1
|
Mele C, De Marchi L, Pitino R, Costantini L, Cavigiolo B, Caputo M, Marzullo P, Aimaretti G. The interplay between thyrotropic axis, neurological complications, and rehabilitation outcomes in patients with traumatic brain injury. Best Pract Res Clin Endocrinol Metab 2025:102001. [PMID: 40307077 DOI: 10.1016/j.beem.2025.102001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and long-term disability, with its pathophysiology encompassing both primary mechanical damage and secondary neuroinflammatory, metabolic, and biochemical alterations. These complex mechanisms contribute to the observed heterogeneous clinical outcomes, including neuroendocrine dysfunctions, post-traumatic seizures, and disorders of consciousness (DoC). Thyroid hormones (THs) play essential roles in synaptic plasticity, neurogenesis and neuronal homeostasis, and the hypothalamic-pituitary-thyroid (HPT) axis has recently emerged as a potential acute and chronic modulator of neurological and functional recovery following TBI, thereby hinting at the potential involvement of THs in post-TBI outcomes. While evidence suggests that alterations in the HPT axis may influence susceptibility to seizures, progression of DoC, and rehabilitation outcomes, an increased blood-brain barrier permeability in concert with dysregulated deiodinase activity and expanding oxidative stress have all been proposed as mechanisms linking THs to post-TBI neurological complications. This review aims to summarize current evidence on the potential role of the thyrotropic axis in neurological and rehabilitation outcomes following TBI.
Collapse
Affiliation(s)
- Chiara Mele
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.
| | - Lucrezia De Marchi
- Department of Endocrinology, UZ Brussel, Laarbeeklaan, Brussels, Belgium
| | - Rosa Pitino
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Luisa Costantini
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Beatrice Cavigiolo
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy; Division of Endocrinology and Diabetology, Sant'Andrea Hospital, Azienda Sanitaria Locale (ASL) Vercelli, Vercelli, Italy
| | - Marina Caputo
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Paolo Marzullo
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Gianluca Aimaretti
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
2
|
Dierichs NTOM, Piersma AH, Peeters RP, Visser WE, Meima ME, Hessel EVS. Mechanisms of developmental neurotoxicity mediated by perturbed thyroid hormone homeostasis in the brain: an adverse outcome pathway network. Crit Rev Toxicol 2025; 55:304-320. [PMID: 40062460 DOI: 10.1080/10408444.2025.2461076] [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: 09/17/2024] [Revised: 12/31/2024] [Accepted: 01/26/2025] [Indexed: 05/24/2025]
Abstract
Thyroid hormone (TH) is crucial for proper neurodevelopment. Insufficient TH concentrations in early life are associated with lower IQ and delayed motor development in children. Intracellular levels of TH are modulated via the transmembrane transport of TH and intracellular deiodination, and can mediate gene transcription via binding to the nuclear TH receptor. Chemical exposure can disrupt TH homeostasis via modes of action targeting intracellular mechanisms, thereby potentially influencing TH transport, deiodination or signaling. Understanding the cause and effect relationships of chemical hazards interfering with TH homeostasis in the developing brain is necessary to identify how chemicals might disturb brain development and result in neurodevelopmental disorders. Adverse Outcome Pathways (AOPs) can provide a template for mapping these relationships, and so far multiple AOPs have been developed for TH homeostasis and adverse effects on cognition. The present review aims to expand current AOP networks by (1) summarizing the most important factors in the regulation of brain development under influence of TH, (2) integrating human-based mechanistic information of biological pathways which can be disturbed by TH disrupting chemicals, and (3) by incorporating brain-specific TH-mediated physiology, including barriers and cell specificity, as well as clinical knowledge. TH-specific pathways in the fetal brain are highlighted and supported by distinguishing cell type specific Molecular Initiating Events (MIEs) and downstream Key Events (KEs) for astrocytes, neurons and oligodendrocytes. Two main pathways leading to adverse outcomes (AOs) in the areas of 'cognition' and 'motor function' are decreased myelination due to oligodendrocyte dysfunction, and decreased synaptogenesis and network formation via the neurons. The proposed AOP framework can form a basis for selecting developmental neurotoxic in vitro and in silico test systems for an innovative human-focused hazard testing strategy and risk assessment of chemical exposure.
Collapse
Affiliation(s)
- Nathalie T O M Dierichs
- Centre for Health Protection (GZB), National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
- Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Aldert H Piersma
- Centre for Health Protection (GZB), National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Robin P Peeters
- Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - W Edward Visser
- Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Marcel E Meima
- Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Ellen V S Hessel
- Centre for Health Protection (GZB), National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| |
Collapse
|
3
|
Sinkó R, Salas-Lucia F, Mohácsik P, Halmos E, Wittmann G, Egri P, Bocco BMLC, Batistuzzo A, Fonseca TL, Fekete C, Bianco AC, Gereben B. Variable transduction of thyroid hormone signaling in structures of the mouse brain. Proc Natl Acad Sci U S A 2025; 122:e2415970122. [PMID: 39903117 PMCID: PMC11831203 DOI: 10.1073/pnas.2415970122] [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: 08/07/2024] [Accepted: 12/31/2024] [Indexed: 02/06/2025] Open
Abstract
L-thyroxine (L-T4) monotherapy is the standard treatment for hypothyroidism, administered daily to normalize TSH levels. Once absorbed, T4 is converted to T3 to alleviate most symptoms. However, this treatment abnormally elevates plasma T4 levels in over 50% of patients. Using L-T4-treated Thyroid Hormone (TH) Action Indicator mice, which express a T3-regulated luciferase (Luc) reporter, we examined whether these T4 elevations disrupt TH signaling. Hypothyroid mice exhibited reduced Luc expression across brain regions, and L-T4 treatment failed to restore T3 signaling uniformly. There was also variability in the activity of type 2 deiodinase (D2), the enzyme that generates most brain T3. Intracerebroventricular T4 administration achieved higher elevation of Luc expression in the mediobasal hypothalamus compared to the cortex, and studies on cultured cortical astrocytes and hypothalamic tanycytes revealed cell-type-specific responses to T4. In tanycytes, exposure to T4 sustained D2 activity, leading to progressive T3 signaling, whereas in astrocytes, T4 exposure triggered a drop in D2 activity, limiting T3 production through a ubiquitin-dependent, self-limiting mechanism. The sustained D2 activity in tanycytes was linked to rapid deubiquitination by USP33, as confirmed using a ubiquitin-specific protease (USP) pan-inhibitor and USP33 knockout mice. In conclusion, the brain's response to L-T4 treatment is heterogeneous, influenced by cell-specific regulation of D2-mediated T3 production. While cortical astrocytes exhibit limited T3 signaling due to D2 ubiquitination, tanycytes coexpressing USP33 amplify T3 signaling by rescuing ubiquitinated D2 from proteasomal degradation. These findings provide mechanistic insights into the limitations of L-T4 therapy and highlight the need for tailored approaches to managing hypothyroidism.
Collapse
Affiliation(s)
- Richárd Sinkó
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest1083, Hungary
| | - Federico Salas-Lucia
- Section of Adult and Pediatric Endocrinology and Metabolism, University of Chicago, Chicago, ILIL60637
| | - Petra Mohácsik
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest1083, Hungary
| | - Emese Halmos
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest1083, Hungary
- János Szentágothai Neurosciences Division, Doctoral College, Semmelweis University, Budapest1085, Hungary
| | - Gábor Wittmann
- Laboratory of Integrative Neuroendocrinology, HUN-REN Institute of Experimental Medicine, Budapest1083, Hungary
| | - Péter Egri
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest1083, Hungary
| | - Barbara M. L. C. Bocco
- Section of Adult and Pediatric Endocrinology and Metabolism, University of Chicago, Chicago, ILIL60637
| | - Alice Batistuzzo
- Section of Adult and Pediatric Endocrinology and Metabolism, University of Chicago, Chicago, ILIL60637
| | - Tatiana L. Fonseca
- Section of Adult and Pediatric Endocrinology and Metabolism, University of Chicago, Chicago, ILIL60637
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, HUN-REN Institute of Experimental Medicine, Budapest1083, Hungary
| | - Antonio C. Bianco
- Section of Adult and Pediatric Endocrinology and Metabolism, University of Chicago, Chicago, ILIL60637
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest1083, Hungary
| |
Collapse
|
4
|
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
|
5
|
Szécsi B, Sinkó R, Vereb A, Khochanskiy D, Benke K, Radovits T, Lakatos B, Kőszegi A, Losoncz E, Kugler S, Szabó M, Merkely B, Székely A, Gereben B. The Perioperative Period of Heart Transplantation Is Affected by Thyroid Hormone Status. Thyroid 2024; 34:774-784. [PMID: 38613807 DOI: 10.1089/thy.2023.0628] [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] [Indexed: 04/15/2024]
Abstract
Background: Orthotopic heart transplantation (HTx) is a long-term surgical therapeutic approach for patients with end-stage heart failure. The objective of the present study was to uncover associations between altered thyroid hormone (TH) status and adverse outcomes after HTx. Methods: In this prospective, single-center cohort study, 283 patients underwent HTx between 2013 and 2020 at the Heart and Vascular Center of Semmelweis University in Hungary. We measured serum free triiodothyronine (fT3), free thyroxine (fT4), and thyrotropin (TSH) pre- and postoperatively. TaqMan qPCR was used to measure type 2 deiodinase and type 3 deiodinase mRNA (Dio2 and Dio3, respectively) levels from the diseased heart bioptates. To assess the local TH action of the heart, mRNA levels of Hcn2 and Myh7 were measured in a subgroup of patients receiving extracorporeal membrane oxygenation (ECMO) postoperatively. Groups were compared using nonparametric tests. Cox regression analysis and logistic regression test were used to investigate the outcomes. The connection between serum TH parameters and cardiac gene expressions was assessed using linear regression. Results: Serum TSH (p = 0.009), fT3 (p < 0.001), and fT4 (p < 0.001) levels were lower after HTx than preoperatively. Levothyroxine (LT4) administered to donors was associated with better survival after 30 days (p = 0.049). LT4 replacement given to recipients after HTx was associated with better survival after 30 days (p = 0.018), 1 year (p = 0.002), and 2 years (p = 0.001). Dio3 mRNA level was significantly increased in patients who were treated with ECMO (p = 0.026), left ventricular assist device (LVAD) (p = 0.008), and biventricular assist device (BiVAD) (p = 0.013) preoperatively, and ECMO (p = 0.042) postoperatively, compared with those who did not require any type of mechanical circulatory support (MCS). We found no significant difference in the expression of the Hcn2 and Myh7 marker genes between patients on postoperative ECMO and those without MCS, and neither did they correlate with serum hormone levels (p = 0.519 and p = 0.056, respectively). Conclusions: We conclude that TH status plays an important role in HTx patients, and monitoring of TH status in the perioperative period may contribute to improved treatment outcomes. Our findings require independent confirmation in a randomized controlled clinical trial.
Collapse
Affiliation(s)
- Balázs Szécsi
- Doctoral School of Theoretical and Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Richárd Sinkó
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Alexandra Vereb
- Doctoral School of Theoretical and Translational Medicine, Semmelweis University, Budapest, Hungary
| | | | - Kálmán Benke
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Bálint Lakatos
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Andrea Kőszegi
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Eszter Losoncz
- Doctoral School of Theoretical and Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Szilvia Kugler
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Márk Szabó
- Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Andrea Székely
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
- Department of Oxiology and Emergency Care, Semmelweis University, Budapest, Hungary
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
6
|
Salas-Lucia F, Escamilla S, Bianco AC, Dumitrescu A, Refetoff S. Impaired T3 uptake and action in MCT8-deficient cerebral organoids underlie Allan-Herndon-Dudley syndrome. JCI Insight 2024; 9:e174645. [PMID: 38376950 PMCID: PMC11128209 DOI: 10.1172/jci.insight.174645] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 02/15/2024] [Indexed: 02/22/2024] Open
Abstract
Patients with mutations in the thyroid hormone (TH) cell transporter monocarboxylate transporter 8 (MCT8) gene develop severe neuropsychomotor retardation known as Allan-Herndon-Dudley syndrome (AHDS). It is assumed that this is caused by a reduction in TH signaling in the developing brain during both intrauterine and postnatal developmental stages, and treatment remains understandably challenging. Given species differences in brain TH transporters and the limitations of studies in mice, we generated cerebral organoids (COs) using human induced pluripotent stem cells (iPSCs) from MCT8-deficient patients. MCT8-deficient COs exhibited (i) altered early neurodevelopment, resulting in smaller neural rosettes with thinner cortical units, (ii) impaired triiodothyronine (T3) transport in developing neural cells, as assessed through deiodinase-3-mediated T3 catabolism, (iii) reduced expression of genes involved in cerebral cortex development, and (iv) reduced T3 inducibility of TH-regulated genes. In contrast, the TH analogs 3,5-diiodothyropropionic acid and 3,3',5-triiodothyroacetic acid triggered normal responses (induction/repression of T3-responsive genes) in MCT8-deficient COs, constituting proof of concept that lack of T3 transport underlies the pathophysiology of AHDS and demonstrating the clinical potential for TH analogs to be used in treating patients with AHDS. MCT8-deficient COs represent a species-specific relevant preclinical model that can be utilized to screen drugs with potential benefits as personalized therapeutics for patients with AHDS.
