal University of São Paulo , São Paulo, Brazil
| | - Tatiana L Fonseca
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
| | - Miriam O Ribeiro
- 5 Developmental Disorders Program, Center for Biological and Health Sciences, Mackenzie Presbyterian University , São Paulo, Brazil
| | - Balázs Gereben
- 6 Department of Endocrine Neurobiology, Institute of Experimental Medicine , Hungarian Academy of Sciences, Budapest, Hungary
| | - Antonio C Bianco
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
| | - Joao P Werneck-de-Castro
- 1 Division of Endocrinology and Metabolism, Rush University Medical Center , Chicago, Illinois
- 2 Biophysics Institute and School of Physical Education and Sports, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
- 3 Nutrition Institute Josué de Castro, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
| |
Collapse
|
48
|
Houbrechts AM, Delarue J, Gabriëls IJ, Sourbron J, Darras VM. Permanent Deiodinase Type 2 Deficiency Strongly Perturbs Zebrafish Development, Growth, and Fertility. Endocrinology 2016; 157:3668-81. [PMID: 27580812 DOI: 10.1210/en.2016-1077] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Iodothyronine deiodinases are selenocysteine-containing enzymes that activate or inactivate thyroid hormones (THs). Deiodinase type 2 (Dio2) catalyzes the conversion of the prohormone T4 into the transcriptionally active T3 and is the predominant activating deiodinase in zebrafish. Using zinc finger nucleases, we generated two different dio2(-/-) mutant zebrafish lines to investigate the physiological function of this TH activator. The first line contains a deletion of 9 bp, resulting in an in-frame elimination of three conserved amino acids. The other line is characterized by an insertion of 4 bp, leading to the introduction of a premature stop-codon. Both lines completely lack Dio2 activity, resulting in a strong reduction of T3 abundancy in all tissues tested. Early development is clearly perturbed in these animals, as shown by a diverse set of morphometric parameters, defects in swim bladder inflation, and disturbed locomotor activity tested between 1 and 7 days after fertilization. Permanent Dio2 deficiency also provokes long-term effects because growth and especially fertility are severely hampered. Possible compensatory mechanisms were investigated in adult dio2(-/-) mutants, revealing a down-regulation of the inactivating deiodinase Dio3 and TH receptor transcript levels. As the first nonmammalian model with permanent Dio2 deficiency, these mutant zebrafish lines provide evidence that Dio2 is essential to assure normal development and to obtain a normal adult phenotype.
Collapse
Affiliation(s)
- Anne M Houbrechts
- Laboratory of Comparative Endocrinology (A.M.H., J.D., I.J.G., V.M.D.), Department of Biology, Division of Animal Physiology and Neurobiology, and Laboratory for Molecular Biodiscovery (J.S.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B-3000, Leuven, Belgium
| | - Julie Delarue
- Laboratory of Comparative Endocrinology (A.M.H., J.D., I.J.G., V.M.D.), Department of Biology, Division of Animal Physiology and Neurobiology, and Laboratory for Molecular Biodiscovery (J.S.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B-3000, Leuven, Belgium
| | - Isabelle J Gabriëls
- Laboratory of Comparative Endocrinology (A.M.H., J.D., I.J.G., V.M.D.), Department of Biology, Division of Animal Physiology and Neurobiology, and Laboratory for Molecular Biodiscovery (J.S.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B-3000, Leuven, Belgium
| | - Jo Sourbron
- Laboratory of Comparative Endocrinology (A.M.H., J.D., I.J.G., V.M.D.), Department of Biology, Division of Animal Physiology and Neurobiology, and Laboratory for Molecular Biodiscovery (J.S.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B-3000, Leuven, Belgium
| | - Veerle M Darras
- Laboratory of Comparative Endocrinology (A.M.H., J.D., I.J.G., V.M.D.), Department of Biology, Division of Animal Physiology and Neurobiology, and Laboratory for Molecular Biodiscovery (J.S.), Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B-3000, Leuven, Belgium
| |
Collapse
|
49
|
Bocco BMLC, Werneck-de-Castro JP, Oliveira KC, Fernandes GW, Fonseca TL, Nascimento BPP, McAninch EA, Ricci E, Kvárta-Papp Z, Fekete C, Bernardi MM, Gereben B, Bianco AC, Ribeiro MO. Type 2 Deiodinase Disruption in Astrocytes Results in Anxiety-Depressive-Like Behavior in Male Mice. Endocrinology 2016; 157:3682-95. [PMID: 27501182 PMCID: PMC5007895 DOI: 10.1210/en.2016-1272] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/03/2016] [Indexed: 12/22/2022]
Abstract
Millions of levothyroxine-treated hypothyroid patients complain of impaired cognition despite normal TSH serum levels. This could reflect abnormalities in the type 2 deiodinase (D2)-mediated T4-to-T3 conversion, given their much greater dependence on the D2 pathway for T3 production. T3 normally reaches the brain directly from the circulation or is produced locally by D2 in astrocytes. Here we report that mice with astrocyte-specific Dio2 inactivation (Astro-D2KO) have normal serum T3 but exhibit anxiety-depression-like behavior as found in open field and elevated plus maze studies and when tested for depression using the tail-suspension and the forced-swimming tests. Remarkably, 4 weeks of daily treadmill exercise sessions eliminated this phenotype. Microarray gene expression profiling of the Astro-D2KO hippocampi identified an enrichment of three gene sets related to inflammation and impoverishment of three gene sets related to mitochondrial function and response to oxidative stress. Despite normal neurogenesis, the Astro-D2KO hippocampi exhibited decreased expression of four of six known to be positively regulated genes by T3, ie, Mbp (∼43%), Mag (∼34%), Hr (∼49%), and Aldh1a1 (∼61%) and increased expression of 3 of 12 genes negatively regulated by T3, ie, Dgkg (∼17%), Syce2 (∼26%), and Col6a1 (∼3-fold) by quantitative real-time PCR. Notably, in Astro-D2KO animals, there was also a reduction in mRNA levels of genes known to be affected in classical animal models of depression, ie, Bdnf (∼18%), Ntf3 (∼43%), Nmdar (∼26%), and GR (∼20%), which were also normalized by daily exercise sessions. These findings suggest that defects in Dio2 expression in the brain could result in mood and behavioral disorders.