Collapse
Affiliation(s)
- Federico Salas-Lucia
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Sergio Escamilla
- Instituto de Neurociencias de Alicante, Miguel Hernández-CSIC University, Sant Joan d’Alacant, Alicante, Spain
| | - Antonio C. Bianco
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Alexandra Dumitrescu
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
- Committee on Molecular Metabolism and Nutrition
| | - Samuel Refetoff
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, and Committee on Genetics, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
7
|
Sabatino L, Lapi D, Del Seppia C. Factors and Mechanisms of Thyroid Hormone Activity in the Brain: Possible Role in Recovery and Protection. Biomolecules 2024; 14:198. [PMID: 38397435 PMCID: PMC10886502 DOI: 10.3390/biom14020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Thyroid hormones (THs) are essential in normal brain development, and cognitive and emotional functions. THs act through a cascade of events including uptake by the target cells by specific cell membrane transporters, activation or inactivation by deiodinase enzymes, and interaction with nuclear thyroid hormone receptors. Several thyroid responsive genes have been described in the developing and in the adult brain and many studies have demonstrated a systemic or local reduction in TH availability in neurologic disease and after brain injury. In this review, the main factors and mechanisms associated with the THs in the normal and damaged brain will be evaluated in different regions and cellular contexts. Furthermore, the most common animal models used to study the role of THs in brain damage and cognitive impairment will be described and the use of THs as a potential recovery strategy from neuropathological conditions will be evaluated. Finally, particular attention will be given to the link observed between TH alterations and increased risk of Alzheimer's Disease (AD), the most prevalent neurodegenerative and dementing condition worldwide.
Collapse
Affiliation(s)
- Laura Sabatino
- Institute of Clinical Physiology, National Council of Research, Via Moruzzi 1, 56124 Pisa, Italy;
| | - Dominga Lapi
- Department of Biology, University of Pisa, 56127 Pisa, Italy;
| | - Cristina Del Seppia
- Institute of Clinical Physiology, National Council of Research, Via Moruzzi 1, 56124 Pisa, Italy;
| |
Collapse
|
8
|
Liu M, Yu Z, Yang F, Zhao Z, Zhou M, Wang C, Zhang B, Liang G, Liu X, Shao J. BDE209-promoted Dio2 degradation in H4 glioma cells through the autophagy pathway, resulting in hypothyroidism and leading to neurotoxicity. Toxicology 2023:153581. [PMID: 37330034 DOI: 10.1016/j.tox.2023.153581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/07/2023] [Accepted: 06/15/2023] [Indexed: 06/19/2023]
Abstract
Decabromodiphenyl ether (BDE209), the homologue with the highest number of brominates in polybrominated diphenyl ethers (PBDEs), is one of the most widespread environmental persistent organic pollutants (POPs) due to its mass production and extensive application in recent decades. BDE209 is neurotoxic, possibly related to its interference with the thyroid hormone (TH) system. However, the underlying molecular mechanisms of BDE209-induced TH interference and neurobehavioral disorders remains unknown. Here, we explored how BDE209 manipulated the major enzyme, human type II iodothyronine deiodinase (Dio2), that is most important in regulating local cerebral TH equilibrium by neuroglial cells, using an in vitro model of human glioma H4 cells. Clonogenic cell survival assay and LC/MS/MS analysis showed that BDE209 could induce chronic neurotoxicity by inducing TH interference. Co-IP assay, RT-qPCR and confocal assay identified that BDE209 destroyed the stability of Dio2 without affecting its expression, and promoted its binding to p62, thereby enhancing its autophagic degradation, thus causing TH metabolism disorder and neurotoxicity. Furthermore, molecular docking studies predicted that BDE209 could effectively suppress Dio2 activity by competing with tetraiodothyronine (T4). Collectively, our study demonstrates that BDE209-induced Dio2 degradation and loss of its enzymatic activity in neuroglial cells are the fundamental pathogenic basis for BDE209-mediated cerebral TH disequilibrium and neurotoxicity, providing a target of interest for further investigation using glial/neuronal cell co-culture system and in vivo models.
Collapse
Affiliation(s)
- Min Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China; Neurology Department, Dalian University Affiliated Xinhua Hospital, Dalian, 116021, China
| | - Zhenlong Yu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Fangyu Yang
- General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Department of Neurosurgery, Shenyang, China
| | - Zikuang Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 116000, China
| | - Meirong Zhou
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Chao Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Baojing Zhang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Guobiao Liang
- General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Department of Neurosurgery, Shenyang, China.
| | - Xiaohui Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Jing Shao
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China; Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine; Liaoning Medical Center for Hematopoietic Stem Cell Transplantation; Dalian Key Laboratory of Hematology; Diamond Bay Institute of Hematology; Second Hospital of Dalian Medical University, Dalian, 116027, China.
| |
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: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 05/18/2023] [Indexed: 05/20/2023] Open
Abstract
The development of the brain, as well as mood and cognitive functions, are affected by thyroid hormone (TH) signaling. Neurons are the critical cellular target for TH action, with T3 regulating the expression of important neuronal gene sets. However, the steps involved in T3 signaling remain poorly known given that neurons express high levels of type 3 deiodinase (D3), which inactivates both T4 and T3. To investigate this mechanism, we used a compartmentalized microfluid device and identified a novel neuronal pathway of T3 transport and action that involves axonal T3 uptake into clathrin-dependent, endosomal/non-degradative lysosomes (NDLs). NDLs-containing T3 are retrogradely transported via microtubules, delivering T3 to the cell nucleus, and doubling the expression of a T3-responsive reporter gene. The NDLs also contain the monocarboxylate transporter 8 (Mct8) and D3, which transport and inactivate T3, respectively. Notwithstanding, T3 gets away from degradation because D3's active center is in the cytosol. Moreover, we used a unique mouse system to show that T3 implanted in specific brain areas can trigger selective signaling in distant locations, as far as the contralateral hemisphere. These findings provide a pathway for L-T3 to reach neurons and resolve the paradox of T3 signaling in the brain amid high D3 activity.
Collapse
Affiliation(s)
- Federico Salas-Lucia
- Section of Adult and Pediatric Endocrinology and Metabolism, University of ChicagoChicagoUnited States
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental MedicineBudapestHungary
| | - Richárd Sinkó
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
- János Szentágothai PhD School of Neurosciences, Semmelweis UniversityBudapestHungary
| | - Péter Egri
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
| | - Kristóf Rada
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
| | - Yvette Ruska
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental MedicineBudapestHungary
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism, Institute of Experimental MedicineBudapestHungary
| | - Antonio C Bianco
- Section of Adult and Pediatric Endocrinology and Metabolism, University of ChicagoChicagoUnited States
| |
Collapse
|
10
|
Liu M, Yu Z, Zhao Z, Yang F, Zhou M, Wang C, Tian X, Zhang B, Liang G, Liu X, Shao J. MiR-24-3p/Dio3 axis is essential for BDE47 to induce local thyroid hormone disorder and neurotoxicity. Toxicology 2023; 491:153527. [PMID: 37116683 DOI: 10.1016/j.tox.2023.153527] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
BDE47 (2,2,4,4-tetrabromodiphenyl ether) is a member of the most important congeners of polybrominated diphenyl ethers (PBDEs) and has been identified as a developmental, reproductive and nervous system toxicant and endocrine system disruptor due to its frequent detection in human tissue and environmental samples. Our preliminary work suggested that high- and low-level of bromodiphenyl ethers have different effects on neuronal cells with differential targets of actions on neural tissues. In this study, we presented the underlying mechanism of BDE47 neurotoxicity from the perspective of thyroid hormone (TH) metabolism using in vitro model of human SK-N-AS neuronal cells. BDE47 could induce local TH metabolism disorder in neuronal cells by inhibiting the expression of the main enzyme, human type III iodothyronine deiodinase (Dio3). Further elucidation revealed that BDE47 effectively up-regulating miR-24-3p, which binds to the 3'-UTR of Dio3 and inhibits its expression. In addition, BDE47 could also inhibit the deiodinase activity of Dio3. Collectively, our study demonstrates the molecular mechanism of BDE47 regulating Dio3-induced TH metabolism disorder through inducing miR-24-3p, providing new clues for the role of miRNAs in neurodevelopmental toxicity mediated by environmental pollutants.
Collapse
Affiliation(s)
- Min Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China; Neurology Department, Dalian University Affiliated Xinhua Hospital, Dalian, 116021, China
| | - Zhenlong Yu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Zikuang Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 116000, China
| | - Fangyu Yang
- General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Department of Neurosurgery, Shenyang, China
| | - Meirong Zhou
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Chao Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Xiangge Tian
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Baojing Zhang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Guobiao Liang
- General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Department of Neurosurgery, Shenyang, China.
| | - Xiaohui Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Jing Shao
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China; Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine; Liaoning Medical Center for Hematopoietic Stem Cell Transplantation; Dalian Key Laboratory of Hematology; Diamond Bay Institute of Hematology; Second Hospital of Dalian Medical University, Dalian, 116027, China.
| |
Collapse
|
11
|
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: 2.0] [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
|
12
|
Okada S, Isoda A, Hoshi H, Okada J, Okada K, Yamada E, Saito T, Watanabe T, Kikkawa K, Ohshima K. The ratio of free triiodothyronine to free thyroxine is regulated differently in patients with type 2 diabetes mellitus treated and not treated with sodium glucose cotransporter 2 inhibitors. Diabetes Metab Syndr 2023; 17:102704. [PMID: 36621107 DOI: 10.1016/j.dsx.2022.102704] [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: 06/24/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS Triiodothyronine reduces sodium glucose cotransporter 2 expression in the kidney and increased glucose excretion in urine of alloxan-induced diabetic rats. Free thyroxine is also negatively associated with islet beta-cell function in euthyroid subjects. However, the influence of sodium glucose cotransporter 2 inhibitor on thyroid function in patients with type 2 diabetes mellitus has not been established. METHODS We investigated thyroid function in patients with type 2 diabetes mellitus in the presence or absence of sodium glucose cotransporter 2 inhibitor in a multicenter retrospective study conducted between 2019 and 2021. All participants visited the hospital monthly for type 2 diabetes mellitus treatment and plasma glucose and glycated hemoglobin level measurements. Furthermore, thyroid-stimulating hormone, free triiodothyronine, and free thyroxine levels were measured annually. RESULTS Free triiodothyronine level and the free triiodothyronine:free thyroxine ratio in the group treated with sodium glucose cotransporter 2 inhibitor were significantly higher than the levels in the group not treated with sodium glucose cotransporter 2 inhibitor. Free triiodothyronine levels in the group treated with sodium glucose cotransporter 2 inhibitor were significantly higher than the levels in the group not treated with sodium glucose cotransporter 2 inhibitor (p = 0.040). Free thyroxine levels in the group treated with sodium glucose cotransporter 2 inhibitor were significantly lower than the levels in the group not treated with sodium glucose cotransporter 2 inhibitor (p = 0.002). Thyroid-stimulating hormone levels did not differ significantly between the two groups. CONCLUSIONS Our findings show that sodium glucose cotransporter 2 inhibitor affects free triiodothyronine levels free thyroxine levels, and the free triiodothyronine:free thyroxine ratio.
Collapse
Affiliation(s)
- Shuichi Okada
- Hoshi-Iin, 204-1 Nishizen-machi, Maebashi, Gunma, 379-2131, Japan; Hidaka Hospital, 886 Nakao-machi, Takasaki, Gunma, 370-0001, Japan.
| | - Atsushi Isoda
- Hoshi-Iin, 204-1 Nishizen-machi, Maebashi, Gunma, 379-2131, Japan
| | - Hiroto Hoshi
- Hoshi-Iin, 204-1 Nishizen-machi, Maebashi, Gunma, 379-2131, Japan
| | - Junichi Okada
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, 1301 Morris Park Ave., Price 369, Bronx, NY, 10461, USA
| | - Kazuya Okada
- Department of Orthopedic Surgery, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Eijiro Yamada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Tsugumichi Saito
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Takuya Watanabe
- Endocrinology and Metabolism, Saku Central Hospital Advanced Care Center, 28-3400 Nakagomi, Saku, Nagano, 385-0051, Japan
| | - Koji Kikkawa
- Hidaka Hospital, 886 Nakao-machi, Takasaki, Gunma, 370-0001, Japan
| | - Kihachi Ohshima
- Hidaka Hospital, 886 Nakao-machi, Takasaki, Gunma, 370-0001, Japan
| |
Collapse
|
13
|
Sinkó R, Mohácsik P, Kővári D, Penksza V, Wittmann G, Mácsai L, Fonseca TL, Bianco AC, Fekete C, Gereben B. Different Hypothalamic Mechanisms Control Decreased Circulating Thyroid Hormone Levels in Infection and Fasting-Induced Non-Thyroidal Illness Syndrome in Male Thyroid Hormone Action Indicator Mice. Thyroid 2023; 33:109-118. [PMID: 36322711 PMCID: PMC9885537 DOI: 10.1089/thy.2022.0404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background: Non-Thyroidal Illness Syndrome (NTIS) caused by infection or fasting is hallmarked by reduced circulating thyroid hormone (TH) levels. To better understand the role of local TH-action in the development of NTIS, we assessed tissue-specific changes of TH signaling in Thyroid Hormone Action Indicator (THAI) mice. Methods: NTIS was induced in young adult THAI mice by bacterial lipopolysaccharide (LPS)-administration or by 24 or 48 hours' fasting. Tissue-specific TH-action was assessed by the detection of changes of the Luciferase reporter of THAI mice with quantitative polymerase chain reaction along with tissue-specific examination of regulators of TH metabolism and signaling. Age dependence of revealed alterations of hypothalamic TH-action was also studied in 1-year-old male THAI mice. Results: LPS-treatment increased TH-action in the hypothalamic arcuate nucleus-median eminence (ARC-ME) region preceded by an increase of type 2 deiodinase (D2) expression in the same region and followed by the suppression of proTrh expression in the hypothalamic paraventricular nucleus (PVN). In contrast, LPS decreased both TH-action and D2 activity in the pituitary at both ages. Tshβ expression and serum free thyroxine (fT4) and free triiodothyronine (fT3) levels decreased in LPS-treated young adults. Tshβ expression and serum fT4 levels were not significantly affected by LPS treatment in aged animals. In contrast to LPS treatment, TH-action remained unchanged in the ARC-ME of 24 and 48 hours fasted animals accompanied with a modest decrease of proTrh expression in the PVN in the 24-hour group. Tshβ expression and fT3 level were decreased in both fasted groups, but the fT4 decreased only in the 48 hours fasted animals. Conclusions: Although the hypothalamo-pituitary-thyroid (HPT) axis is inhibited both in LPS and fasting-induced NTIS, LPS achieves this by centrally inducing local hyperthyroidism in the ARC-ME region, while fasting acts without affecting hypothalamic TH signaling. Lack of downregulation of Tshβ and fT4 in LPS-treated aged THAI mice suggests age-dependent alterations in the responsiveness of the HPT axis. The LPS-induced tissue-specific hypo-, eu-, and hyperthyroidism in different tissues of the same animal indicate that under certain conditions TH levels alone could be a poor marker of tissue TH signaling. In conclusion, decreased circulating TH levels in these two forms of NTIS are associated with different patterns of hypothalamic TH signaling.
Collapse
Affiliation(s)
- Richárd Sinkó
- Laboratory of Molecular Cell Metabolism, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai PhD School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Petra Mohácsik
- Laboratory of Molecular Cell Metabolism, Institute of Experimental Medicine, Budapest, Hungary
| | - Dóra Kővári
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| | - Veronika Penksza
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| | - Gábor Wittmann
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| | - Lilla Mácsai
- Laboratory of Molecular Cell Metabolism, Institute of Experimental Medicine, Budapest, Hungary
| | - Tatiana L. Fonseca
- Section of Adult and Pediatric Endocrinology, University of Chicago, Chicago, Illinois, USA
| | - Antonio C. Bianco
- Section of Adult and Pediatric Endocrinology, University of Chicago, Chicago, Illinois, USA
| | - Csaba Fekete
- Laboratory of Integrative Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Gereben
- Laboratory of Molecular Cell Metabolism, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
14
|
Wibowo A, Hidayat T, Wahyuningrum SN. Type 2 Deiodinase A/G (Thr92Ala) Polymorphism and Circulating Thyroid Hormone Level of Childbearing Age Women in Area Replete with Iodine Deficiency Disorders. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.11017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND: Iodothyronine deiodinase (DIO) is an enzyme that regulates thyroid hormone activity. DIO consists of three types: deiodinase 1 (D1), 2 (D2), and 3 (D3). D2 is a gene that plays an important role in regulation of the biochemistry of the thyroid hormone in several tissues. D2 also plays a role in the production of triiodothyronine and controlling thyroid hormone signals. This study measured the observation that about 15% of the normal population show that D2 gene polymorphism (Thr92Ala) potentially affects the activity of D2.
AIM: This study aimed to determine D2 polymorphisms and their association with thyroid hormone levels in women of childbearing age in replete iodine deficiency disorder areas.
METHODS: Total number of subjects was 131. Analysis of serum TSH, T3, fT3, T4, and fT4 levels was done using ELISA. Polymorphism of Thr92Ala was analyzed by PCR-RFLP method.
RESULTS: The results showed that the frequencies of the genotypes Thr92Ala were AA 16.79%, AG 41.22%, and GG 41.99%, whereas the allele frequency A 37.5% and G 62.5% (p HWE = 0.171). In this study, we found no differences of TSH and thyroid hormone level between group of each allel. Mean of TSH and thyroid hormone level was on normal range.
CONCLUSION: This D2 polymorphism is associated with fT4 levels rather than fT3 but not statistically significant. Heterozygous alleles at D2 AG have higher TSH levels compared with homozygous alleles.
Collapse
|
15
|
Abstract
The clinical availability of tissue-specific biomarkers of thyroid hormone (TH) action constitutes a "holy grail" for the field. Scientists have investigated several TH-dependent markers, including the tissue content of triiodothyronine (T3)-the active form of TH. The study of animal models and humans indicates that the T3 content varies among different tissues, mostly due to the presence of low-affinity, high-capacity cytoplasmic T3 binding proteins. Nonetheless, given that T3 levels in the plasma and tissues are in equilibrium, T3 signaling is defined by the intracellular free T3 levels. The available techniques to assess tissue T3 are invasive and not clinically applicable. However, the tracer kinetic studies revealed that serum T3 levels can accurately predict tissue T3 content and T3 signaling in most tissues, except for the brain and pituitary gland. This is true not only for normal individuals but also for patients with hypo or hyperthyroidism-but not for patients with non-thyroidal illness syndrome. Given this direct relationship between serum and tissue T3 contents and T3 signaling in most tissues, clinicians managing patients with hypothyroidism could refocus attention on monitoring serum T3 levels. Future clinical trials should aim at correlating clinical outcomes with serum T3 levels.
Collapse
Affiliation(s)
- Federico Salas-Lucia
- Section of Adult and Pediatric Endocrinology and Metabolism, The University of Chicago, Chicago, IL, United States
| | | |
Collapse
|
16
|
Shcherbitskaia AD, Kovalenko AA, Milyutina YP, Vasilev DS. Thyroid Hormone Production and Transplacental Transfer in the “Mother–Fetus” System during Gestational Hyperhomocysteinemia. NEUROCHEM J+ 2022. [DOI: 10.1134/s1819712422030102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
17
|
Effects of iodine excess on serum thyrotropin-releasing hormone levels and type 2 deiodinase in the hypothalamus of Wistar rats. Br J Nutr 2022; 127:1631-1638. [PMID: 34250878 DOI: 10.1017/s0007114521002592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Iodine is an important element in thyroid hormone biosynthesis. Thyroid function is regulated by the hypothalamic-pituitary-thyroid axis. Excessive iodine leads to elevated thyroid-stimulating hormone (TSH) levels, but the mechanism is not yet clear. Type 2 deiodinase (Dio2) is a Se-containing protease that plays a vital role in thyroid function. The purpose of this study was to explore the role of hypothalamus Dio2 in regulating TSH increase caused by excessive iodine and to determine the effects of iodine excess on thyrotropin-releasing hormone (TRH) levels. Male Wistar rats were randomised into five groups and administered different iodine dosages (folds of physiological dose): normal iodine, 3-fold iodine, 6-fold iodine, 10-fold iodine and 50-fold iodine. Rats were euthanised at 4, 8, 12 or 24 weeks after iodine administration. Serum TRH, TSH, total thyroxine (TT4) and total triiodothyronine (TT3) were determined. Hypothalamus tissues were frozen and sectioned to evaluate the expression of Dio2, Dio2 activity and monocarboxylate transporter 8 (MCT8). Prolonged high iodine intake significantly increased TSH expression (P < 0·05) but did not affect TT3 and TT4 levels. Prolonged high iodine intake decreased serum TRH levels in the hypothalamus (P < 0·05). Dio2 expression and activity in the hypothalamus exhibited an increasing trend compared at each time point with increasing iodine intake (P < 0·05). Hypothalamic MCT8 expression was increased in rats with prolonged high iodine intake (P < 0·05). These results indicate that iodine excess affects the levels of Dio2, TRH and MCT8 in the hypothalamus.
Collapse
|
18
|
Hegedüs L, Bianco AC, Jonklaas J, Pearce SH, Weetman AP, Perros P. Primary hypothyroidism and quality of life. Nat Rev Endocrinol 2022; 18:230-242. [PMID: 35042968 PMCID: PMC8930682 DOI: 10.1038/s41574-021-00625-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/15/2022]
Abstract
In the 1970s, treatment with thyroid extract was superseded by levothyroxine, a synthetic L form of tetraiodothyronine. Since then, no major innovation has emerged for the treatment of hypothyroidism. The biochemical definition of subclinical hypothyroidism is a matter of debate. Indiscriminate screening for hypothyroidism has led to overdiagnosis and treatment initiation at lower serum levels of thyroid-stimulating hormone (TSH) than previously. Adverse health effects have been documented in individuals with hypothyroidism or hyperthyroidism, and these adverse effects can affect health-related quality of life (QOL). Levothyroxine substitution improves, but does not always normalize, QOL, especially for individuals with mild hypothyroidism. However, neither studies combining levothyroxine and liothyronine (the synthetic form of tri-iodothyronine) nor the use of desiccated thyroid extract have shown robust improvements in patient satisfaction. Future studies should focus not only on a better understanding of an individual's TSH set point (the innate narrow physiological range of serum concentration of TSH in an individual, before the onset of hypothyroidism) and alternative thyroid hormone combinations and formulations, but also on autoimmunity and comorbidities unrelated to hypothyroidism as drivers of patient dissatisfaction. Attention to the long-term health consequences of hypothyroidism, beyond QOL, and the risks of overtreatment is imperative.