Collapse
Affiliation(s)
- Barbara M L C Bocco
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - João Pedro Werneck-de-Castro
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Kelen C Oliveira
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Gustavo W Fernandes
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Tatiana L Fonseca
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Bruna P P Nascimento
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Elizabeth A McAninch
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Esther Ricci
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Zsuzsanna Kvárta-Papp
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Csaba Fekete
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Maria Martha Bernardi
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Balázs Gereben
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Antonio C Bianco
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| | - Miriam O Ribeiro
- Division of Endocrinology and Metabolism (B.M.L.C.B., J.P.W.-d.C., G.W.F., T.L.F., E.A.M., A.C.B.), Rush University Medical Center, Chicago, Illinois 60612; Department of Translational Medicine (B.M.L.C.B., G.W.F., B.P.P.N.), Federal University of Sao Paulo, Sao Paulo SP, 04039-002, Brazil; Biophysics Institute and School of Physical Education and Sports (J.P.W.-d.C.), Federal University of Rio de Janeiro, RJ 21941-599, Brazil; Department of Clinic Endocrinology (K.C.O.), Federal University of Sao Paulo, Sao Paulo SP 04039-032, Brazil; Developmental Disorders Program (B.P.P.N., E.R., M.O.R.), Center of Biological Science and Health, Mackenzie Presbyterian University, Sao Paulo SP 01302-900 Brazil; Department of Endocrine Neurobiology (Z.K.-P., C.F., B.G.), Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest H-1083, Hungary; Department of Medicine (C.F.), Division of Endocrinology, Diabetes, and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts 02111; and Graduate Program of Environmental and Experimental Pathology (M.M.B.), Graduate Program of Dentistry, Universidade Paulista, Sao Paulo SP 04026-002, Brazil
| |
Collapse
|
50
|
Ortiga-Carvalho TM, Chiamolera MI, Pazos-Moura CC, Wondisford FE. Hypothalamus-Pituitary-Thyroid Axis. Compr Physiol 2016; 6:1387-428. [PMID: 27347897 DOI: 10.1002/cphy.c150027] [Citation(s) in RCA: 222] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The hypothalamus-pituitary-thyroid (HPT) axis determines the set point of thyroid hormone (TH) production. Hypothalamic thyrotropin-releasing hormone (TRH) stimulates the synthesis and secretion of pituitary thyrotropin (thyroid-stimulating hormone, TSH), which acts at the thyroid to stimulate all steps of TH biosynthesis and secretion. The THs thyroxine (T4) and triiodothyronine (T3) control the secretion of TRH and TSH by negative feedback to maintain physiological levels of the main hormones of the HPT axis. Reduction of circulating TH levels due to primary thyroid failure results in increased TRH and TSH production, whereas the opposite occurs when circulating THs are in excess. Other neural, humoral, and local factors modulate the HPT axis and, in specific situations, determine alterations in the physiological function of the axis. The roles of THs are vital to nervous system development, linear growth, energetic metabolism, and thermogenesis. THs also regulate the hepatic metabolism of nutrients, fluid balance and the cardiovascular system. In cells, TH actions are mediated mainly by nuclear TH receptors (210), which modify gene expression. T3 is the preferred ligand of THR, whereas T4, the serum concentration of which is 100-fold higher than that of T3, undergoes extra-thyroidal conversion to T3. This conversion is catalyzed by 5'-deiodinases (D1 and D2), which are TH-activating enzymes. T4 can also be inactivated by conversion to reverse T3, which has very low affinity for THR, by 5-deiodinase (D3). The regulation of deiodinases, particularly D2, and TH transporters at the cell membrane control T3 availability, which is fundamental for TH action. © 2016 American Physiological Society. Compr Physiol 6:1387-1428, 2016.
Collapse
Affiliation(s)
- Tania M Ortiga-Carvalho
- Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Maria I Chiamolera
- Department of Medicine, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Carmen C Pazos-Moura
- Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Fredic E Wondisford
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|