Collapse
Affiliation(s)
- Laszlo Hegedüs
- Department of Endocrinology, Odense University Hospital, Odense, Denmark.
| | - Antonio C Bianco
- Section of Adult and Paediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | - Simon H Pearce
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Endocrinology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Anthony P Weetman
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Petros Perros
- Department of Endocrinology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| |
Collapse
|
19
|
Mele C, Pagano L, Franciotta D, Caputo M, Nardone A, Aimaretti G, Marzullo P, Pingue V. Thyroid function in the subacute phase of traumatic brain injury: a potential predictor of post-traumatic neurological and functional outcomes. J Endocrinol Invest 2022; 45:379-389. [PMID: 34351610 PMCID: PMC8783844 DOI: 10.1007/s40618-021-01656-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 07/29/2021] [Indexed: 01/28/2023]
Abstract
PURPOSE That thyroid hormones exert pleiotropic effects and have a contributory role in triggering seizures in patients with traumatic brain injury (TBI) can be hypothesized. We aimed at investigating thyroid function tests as prognostic factors of the development of seizures and of functional outcome in TBI. METHODS This retrospective study enrolled 243 adult patients with a diagnosis of mild-to-severe TBI, consecutively admitted to our rehabilitation unit for a 6-month neurorehabilitation program. Data on occurrence of seizures, brain imaging, injury characteristics, associated neurosurgical procedures, neurologic and functional assessments, and death during hospitalization were collected at baseline, during the workup and on discharge. Thyroid function tests (serum TSH, fT4, and fT3 levels) were performed upon admission to neurorehabilitation. RESULTS Serum fT3 levels were positively associated with an increased risk of late post-traumatic seizures (LPTS) in post-TBI patients independent of age, sex and TBI severity (OR = 1.85, CI 95% 1.22-2.61, p < 0.01). Measured at admission, fT3 values higher than 2.76 pg/mL discriminated patients with late post-traumatic seizures from those without, with a sensitivity of 74.2% and a specificity of 60.9%. Independently from the presence of post-traumatic epilepsy and TBI severity, increasing TSH levels and decreasing fT3 levels were associated with worse neurological and functional outcome, as well as with higher risk of mortality within 6 months from the TBI event. CONCLUSIONS Serum fT3 levels assessed in the subacute phase post-TBI are associated with neurological and functional outcome as well as with the risk of seizure occurrence. Further studies are needed to investigate the mechanisms underlying these associations.
Collapse
Affiliation(s)
- C Mele
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| | - L Pagano
- Division of Endocrinology, Diabetology and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - D Franciotta
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - M Caputo
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - A Nardone
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Neurorehabilitation and Spinal Unit, Istituti Clinici Scientifici Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - G Aimaretti
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - P Marzullo
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Division of General Medicine, IRCCS Istituto Auxologico Italiano, Ospedale San Giuseppe, Verbania, Italy
| | - V Pingue
- Neurorehabilitation and Spinal Unit, Istituti Clinici Scientifici Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| |
Collapse
|
20
|
Mele C, De Tanti A, Bagnato S, Lucca LF, Saviola D, Estraneo A, Moretta P, Marcuccio L, Lanzillo B, Aimaretti G, Nardone A, Marzullo P, Pingue V. Thyrotropic Axis and Disorders of Consciousness in Acquired Brain Injury: A Potential Intriguing Association? Front Endocrinol (Lausanne) 2022; 13:887701. [PMID: 35872992 PMCID: PMC9302487 DOI: 10.3389/fendo.2022.887701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSE A potential involvement of thyrotropic axis in influencing the state of consciousness could be hypothesized. We aimed at investigating thyroid function tests as predictors of disorders of consciousness (DoC) and relating recovery in a large cohort of patients with DoC secondary to acquired brain injury (ABI). METHODS This retrospective, multicenter, cohort study included 151 patients with DoC following ABI, consecutively admitted for a 6-month neurorehabilitation program. Data on etiology of brain injury, evolution of DoC, disability and rehabilitation assessments, and death during rehabilitation were collected at baseline and on discharge. Thyroid function tests (serum TSH, fT4 and fT3 levels) were assessed on admission in all patients and at final discharge in 50 patients. RESULTS Lower baseline TSH levels and greater TSH increments (ΔTSH) after neurorehabilitation predicted a favorable change in DoC independent of age, sex, BMI, etiology of brain injury and initial DoC subtype (TSH: OR=0.712, CI 95% 0.533-0.951, p=0.01; ΔTSH: OR=2.878, CI 95% 1.147-7.223, p=0.02). On the other hand, neither fT4 nor fT3 or their variations appeared to play any role on DoC changes after 6-months inpatient neurorehabilitation. A lower magnitude of ΔfT4 acted as a strong predictor of improved functional disability level (β=0.655, p=0.002) and cognitive functions (β=-0.671, p=0.003), implying that smaller changes in fT4 were associated with higher outcomes. CONCLUSIONS Serum TSH levels assessed in the subacute post-ABI phase and its variation during neurorehabilitation could represent a potential biomarker of DoC evolution, while variations in fT4 levels seem to be associated with rehabilitation and cognitive functions. Further studies are needed to investigate the mechanisms underlying these associations.
Collapse
Affiliation(s)
- Chiara Mele
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- *Correspondence: Chiara Mele,
| | - Antonio De Tanti
- Cardinal Ferrari Centre, Santo Stefano Riabilitazione KOS-CARE, Fontanellato, Parma, Italy
| | - Sergio Bagnato
- Unit of Neurophysiology and Unit for Severe Acquired Brain Injuries, Rehabilitation Department, Giuseppe Giglio Foundation, Cefalù, Italy
| | | | - Donatella Saviola
- Cardinal Ferrari Centre, Santo Stefano Riabilitazione KOS-CARE, Fontanellato, Parma, Italy
| | - Anna Estraneo
- Department of Neurorehabilitation for Severe Acquired Brain Injury, Don Carlo Gnocchi Foundation, Scientific Institute for Research and Health Care, Florence, Italy
- Neurology Unit, Santa Maria della Pietà General Hospital, Nola, Italy
| | - Pasquale Moretta
- Istituti Clinici Scientifici Maugeri IRCCS, Neurorehabilitation Unit of Telese Terme Institute, Telese Terme, Italy
| | - Laura Marcuccio
- Istituti Clinici Scientifici Maugeri IRCCS, Neurorehabilitation Unit of Telese Terme Institute, Telese Terme, Italy
| | - Bernardo Lanzillo
- Istituti Clinici Scientifici Maugeri IRCCS, Neurorehabilitation Unit of Telese Terme Institute, Telese Terme, Italy
| | - Gianluca Aimaretti
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Antonio Nardone
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Istituti Clinici Scientifici Maugeri IRCCS, Neurorehabilitation and Spinal Unit of Pavia Institute and Neurorehabilitation Unit of Montescano Institute, Pavia, Italy
| | - Paolo Marzullo
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Istituto Auxologico Italiano, IRCCS, Laboratory of Metabolic Research, S. Giuseppe Hospital, Piancavallo, Italy
| | - Valeria Pingue
- Istituti Clinici Scientifici Maugeri IRCCS, Neurorehabilitation and Spinal Unit of Pavia Institute and Neurorehabilitation Unit of Montescano Institute, Pavia, Italy
| |
Collapse
|
21
|
Kapri D, Fanibunda SE, Vaidya VA. Thyroid hormone regulation of adult hippocampal neurogenesis: Putative molecular and cellular mechanisms. VITAMINS AND HORMONES 2021; 118:1-33. [PMID: 35180924 DOI: 10.1016/bs.vh.2021.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Adult hippocampal neurogenesis is sensitive to perturbations in thyroid hormone signaling, with evidence supporting a key role for thyroid hormone and thyroid hormone receptors (TRs) in the regulation of postmitotic progenitor survival and neuronal differentiation. In this book chapter we summarize the current understanding of the effects of thyroid hormone signaling on adult hippocampal progenitor development, and also critically address the role of TRs in regulation of distinct aspects of stage-specific hippocampal progenitor progression. We highlight actions of thyroid hormone on thyroid hormone responsive target genes, and the implications for hippocampal progenitor regulation. Given the influence of thyroid hormone on both mitochondrial and lipid metabolism, we discuss a putative role for regulation of metabolism in the effects of thyroid hormone on adult hippocampal neurogenesis. Finally, we highlight specific ideas that require detailed experimental investigation, and the need for future studies to obtain a deeper mechanistic insight into the influence of thyroid hormone and TRs in the developmental progression of adult hippocampal progenitors.
Collapse
Affiliation(s)
- Darshana Kapri
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sashaina E Fanibunda
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India; Medical Research Centre, Kasturba Health Society, Mumbai, India
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
22
|
Shakir MKM, Brooks DI, McAninch EA, Fonseca TL, Mai VQ, Bianco AC, Hoang TD. Comparative Effectiveness of Levothyroxine, Desiccated Thyroid Extract, and Levothyroxine+Liothyronine in Hypothyroidism. J Clin Endocrinol Metab 2021; 106:e4400-e4413. [PMID: 34185829 PMCID: PMC8530721 DOI: 10.1210/clinem/dgab478] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Studies comparing levothyroxine (LT4) therapy with LT4 + liothyronine (LT3) or desiccated thyroid extract (DTE) did not detect consistent superiority of either treatment. Here, we investigated these therapies, focusing on the whole group of LT4-treated hypothyroid patients, while also exploring the most symptomatic patients. METHODOLOGY Prospective, randomized, double-blind, crossover study of 75 hypothyroid patients randomly allocated to 1 of 3 treatment arms, LT4, LT4 + LT3, and DTE, for 22 weeks. The primary outcomes were posttreatment scores on the 36-point thyroid symptom questionnaire (TSQ-36), 12-point quality of life general health questionnaire (GHQ-12), the Wechsler memory scale-version IV (VMS-IV), and the Beck Depression Inventory (BDI). Secondary endpoints included treatment preference, biochemical and metabolic parameters, etiology of hypothyroidism, and Thr92Ala-DIO2 gene polymorphism. Analyses were performed with a linear mixed model using subject as a random factor and group as a fixed effect. RESULTS Serum TSH remained within reference range across all treatment arms. There were no differences for primary and secondary outcomes, except for a minor increase in heart rate caused by DTE. Treatment preference was not different and there were no interferences of the etiology of hypothyroidism or Thr92Ala-DIO2 gene polymorphism in the outcomes. Subgroup analyses of the 1/3 most symptomatic patients on LT4 revealed strong preference for treatment containing T3, which improved performance on TSQ-36, GHQ-12, BDI, and visual memory index (VMS-IV component). CONCLUSIONS As a group, outcomes were similar among hypothyroid patients taking DTE vs LT4 + T3 vs LT4. However, those patients that were most symptomatic on LT4 preferred and responded positively to therapy with LT4 + LT3 or DTE.
Collapse
Affiliation(s)
- Mohamed K M Shakir
- Walter Reed National Military Medical Center, Bethesda, MD 20889-5600, USA
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Daniel I Brooks
- Walter Reed National Military Medical Center, Bethesda, MD 20889-5600, USA
| | - Elizabeth A McAninch
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, IL 60612, USA
| | - Tatiana L Fonseca
- Section of Adult and Pediatric Endocrinology, University of Chicago, Chicago, IL 60637, USA
| | - Vinh Q Mai
- Walter Reed National Military Medical Center, Bethesda, MD 20889-5600, USA
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Antonio C Bianco
- Section of Adult and Pediatric Endocrinology, University of Chicago, Chicago, IL 60637, USA
| | - Thanh D Hoang
- Walter Reed National Military Medical Center, Bethesda, MD 20889-5600, USA
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
23
|
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: 65] [Impact Index Per Article: 16.3] [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
|
24
|
Agarwal S, Koh KH, Tardi NJ, Chen C, Dande RR, WerneckdeCastro JP, Sudhini YR, Luongo C, Salvatore D, Samelko B, Altintas MM, Mangos S, Bianco A, Reiser J. Deiodinase-3 is a thyrostat to regulate podocyte homeostasis. EBioMedicine 2021; 72:103617. [PMID: 34649077 PMCID: PMC8517284 DOI: 10.1016/j.ebiom.2021.103617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Nephrotic syndrome (NS) is associated with kidney podocyte injury and may occur as part of thyroid autoimmunity such as Graves' disease. Therefore, the present study was designed to ascertain if and how podocytes respond to and regulate the input of biologically active thyroid hormone (TH), 3,5,3'-triiodothyronine (T3); and also to decipher the pathophysiological role of type 3 deiodinase (D3), a membrane-bound selenoenzyme that inactivates TH, in kidney disease. METHODS To study D3 function in healthy and injured (PAN, puromycin aminonucleoside and LPS, Lipopolysaccharide-mediated) podocytes, immunofluorescence, qPCR and podocyte-specific D3 knockout mouse were used. Surface plasmon resonance (SPR), co-immunoprecipitation and Proximity Ligation Assay (PLA) were used for the interaction studies. FINDINGS Healthy podocytes expressed D3 as the predominant deiodinase isoform. Upon podocyte injury, levels of Dio3 transcript and D3 protein were dramatically reduced both in vitro and in the LPS mouse model of podocyte damage. D3 was no longer directed to the cell membrane, it accumulated in the Golgi and nucleus instead. Further, depleting D3 from the mouse podocytes resulted in foot process effacement and proteinuria. Treatment of mouse podocytes with T3 phenocopied the absence of D3 and elicited activation of αvβ3 integrin signaling, which led to podocyte injury. We also confirmed presence of an active thyroid stimulating hormone receptor (TSH-R) on mouse podocytes, engagement and activation of which resulted in podocyte injury. INTERPRETATION The study provided a mechanistic insight into how D3-αvβ3 integrin interaction can minimize T3-dependent integrin activation, illustrating how D3 could act as a renoprotective thyrostat in podocytes. Further, injury caused by binding of TSH-R with TSH-R antibody, as found in patients with Graves' disease, explained a plausible link between thyroid disorder and NS. FUNDING This work was supported by American Thyroid Association (ATA-2018-050.R1).
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | - Kwi Hye Koh
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | - Nicholas J Tardi
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | - Chuang Chen
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | | | | | | | - Cristina Luongo
- Department of Public Health, University of Naples "Federico II," Naples, Italy
| | - Domenico Salvatore
- Department of Public Health, University of Naples "Federico II," Naples, Italy
| | - Beata Samelko
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | | | - Steve Mangos
- Department of Internal Medicine, Rush University, Chicago, IL 60612
| | - Antonio Bianco
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - Jochen Reiser
- Department of Internal Medicine, Rush University, Chicago, IL 60612.
| |
Collapse
|
25
|
Abstract
Deiodinases modify the biological activity of thyroid hormone (TH) molecules, ie, they may activate thyroxine (T4) to 3,5,3'-triiodothyronine (T3), or they may inactivate T3 to 3,3'-diiodo-L-thyronine (T2) or T4 to reverse triiodothyronine (rT3). Although evidence of deiodination of T4 to T3 has been available since the 1950s, objective evidence of TH metabolism was not established until the 1970s. The modern paradigm considers that the deiodinases not only play a role in the homeostasis of circulating T3, but they also provide dynamic control of TH signaling: cells that express the activating type 2 deiodinase (D2) have enhanced TH signaling due to intracellular build-up of T3; the opposite is seen in cells that express type 3 deiodinase (D3), the inactivating deiodinase. D2 and D3 are expressed in metabolically relevant tissues such as brown adipose tissue, skeletal muscle and liver, and their roles have been investigated using cell, animal, and human models. During development, D2 and D3 expression customize for each tissue/organ the timing and intensity of TH signaling. In adult cells, D2 is induced by cyclic adenosine monophosphate (cAMP), and its expression is invariably associated with enhanced T3 signaling, expression of PGC1 and accelerated energy expenditure. In contrast, D3 expression is induced by hypoxia-inducible factor 1α (HIF-1a), dampening T3 signaling and the metabolic rate. The coordinated expression of these enzymes adjusts TH signaling in a time- and tissue-specific fashion, affecting metabolic pathways in health and disease states.
Collapse
Affiliation(s)
- Samuel C Russo
- Section of Endocrinology, Diabetes & Metabolism, University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Federico Salas-Lucia
- Section of Endocrinology, Diabetes & Metabolism, University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Antonio C Bianco
- Section of Endocrinology, Diabetes & Metabolism, University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
26
|
E Marcondes AA, Gomez TGB, Ravache TT, Batistuzzo A, Lorena FB, de Paula CS, Lowenthal R, Bianco AC, Ribeiro MO. Assessment of children in the autistic spectrum disorder that carry the Thr92Ala-DIO2 polymorphism. J Endocrinol Invest 2021; 44:1775-1782. [PMID: 33449341 PMCID: PMC8693502 DOI: 10.1007/s40618-020-01497-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/31/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION A polymorphism in the type 2 deiodinase (Thr92Ala-DIO2) gene has been associated with behavioral and cognitive dysfunction as well as neurodegeneration and oxidative stress in the central nervous system. OBJECTIVE To test whether the minor allele (Ala92) frequency (MAF) is increased in children in the autism spectrum disorder (ASD), and whether carriers of the minor allele exhibit more severe symptoms and/or worse adaptive behavior. STUDY DESIGN ASD children were evaluated at baseline and yearly throughout the study by psychologists using the following tools: autism behavior checklist, Vineland Adaptative Behaviour Scales II, non-verbal intelligence test SON-R 21/2-7, SON-R 6-40, Weschler scale for intelligence, and autism treatment evaluation checklist. SETTINGS Academic outpatient mental health facility in Sao Paulo, Brazil. PARTICIPANTS ASD boys and girls younger than 18 years of age. 132 consecutive ASD children, mostly boys (~ 80%); ~ 50% was classified as verbal. Exclusion criteria were coexistence of sensory and/or physical impairment, or any associated genetic syndromes. RESULTS Median follow-up was for an uninterrupted period of 937 days (139-1375 days), which did not vary significantly among the genotypes. The MAF was 47% in ASD patients vs. 51% in a local reference population with similar ethnic background; the clinical severity and progression were not affected by the minor allele. Carriers of the minor allele exhibited higher adaptive behavior in the domains "daily living skills" and "communication", which correlated positively with the dose of the minor allele. CONCLUSION The MAF is not different in ASD children, but carriers of the Thr92Ala-DIO2 polymorphism exhibited higher adaptive behavior.
Collapse
Affiliation(s)
- A A E Marcondes
- Developmental Disorders Program, CCBS, Center of Biological Science and Health, Mackenzie Presbyterian University, Rua da Consolação, 930 Bld. #28, São Paulo, SP, 01302-907, Brazil
| | - T G B Gomez
- Developmental Disorders Program, CCBS, Center of Biological Science and Health, Mackenzie Presbyterian University, Rua da Consolação, 930 Bld. #28, São Paulo, SP, 01302-907, Brazil
| | - T T Ravache
- Developmental Disorders Program, CCBS, Center of Biological Science and Health, Mackenzie Presbyterian University, Rua da Consolação, 930 Bld. #28, São Paulo, SP, 01302-907, Brazil
| | - A Batistuzzo
- Developmental Disorders Program, CCBS, Center of Biological Science and Health, Mackenzie Presbyterian University, Rua da Consolação, 930 Bld. #28, São Paulo, SP, 01302-907, Brazil
| | - F B Lorena
- Developmental Disorders Program, CCBS, Center of Biological Science and Health, Mackenzie Presbyterian University, Rua da Consolação, 930 Bld. #28, São Paulo, SP, 01302-907, Brazil
- Federal University of Sao Paulo, São Paulo, Brazil
| | - C S de Paula
- Developmental Disorders Program, CCBS, Center of Biological Science and Health, Mackenzie Presbyterian University, Rua da Consolação, 930 Bld. #28, São Paulo, SP, 01302-907, Brazil
| | - R Lowenthal
- Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - A C Bianco
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - M O Ribeiro
- Developmental Disorders Program, CCBS, Center of Biological Science and Health, Mackenzie Presbyterian University, Rua da Consolação, 930 Bld. #28, São Paulo, SP, 01302-907, Brazil.
| |
Collapse
|
27
|
Abstract
Hormones are key drivers of cancer development, and alteration of the intratumoral concentration of thyroid hormone (TH) is a common feature of many human neoplasias. Besides the systemic control of TH levels, the expression and activity of deiodinases constitute a major mechanism for the cell-autonomous, prereceptoral control of TH action. The action of deiodinases ensures tight control of TH availability at intracellular level in a time- and tissue-specific manner, and alterations in deiodinase expression are frequent in tumors. Research over the past decades has shown that in cancer cells, a complex and dynamic expression of deiodinases is orchestrated by a network of growth factors, oncogenic proteins, and miRNA. It has become increasingly evident that this fine regulation exposes cancer cells to a dynamic concentration of TH that is functional to stimulate or inhibit various cellular functions. This review summarizes recent advances in the identification of the complex interplay between deiodinases and cancer and how this family of enzymes is relevant in cancer progression. We also discuss whether deiodinase expression could represent a diagnostic tool with which to define tumor staging in cancer treatment or even a therapeutic tool against cancer.
Collapse
Affiliation(s)
- Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples “Federico II,” Naples, Italy
| | - Maria Angela De Stefano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II,” Naples, Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples “Federico II,” Naples, Italy
| | - Domenico Salvatore
- Department of Public Health, University of Naples “Federico II,” Naples, Italy
- Correspondence: Domenico Salvatore, Department of Public Health, University of Naples “Federico II”, Napoli, Italy.
| |
Collapse
|
28
|
Marcelino CP, McAninch EA, Fernandes GW, Bocco BMLC, Ribeiro MO, Bianco AC. Temporal Pole Responds to Subtle Changes in Local Thyroid Hormone Signaling. J Endocr Soc 2020; 4:bvaa136. [PMID: 33123655 PMCID: PMC7575126 DOI: 10.1210/jendso/bvaa136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
To study thyroid hormone (TH) signaling in the human brain, we analyzed published microarray data sets of the temporal pole (Brodmann area 38) of 19 deceased donors. An index of TH signaling built on the expression of 19 well known TH-responsive genes in mouse brains (T3S+) varied from 0.92 to 1.1. After Factor analysis, T3S+ correlated independently with the expression of TH transporters (MCT8, LAT2), TH receptor (TR) beta and TR coregulators (CARM1, MED1, KAT2B, SRC2, SRC3, NCOR2a). Unexpectedly, no correlation was found between T3S+ vs DIO2, DIO3, SRC1, or TRα. An unbiased systematic analysis of the entire transcriptome identified a set of 1649 genes (set #1) with strong positive correlation with T3S+ (r > 0.75). Factor analysis of set #1 identified 2 sets of genes that correlated independently with T3S+, sets #2 (329 genes) and #3 (191 genes). When processed through the Molecular Signatures Data Base (MSigDB), both sets #2 and #3 were enriched with Gene Ontology (GO)-sets related to synaptic transmission and metabolic processes. Ranking individual human brain donors according to their T3S+ led us to identify 1262 genes (set #4) with >1.3-fold higher expression in the top half. The analysis of the overlapped genes between sets #1 and #4 resulted in 769 genes (set #5), which have a very similar MSigDB signature as sets #2 and #3. In conclusion, gene expression in the human temporal pole can be assessed through T3S+ and fluctuates with subtle variations in local TH signaling.
Collapse
Affiliation(s)
- Cícera P Marcelino
- Department of Health and Biological Sciences - CCBS, Mackenzie Presbyterian University, Sao Paulo, Sao Paulo, Brazil
- Department of Translational Medicine, Federal University of Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Elizabeth A McAninch
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois
| | - Gustavo W Fernandes
- Section of Endocrinology and Metabolism, University of Chicago, Chicago, Illinois
| | - Barbara M L C Bocco
- Section of Endocrinology and Metabolism, University of Chicago, Chicago, Illinois
| | - Miriam O Ribeiro
- Department of Health and Biological Sciences - CCBS, Mackenzie Presbyterian University, Sao Paulo, Sao Paulo, Brazil
- Department of Translational Medicine, Federal University of Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Antonio C Bianco
- Section of Endocrinology and Metabolism, University of Chicago, Chicago, Illinois
| |
Collapse
|
29
|
Farkas E, Varga E, Kovács B, Szilvásy-Szabó A, Cote-Vélez A, Péterfi Z, Matziari M, Tóth M, Zelena D, Mezriczky Z, Kádár A, Kővári D, Watanabe M, Kano M, Mackie K, Rózsa B, Ruska Y, Tóth B, Máté Z, Erdélyi F, Szabó G, Gereben B, Lechan RM, Charli JL, Joseph-Bravo P, Fekete C. A Glial-Neuronal Circuit in the Median Eminence Regulates Thyrotropin-Releasing Hormone-Release via the Endocannabinoid System. iScience 2020; 23:100921. [PMID: 32143135 PMCID: PMC7058404 DOI: 10.1016/j.isci.2020.100921] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/20/2019] [Accepted: 02/12/2020] [Indexed: 12/21/2022] Open
Abstract
Based on the type-I cannabinoid receptor (CB1) content of hypophysiotropic axons and the involvement of tanycytes in the regulation of the hypothalamic-pituitary-thyroid (HPT) axis, we hypothesized that endocannabinoids are involved in the tanycyte-induced regulation of TRH release in the median eminence (ME). We demonstrated that CB1-immunoreactive TRH axons were associated to DAGLα-immunoreactive tanycyte processes in the external zone of ME and showed that endocannabinoids tonically inhibit the TRH release in this tissue. We showed that glutamate depolarizes the tanycytes, increases their intracellular Ca2+ level and the 2-AG level of the ME via AMPA and kainite receptors and glutamate transport. Using optogenetics, we demonstrated that glutamate released from TRH neurons influences the tanycytes in the ME. In summary, tanycytes regulate TRH secretion in the ME via endocannabinoid release, whereas TRH axons regulate tanycytes by glutamate, suggesting the existence of a reciprocal microcircuit between tanycytes and TRH terminals that controls TRH release. Tanycytes tonically inhibit the activity of TRH axons via endocannabinoid release Glutamate depolarizes the tanycytes and regulates their 2-AG synthesis Glutamate released from the hypophysiotropic TRH axons influences tanycytes A microcircuit utilizing glutamate and endocannabinoids regulates TRH release
Collapse
Affiliation(s)
- Erzsébet Farkas
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Edina Varga
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Balázs Kovács
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Anett Szilvásy-Szabó
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Antonieta Cote-Vélez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, México
| | - Zoltán Péterfi
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Magdalini Matziari
- Department of Chemistry, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Mónika Tóth
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Dóra Zelena
- Department of Behavioral Neurobiology, Institute of Experimental Medicine, Budapest, Hungary; Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, Pécs 7624, Hungary
| | - Zsolt Mezriczky
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest 1088, Hungary
| | - Andrea Kádár
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Dóra Kővári
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo 060-8638, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University, Bloomington 474052, IN, USA
| | - Balázs Rózsa
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Yvette Ruska
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Blanka Tóth
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Szent Gellert ter 4, Budapest 1111, Hungary
| | - Zoltán Máté
- Medical Gene Technology Unit, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Ferenc Erdélyi
- Medical Gene Technology Unit, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Gábor Szabó
- Medical Gene Technology Unit, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary
| | - Ronald M Lechan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston 02111, MA, USA; Department of Neuroscience, Tufts University School of Medicine, Boston 02111, MA, USA
| | - Jean-Louis Charli
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, México
| | - Patricia Joseph-Bravo
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca 62210, México
| | - Csaba Fekete
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Szigony u. 43, Budapest 1083, Hungary; Department of Neuroscience, Tufts University School of Medicine, Boston 02111, MA, USA.
| |
Collapse
|
30
|
Taroza S, Rastenytė D, Podlipskytė A, Patamsytė V, Mickuvienė N. Deiodinases, organic anion transporter polypeptide polymorphisms and ischemic stroke outcomes. J Neurol Sci 2019; 407:116457. [PMID: 31677555 DOI: 10.1016/j.jns.2019.116457] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ischemic stroke is a major cause of premature death and chronic disability worldwide, and individual variation in functional outcome is strongly influenced by genetic factors. Neuroendocrine signaling by the hypothalamic-hypophyseal-thyroid axis is a critical regulator of post-stroke pathogenesis, suggesting that allelic variants in thyroid hormone (TH) signaling can influence stroke outcome. AIM To examine associations between acute ischemic stroke (AIS) outcome and allelic variants of the TH metabolizing enzymes deiodinase type 1-3 (DIO1-3) and membrane transporting organic anion polypeptide C1 (OATP1C1). METHODS Eligible AIS patients from Lithuania (n = 248) were genotyped for ten DIO1-3 and OATP1C1 single nucleotide polymorphisms (SNPs): DIO1 rs12095080-A/G, rs11206244-C/T, and rs2235544-A/C; DIO2 rs225014-T/C and rs225015-G/A; DIO3 rs945006-T/G; OATP1C1 rs974453-G/A, rs10444412-T/C, rs10770704-C/T, and rs1515777-A/G. Functional outcome was evaluated one year after index AIS using the modified Rankin Scale. Analyses were adjusted for important confounders, including serum free triiodothyronine. RESULTS After adjustment for potential confounders, the major allelic (wild-type) DIO3 genotype rs945006-TT was associated with better 1-year AIS functional outcome (odds ratio [OR] = 0.25; 95% confidence interval [CI]: 0.08-0.74; p = .013), while the wild-type OATP1C1 genotype rs10770704-CC was associated with poorer outcome (OR = 2.00, 95%CI: 1.04-3.86; p = .038). CONCLUSION Allelic variants in thyroid axis genes may prove useful for prognosis and treatment guidance.
Collapse
Affiliation(s)
- Saulius Taroza
- Laboratory of Behavioral Medicine (Palanga), Neuroscience Institute, Lithuanian University of Health Sciences, Lithuania.
| | - Daiva Rastenytė
- Department of Neurology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Aurelija Podlipskytė
- Laboratory of Behavioral Medicine (Palanga), Neuroscience Institute, Lithuanian University of Health Sciences, Lithuania
| | - Vaiva Patamsytė
- Institute of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Narseta Mickuvienė
- Laboratory of Behavioral Medicine (Palanga), Neuroscience Institute, Lithuanian University of Health Sciences, Lithuania
| |
Collapse
|
31
|
Talhada D, Santos CRA, Gonçalves I, Ruscher K. Thyroid Hormones in the Brain and Their Impact in Recovery Mechanisms After Stroke. Front Neurol 2019; 10:1103. [PMID: 31681160 PMCID: PMC6814074 DOI: 10.3389/fneur.2019.01103] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/02/2019] [Indexed: 12/23/2022] Open
Abstract
Thyroid hormones are of fundamental importance for brain development and essential factors to warrant brain functions throughout life. Their actions are mediated by binding to specific intracellular and membranous receptors regulating genomic and non-genomic mechanisms in neurons and populations of glial cells, respectively. Among others, mechanisms include the regulation of neuronal plasticity processes, stimulation of angiogenesis and neurogenesis as well modulating the dynamics of cytoskeletal elements and intracellular transport processes. These mechanisms overlap with those that have been identified to enhance recovery of lost neurological functions during the first weeks and months after ischemic stroke. Stimulation of thyroid hormone signaling in the postischemic brain might be a promising therapeutic strategy to foster endogenous mechanisms of repair. Several studies have pointed to a significant association between thyroid hormones and outcome after stroke. With this review, we will provide an overview on functions of thyroid hormones in the healthy brain and summarize their mechanisms of action in the developing and adult brain. Also, we compile the major thyroid-modulated molecular pathways in the pathophysiology of ischemic stroke that can enhance recovery, highlighting thyroid hormones as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Daniela Talhada
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilha, Portugal
- LUBIN Lab-Lunds Laboratorium för Neurokirurgisk Hjärnskadeforskning, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Cecília Reis Alves Santos
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilha, Portugal
| | - Isabel Gonçalves
- CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilha, Portugal
| | - Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
- LUBIN Lab-Lunds Laboratorium för Neurokirurgisk Hjärnskadeforskning, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW This review summarizes recent literature on nonthyroidal illness syndrome (NTI) and outcome of pediatric critical illness, to provide insight in pathophysiology and therapeutic implications. RECENT FINDINGS NTI is typically characterized by lowered triiodothyronine levels without compensatory TSH rise. Although NTI severity is associated with poor outcome of pediatric critical illness, it remains unclear whether this association reflects an adaptive protective response or contributes to poor outcome. Recently, two metabolic interventions that improved outcome also altered NTI in critically ill children. These studies shed new light on the topic, as the results suggested that the peripheral NTI component, with inactivation of thyroid hormone, may represent a beneficial adaptation, whereas the central component, with suppressed TSH-driven thyroid hormone secretion, may be maladaptive. There is currently insufficient evidence for treatment of NTI in children. However, the recent findings raised the hypothesis that reactivation of the central NTI component could offer benefit, which should be tested in RCTs. SUMMARY NTI in critically ill children can be modified by metabolic interventions. The peripheral, but not the central, component of NTI may be a beneficial adaptive response. These findings open perspectives for the development of novel strategies to improve outcome of critical illness in children.
Collapse
Affiliation(s)
- An Jacobs
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven University Hospital, Leuven, Belgium
| | | | | |
Collapse
|
33
|
de Vries EM, Surovtseva O, Vos WG, Kunst RF, van Beeren M, Kwakkel J, Chassande O, Ackermans MT, Fliers E, Boelen A. Downregulation of Type 3 Deiodinase in the Hypothalamus During Inflammation. Thyroid 2019; 29:1336-1343. [PMID: 31303139 DOI: 10.1089/thy.2019.0201] [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] [Indexed: 12/28/2022]
Abstract
Background: Inflammation is associated with marked changes in cellular thyroid hormone (TH) metabolism in triiodothyronine (T3) target organs. In the hypothalamus, type 2 deiodinase (D2), the main T3 producing enzyme, increases upon inflammation, leading to an increase in local T3 availability, which in turn decreases thyrotropin releasing hormone expression in the paraventricular nucleus. Type 3 deiodinase (D3), the T3 inactivating enzyme, decreases during inflammation, which might also contribute to the increased T3 availability in the hypothalamus. While it is known that D2 is regulated by nuclear factor κB (NF-κB) during inflammation, the underlying mechanisms of D3 regulation are unknown. Therefore, the aim of the present study was to investigate inflammation-induced D3 regulation using in vivo and in vitro models. Methods: Mice were injected with a sublethal dose of bacterial endotoxin (lipopolysaccharide [LPS]) to induce a systemic acute-phase response. A human neuroblastoma (SK-N-AS) cell line was used to test the involvement of the thyroid hormone receptor alpha 1 (TRα1) as well as the activator protein-1 (AP-1) and NF-κB inflammatory pathways in the inflammation-induced decrease of D3. Results: D3 expression in the hypothalamus was decreased 24 hours after LPS injection in mice. This decrease was similar in mice lacking the TRα. Incubation of SK-N-AS cells with LPS robustly decreased both D3 mRNA expression and activity. This led to increased intracellular T3 concentrations. The D3 decrease was prevented when NF-κB or AP-1 was inhibited. TRα1 mRNA expression decreased in SK-N-AS cells incubated with LPS, but knockdown of the TRα in SK-N-AS cells did not prevent the LPS-induced D3 decrease. Conclusions: We conclude that the inflammation-induced D3 decrease in the hypothalamus is mediated by the inflammatory pathways NF-κB and AP-1, but not TRα1. Furthermore, the observed decrease modulates intracellular T3 concentrations. Our results suggest a concerted action of inflammatory modulators to regulate both hypothalamic D2 and D3 activities to increase the local TH concentrations.
Collapse
Affiliation(s)
- Emmely M de Vries
- Endocrine Laboratory, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Olga Surovtseva
- Endocrine Laboratory, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Winnie G Vos
- Endocrine Laboratory, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Roni F Kunst
- Endocrine Laboratory, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mieke van Beeren
- Endocrine Laboratory, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Joan Kwakkel
- Endocrine Laboratory, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Olivier Chassande
- Tissue Bioengineering, U1026, F-33076, University of Bordeaux, Bordeaux, France
| | - Mariette T Ackermans
- Endocrine Laboratory, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anita Boelen
- Endocrine Laboratory, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Research Institute Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
34
|
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: 177] [Impact Index Per Article: 29.5] [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
|
35
|
Wray JR, Davies A, Sefton C, Allen TJ, Adamson A, Chapman P, Lam BYH, Yeo GSH, Coll AP, Harno E, White A. Global transcriptomic analysis of the arcuate nucleus following chronic glucocorticoid treatment. Mol Metab 2019; 26:5-17. [PMID: 31176677 PMCID: PMC6667392 DOI: 10.1016/j.molmet.2019.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/03/2019] [Accepted: 05/16/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Glucocorticoids (GCs) are widely prescribed medications that are well recognized to cause adverse metabolic effects including hyperphagia, obesity, and hyperglycemia. These effects have been recapitulated in a murine model of GC excess, and we hypothesize that they are mediated, in part, through central mechanisms. This study aimed to identify genes in the hypothalamic arcuate nucleus (ARC) that are altered with GC treatment and evaluate their contribution to GC-induced metabolic abnormalities. METHODS Corticosterone (Cort; 75 μg/ml) was administered in the drinking water to male C57Bl/6J mice for 2 days or 4 weeks. Phenotypic analysis of each group was undertaken and central and peripheral tissues were collected for biochemical and mRNA analyses. Arcuate nuclei were isolated by laser capture microdissection and tissue analyzed by RNA-seq. RESULTS RNA-seq analysis of ARC tissue from 4 week Cort treated mice revealed 21 upregulated and 22 downregulated genes at a time when mice had increased food intake, expansion of adipose tissue mass, and insulin resistance. In comparison, after 2 days Cort treatment, when the main phenotypic change was increased food intake, RNA-seq identified 30 upregulated and 16 downregulated genes. Within the genes altered at 2 days were a range of novel genes but also those known to be regulated by GCs, including Fkbp5, Mt2, Fam107a, as well as some involved in the control of energy balance, such as Agrp, Sepp1, Dio2, and Nmb. Of the candidate genes identified by RNA-seq, type-II iodothyronine deiodinase (Dio2) was chosen for further investigation as it was increased (2-fold) with Cort, and has been implicated in the control of energy balance via the modulation of hypothalamic thyroid hormone availability. Targeted knockdown of Dio2 in the MBH using AAV-mediated CRISPR-Cas9 produced a mild attenuation in GC-induced brown adipose tissue weight gain, as well as a 56% reduction in the GC-induced increase in Agrp. However, this conferred no protection from GC-induced hyperphagia, obesity, or hyperglycemia. CONCLUSIONS This study identified a comprehensive set of genes altered by GCs in the ARC and enabled the selection of key candidate genes. Targeted knockdown of hypothalamic Dio2 revealed that it did not mediate the chronic GC effects on hyperphagia and hyperglycemia.
Collapse
Affiliation(s)
- Jonathan R Wray
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Alison Davies
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Charlotte Sefton
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Tiffany-Jayne Allen
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | - Antony Adamson
- Manchester Transgenic Unit, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK
| | | | - Brian Y H Lam
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Giles S H Yeo
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Anthony P Coll
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Erika Harno
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK.
| | - Anne White
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, M13 9PT, UK.
| |
Collapse
|
36
|
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: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022]
|
37
|
Zevenbergen C, Groeneweg S, Swagemakers SMA, de Jong A, Medici-Van den Herik E, Rispens M, Klootwijk W, Medici M, de Rijke YB, Meima ME, Larsen PR, Chavatte L, Venter D, Peeters RP, Van der Spek PJ, Visser WE. Functional Analysis of Genetic Variation in the SECIS Element of Thyroid Hormone Activating Type 2 Deiodinase. J Clin Endocrinol Metab 2019; 104:1369-1377. [PMID: 30423129 DOI: 10.1210/jc.2018-01605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/08/2018] [Indexed: 01/05/2023]
Abstract
CONTEXT Thyroid hormone is important for normal brain development. The type 2 deiodinase (D2) controls thyroid hormone action in the brain by activating T4 to T3. The enzymatic activity of D2 depends on the incorporation of selenocysteine for which the selenocysteine-insertion sequence (SECIS) element located in the 3' untranslated region is indispensable. We hypothesized that mutations in the SECIS element could affect D2 function, resulting in a neurocognitive phenotype. OBJECTIVE To identify mutations in the SECIS element of DIO2 in patients with intellectual disability and to test their functional consequences. DESIGN, SETTING, AND PATIENTS The SECIS element of DIO2 was sequenced in 387 patients with unexplained intellectual disability using a predefined pattern of thyroid function tests. SECIS element read-through in wild-type or mutant D2 was quantified by a luciferase reporter system in transfected cells. Functional consequences were assessed by quantifying D2 activity in cell lysate or intact cell metabolism studies. RESULTS Sequence analysis revealed 2 heterozygous mutations: c.5703C>T and c.5730A>T, which were also present in the unaffected family members. The functional evaluation showed that both mutations did not affect D2 enzyme activity in cell lysates or intact cells, although the 5730A>T mutation decreased SECIS element read-through by 75%. In the patient harboring the c.5730A>T variant, whole genome sequencing revealed a pathogenic deletion of the STXBP1 gene. CONCLUSIONS We report on two families with mutations in the SECIS element of D2. Although functional analysis showed that nucleotide 5730 is important for normal SECIS element read-through, the two variants did not segregate with a distinct phenotype.
Collapse
Affiliation(s)
- Chantal Zevenbergen
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Stefan Groeneweg
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Sigrid M A Swagemakers
- Department of Bioinformatics, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Pathology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | | | - Evita Medici-Van den Herik
- Department of Child Neurology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | | | - Wim Klootwijk
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Marco Medici
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Marcel E Meima
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - P Reed Larsen
- Department of Internal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Laurent Chavatte
- Centre International de Recherche en Infectiologie, CIRI, INSERM U1111, CNRS/ENS/UCBL1 UMR5308, Lyon, France
| | - Deon Venter
- Department of Pathology, Mater Health Services, South Brisbane, Queensland, Australia
| | - Robin P Peeters
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Peter J Van der Spek
- Department of Bioinformatics, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Pathology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
38
|
Morita M, Ikeshima-Kataoka H, Kreft M, Vardjan N, Zorec R, Noda M. Metabolic Plasticity of Astrocytes and Aging of the Brain. Int J Mol Sci 2019; 20:ijms20040941. [PMID: 30795555 PMCID: PMC6413111 DOI: 10.3390/ijms20040941] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 01/03/2023] Open
Abstract
As part of the blood-brain-barrier, astrocytes are ideally positioned between cerebral vasculature and neuronal synapses to mediate nutrient uptake from the systemic circulation. In addition, astrocytes have a robust enzymatic capacity of glycolysis, glycogenesis and lipid metabolism, managing nutrient support in the brain parenchyma for neuronal consumption. Here, we review the plasticity of astrocyte energy metabolism under physiologic and pathologic conditions, highlighting age-dependent brain dysfunctions. In astrocytes, glycolysis and glycogenesis are regulated by noradrenaline and insulin, respectively, while mitochondrial ATP production and fatty acid oxidation are influenced by the thyroid hormone. These regulations are essential for maintaining normal brain activities, and impairments of these processes may lead to neurodegeneration and cognitive decline. Metabolic plasticity is also associated with (re)activation of astrocytes, a process associated with pathologic events. It is likely that the recently described neurodegenerative and neuroprotective subpopulations of reactive astrocytes metabolize distinct energy substrates, and that this preference is supposed to explain some of their impacts on pathologic processes. Importantly, physiologic and pathologic properties of astrocytic metabolic plasticity bear translational potential in defining new potential diagnostic biomarkers and novel therapeutic targets to mitigate neurodegeneration and age-related brain dysfunctions.
Collapse
Affiliation(s)
- Mitsuhiro Morita
- Department of Biology, Graduate School of Sciences, Kobe University, 657-8501 Kobe, Japan.
| | - Hiroko Ikeshima-Kataoka
- Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan.
| | - Marko Kreft
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
- Department of Biology, Biotechnical Faculty University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Nina Vardjan
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
39
|
Jo S, Fonseca TL, Bocco BMLC, Fernandes GW, McAninch EA, Bolin AP, Da Conceição RR, Werneck-de-Castro JP, Ignacio DL, Egri P, Németh D, Fekete C, Bernardi MM, Leitch VD, Mannan NS, Curry KF, Butterfield NC, Bassett JD, Williams GR, Gereben B, Ribeiro MO, Bianco AC. Type 2 deiodinase polymorphism causes ER stress and hypothyroidism in the brain. J Clin Invest 2019; 129:230-245. [PMID: 30352046 PMCID: PMC6307951 DOI: 10.1172/jci123176] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/11/2018] [Indexed: 12/31/2022] Open
Abstract
Levothyroxine (LT4) is a form of thyroid hormone used to treat hypothyroidism. In the brain, T4 is converted to the active form T3 by type 2 deiodinase (D2). Thus, it is intriguing that carriers of the Thr92Ala polymorphism in the D2 gene (DIO2) exhibit clinical improvement when liothyronine (LT3) is added to LT4 therapy. Here, we report that D2 is a cargo protein in ER Golgi intermediary compartment (ERGIC) vesicles, recycling between ER and Golgi. The Thr92-to-Ala substitution (Ala92-D2) caused ER stress and activated the unfolded protein response (UPR). Ala92-D2 accumulated in the trans-Golgi and generated less T3, which was restored by eliminating ER stress with the chemical chaperone 4-phenyl butyric acid (4-PBA). An Ala92-Dio2 polymorphism-carrying mouse exhibited UPR and hypothyroidism in distinct brain areas. The mouse refrained from physical activity, slept more, and required additional time to memorize objects. Enhancing T3 signaling in the brain with LT3 improved cognition, whereas restoring proteostasis with 4-PBA eliminated the Ala92-Dio2 phenotype. In contrast, primary hypothyroidism intensified the Ala92-Dio2 phenotype, with only partial response to LT4 therapy. Disruption of cellular proteostasis and reduced Ala92-D2 activity may explain the failure of LT4 therapy in carriers of Thr92Ala-DIO2.
Collapse
Affiliation(s)
- Sungro Jo
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
| | - Tatiana L. Fonseca
- Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Barbara M. L. C. Bocco
- Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Gustavo W. Fernandes
- Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Elizabeth A. McAninch
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
| | - Anaysa P. Bolin
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
- Department of Pharmacology, Biomedical Science Institute, University of São Paulo, and
| | - Rodrigo R. Da Conceição
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | | | - Daniele L. Ignacio
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
| | - Péter Egri
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Dorottya Németh
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Csaba Fekete
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Maria Martha Bernardi
- Graduate Program of Environmental and Experimental Pathology, Graduate Program of Dentistry, Universidade Paulista, São Paulo, SP, Brazil
| | - Victoria D. Leitch
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Naila S. Mannan
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Katharine F. Curry
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Natalie C. Butterfield
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - J.H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Miriam O. Ribeiro
- Developmental Disorders Program, Center of Biological Science and Health, Mackenzie Presbyterian University, São Paulo, SP, Brazil
| | - Antonio C. Bianco
- Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
40
|
Abstract
Treatment of hypothyroidism involves the endogenous conversion of thyroxine (T4) to 3,5,3'-triiodothyronine (T3) and may not be optimal in some cases when based on T4 alone. In the current issue of the JCI, Jo et al. present results that explain the reduced enzymatic activity of a common genetic variant of the enzyme responsible for this conversion, type 2 deiodinase (DIO2). The authors further explore the functional consequences of this variant on brain T3 activity, endoplasmic reticulum stress in glial cells, and cognitive function. These findings have important implications for the clinical treatment of hypothyroidism and for susceptibility to other neurological and metabolic diseases.
Collapse
|
41
|
Abstract
OBJECTIVE The aim of this study was to determine the prevalence and type of thyroid hormone levels alterations in patients with acute pancreatitis (AP) and analyze if variations are useful AP progression predictors. METHODS Three groups of patients were analyzed: AP patients (n = 90), abdominal pain patients (n = 30), and healthy control subjects (n = 40). Usual blood parameters for AP diagnosis and prognosis, thyroid-stimulating hormone (or thyrotropin), FT4 (free thyroxine), FT3 (free triiodothyronine), and TT3 (total triiodothyronine) levels were analyzed. RESULTS Thyroid hormone level alterations were detected only within the AP group (41% of total cases), being the reduction in T3 levels the most frequently detected deviation (15.6% of FT3 and 8.3% of TT3 cases). Alterations were not influenced by age or sex. Free thyroxine average values were also significantly higher in the AP group, compared with the healthy control group (P = 0.0005), resulting as independent predictors of both severity and mortality. Mortality in this group was 50%, with deceased patients showing FT4 levels above the reference limit. CONCLUSIONS Our results show that FT4 level determination during the initial clinical evaluation of patients admitted to the emergency service with AP can be included as a severity indicator to help determine the differential care of these cases.
Collapse
|
42
|
Nascimento BPP, Bocco BMLC, Fernandes GW, Fonseca TL, McAninch EA, Cardoso CV, Bondan EF, Nassif RJ, Cysneiros RM, Bianco AC, Ribeiro MO. Induction of Type 2 Iodothyronine Deiodinase After Status Epilepticus Modifies Hippocampal Gene Expression in Male Mice. Endocrinology 2018; 159:3090-3104. [PMID: 29905787 PMCID: PMC6669821 DOI: 10.1210/en.2018-00146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/06/2018] [Indexed: 11/19/2022]
Abstract
Status epilepticus (SE) is an abnormally prolonged seizure that results from either a failure of mechanisms that terminate seizures or from initiating mechanisms that inherently lead to prolonged seizures. Here we report that mice experiencing a 3 hours of SE caused by pilocarpine exhibit a rapid increase in expression of type 2 iodothyronine deiodinase gene (Dio2) and a decrease in the expression of type 3 iodothyronine deiodinase gene in hippocampus, amygdala and prefrontal cortex. Type 3 iodothyronine deiodinase in hippocampal sections was seen concentrated in the neuronal nuclei, typical of ischemic injury of the brain. An unbiased analysis of the hippocampal transcriptome of mice undergoing 3 hours of SE revealed a number of genes, including those involved with response to oxidative stress, cellular homeostasis, cell signaling, and mitochondrial structure. In contrast, in mice with targeted disruption of Dio2 in astrocytes (Astro D2KO mouse), the highly induced genes in the hippocampus were related to inflammation, apoptosis, and cell death. We propose that Dio2 induction caused by SE accelerates production of T3 in different areas of the central nervous system and modifies the hippocampal gene expression profile, affecting the balance between adaptive and maladaptive mechanisms.
Collapse
Affiliation(s)
- Bruna P P Nascimento
- Graduate Program of Translational Medicine, Department of Medicine, Federal University of São Paulo, São Paulo-SP, Brazil
- Developmental Disorders Program, Center of Biological Sciences and Health, Mackenzie Presbyterian University, São Paulo-SP, Brazil
| | - Barbara M L C Bocco
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Gustavo W Fernandes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Tatiana L Fonseca
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Elizabeth A McAninch
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Carolina V Cardoso
- Department of Environmental and Experimental Pathology, Paulista University, São Paulo-SP, Brazil
| | - Eduardo F Bondan
- Department of Environmental and Experimental Pathology, Paulista University, São Paulo-SP, Brazil
| | - Renata J Nassif
- Neuroscience Sector, Department of Neurology and Neurosurgery, Federal University of São Paulo, São Paulo-SP, Brazil
| | - Roberta M Cysneiros
- Developmental Disorders Program, Center of Biological Sciences and Health, Mackenzie Presbyterian University, São Paulo-SP, Brazil
| | - Antonio C Bianco
- Division of Endocrinology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Miriam O Ribeiro
- Graduate Program of Translational Medicine, Department of Medicine, Federal University of São Paulo, São Paulo-SP, Brazil
- Developmental Disorders Program, Center of Biological Sciences and Health, Mackenzie Presbyterian University, São Paulo-SP, Brazil
| |
Collapse
|
43
|
Bradley D, Liu J, Blaszczak A, Wright V, Jalilvand A, Needleman B, Noria S, Renton D, Hsueh W. Adipocyte DIO2 Expression Increases in Human Obesity but Is Not Related to Systemic Insulin Sensitivity. J Diabetes Res 2018; 2018:2464652. [PMID: 30116736 PMCID: PMC6079440 DOI: 10.1155/2018/2464652] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/25/2018] [Indexed: 11/17/2022] Open
Abstract
Deiodinase type II (D2), encoded by DIO2, catalyzes the conversion of T4 to bioactive T3. T3 not only stimulates adaptive thermogenesis but also affects adipose tissue (AT) lipid accumulation, mitochondrial function, inflammation, and potentially systemic metabolism. Although better defined in brown AT, the precise role of DIO2 expression in white AT remains largely unknown, with data derived only from whole fat. Therefore, the purpose of this study was to determine whether subcutaneous (SAT) and visceral (VAT) adipocyte-specific gene expression of DIO2 differs between obese and lean patients and whether these differences relate to alterations in mitochondrial function, fatty acid flux, inflammatory cytokines/adipokines, and ultimately insulin sensitivity. Accordingly, adipocytes of 73 obese and 21 lean subjects were isolated and subjected to gene expression analyses. Our results demonstrate that obese compared to lean human individuals have increased adipocyte-specific DIO2 expression in both SAT and VAT. Although higher DIO2 was strongly related to reduced fatty acid synthesis/oxidation and mitochondrial function, we found no relationship to proinflammatory cytokines or insulin resistance and no difference based on diabetic status. Our results suggest that adipocyte-derived DIO2 may play a role in weight maintenance but is likely not a major contributor to obesity-related insulin resistance.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Joey Liu
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Alecia Blaszczak
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Valerie Wright
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Anahita Jalilvand
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Bradley Needleman
- Center for Minimally Invasive Surgery, Department of General Surgery, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Sabrena Noria
- Center for Minimally Invasive Surgery, Department of General Surgery, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - David Renton
- Center for Minimally Invasive Surgery, Department of General Surgery, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Willa Hsueh
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| |
Collapse
|
44
|
Hippocampus-dependent memory and allele-specific gene expression in adult offspring of alcohol-consuming dams after neonatal treatment with thyroxin or metformin. Mol Psychiatry 2018; 23:1643-1651. [PMID: 28727687 PMCID: PMC5775940 DOI: 10.1038/mp.2017.129] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 02/06/2023]
Abstract
Fetal alcohol spectrum disorder (FASD), the result of fetal alcohol exposure (FAE), affects 2-11% of children worldwide, with no effective treatments. Hippocampus-based learning and memory deficits are key symptoms of FASD. Our previous studies show hypothyroxinemia and hyperglycemia of the alcohol-consuming pregnant rat, which likely affects fetal neurodevelopment. We administered vehicle, thyroxine (T4) or metformin to neonatal rats post FAE and rats were tested in the hippocampus-dependent contextual fear-conditioning paradigm in adulthood. Both T4 and metformin alleviated contextual fear memory deficit induced by FAE, and reversed the hippocampal expression changes in the thyroid hormone-inactivating enzyme, deiodinase-III (Dio3) and insulin-like growth factor 2 (Igf2), genes that are known to modulate memory processes. Neonatal T4 restored maternal allelic expressions of the imprinted Dio3 and Igf2 in the adult male hippocampus, while metformin restored FAE-caused changes in Igf2 expression only. The decreased hippocampal expression of DNA methyltransferase 1 (Dnmt1) that maintains the imprinting of Dio3 and Igf2 during development was normalized by both treatments. Administering Dnmt1 inhibitor to control neonates resulted in FAE-like deficits in fear memory and hippocampal allele-specific expression of Igf2, which were reversed by metformin. We propose that neonatal administration of T4 and metformin post FAE affect memory via elevating Dnmt1 and consequently normalizing hippocampal Dio3 and Igf2 expressions in the adult offspring. The present results indicate that T4 and metformin, administered during the neonatal period that is equivalent to the third trimester of human pregnancy, are potential treatments for FASD and conceivably for other neurodevelopmental disorders with cognitive deficits.
Collapse
|
45
|
Hernandez A, Stohn JP. The Type 3 Deiodinase: Epigenetic Control of Brain Thyroid Hormone Action and Neurological Function. Int J Mol Sci 2018; 19:ijms19061804. [PMID: 29921775 PMCID: PMC6032375 DOI: 10.3390/ijms19061804] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 12/31/2022] Open
Abstract
Thyroid hormones (THs) influence multiple processes in the developing and adult central nervous system, and their local availability needs to be maintained at levels that are tailored to the requirements of their biological targets. The local complement of TH transporters, deiodinase enzymes, and receptors is critical to ensure specific levels of TH action in neural cells. The type 3 iodothyronine deiodinase (DIO3) inactivates THs and is highly present in the developing and adult brain, where it limits their availability and action. DIO3 deficiency in mice results in a host of neurodevelopmental and behavioral abnormalities, demonstrating the deleterious effects of TH excess, and revealing the critical role of DIO3 in the regulation of TH action in the brain. The fact the Dio3 is an imprinted gene and that its allelic expression pattern varies across brain regions and during development introduces an additional level of control to deliver specific levels of hormone action in the central nervous system (CNS). The sensitive epigenetic nature of the mechanisms controlling the genomic imprinting of Dio3 renders brain TH action particularly susceptible to disruption due to exogenous treatments and environmental exposures, with potential implications for the etiology of human neurodevelopmental disorders.
Collapse
Affiliation(s)
- Arturo Hernandez
- Center for Molecular Medicine, Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, USA.
- Graduate School for Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA.
- Department of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA.
| | - J Patrizia Stohn
- Center for Molecular Medicine, Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, USA.
| |
Collapse
|
46
|
Thyroid Hormone Supplementation Restores Spatial Memory, Hippocampal Markers of Neuroinflammation, Plasticity-Related Signaling Molecules, and β-Amyloid Peptide Load in Hypothyroid Rats. Mol Neurobiol 2018; 56:722-735. [DOI: 10.1007/s12035-018-1111-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/09/2018] [Indexed: 10/16/2022]
|
47
|
Comparative Analysis of Zearalenone Effects on Thyroid Receptor Alpha (TRα) and Beta (TRβ) Expression in Rat Primary Cerebellar Cell Cultures. Int J Mol Sci 2018; 19:ijms19051440. [PMID: 29751674 PMCID: PMC5983839 DOI: 10.3390/ijms19051440] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/20/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022] Open
Abstract
Thyroid receptors play an important role in postnatal brain development. Zearalenone (ZEN), a major mycotoxin of Fusarium fungi, is well known to cause serious health problems in animals and humans through various mechanisms, including the physiological pathways of thyroid hormone (TH). In the present study, we aimed to investigate the expression of thyroid receptors α (TRα) and β (TRβ) in primary cerebellar neurons in the presence or absence of glia and following ZEN treatment, using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot. Primary cerebellar granule cells were treated with low doses of ZEN (0.1 nM) in combination with physiologically relevant concentrations of l-thyroxine (T4), 3,3′,5-triiodo-l-thyronine (T3) and 17β-estradiol (E2). Expression levels of TRα and TRβ at mRNA and protein levels were slightly modified by ZEN administered alone; however, along with thyroid and steroid hormones, modelling the physiological conditions, expression levels of TRs varied highly depending on the given treatment. Gene expression levels were also highly modulated by the presence or absence of glial cells, with mostly contrasting effects. Our results demonstrate divergent transcriptional and translational mechanisms involved in the expression of TRs implied by ZEN and hormonal milieu, as well as culturing conditions.
Collapse
|
48
|
Naicker M, Naidoo S. Expression of thyroid-stimulating hormone receptors and thyroglobulin in limbic regions in the adult human brain. Metab Brain Dis 2018; 33:481-489. [PMID: 28776278 DOI: 10.1007/s11011-017-0076-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/19/2017] [Indexed: 10/19/2022]
Abstract
Expression of the human thyroid-specific proteins, thyroid-stimulating hormone receptor (TSH-R) and thyroglobulin (TG) in non-thyroid tissue is well-documented. TSH-R has been identified in the heart, kidney, bone, pituitary, adipose tissue, skin and astrocyte cultures. TG has been identified in the skin, thymus and kidney. However, none of those previous studies had identified TSH-R or TG in specific human brain regions. Previously, a pilot study conducted by our group on normal adult human brain demonstrated TSH-R and TG in cortical neurons and cerebral vasculature, respectively, within various brain areas. In the present study, we extend this investigation of thyroid proteins specifically in limbic regions of normal human brain. Forensic human samples of amygdalae, cingulate gyrii, frontal cortices, hippocampii, hypothalamii, and thalamii were obtained from five individuals who had died of causes unrelated to head injury and had no evidence of brain disease or psychological abnormality. Tissues were probed with commercial polyclonal antibodies against human TSH-R and TG which resulted in the significant demonstration of neuronal TSH-R in all limbic regions examined. Other novel results demonstrated TG in vascular smooth muscle of all limbic regions and in some neurons. Finding thyroid proteins in limbic areas of the human brain is unique, and this study demonstrates that cerebro-limbic localisation of thyroid proteins may have potential roles in neuro-psycho-pharmacology.
Collapse
Affiliation(s)
- Meleshni Naicker
- Therapeutics and Medicines Management, Pharmaceutical Sciences, Nelson, R Mandela School of Medicine, University of KwaZulu-Natal, Private bag X7, Durban, 4001, South Africa.
| | - Strinivasen Naidoo
- Therapeutics and Medicines Management, Pharmaceutical Sciences, Nelson, R Mandela School of Medicine, University of KwaZulu-Natal, Private bag X7, Durban, 4001, South Africa
| |
Collapse
|
49
|
Calzà L, Baldassarro VA, Fernandez M, Giuliani A, Lorenzini L, Giardino L. Thyroid Hormone and the White Matter of the Central Nervous System: From Development to Repair. VITAMINS AND HORMONES 2018; 106:253-281. [DOI: 10.1016/bs.vh.2017.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
50
|
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
|