1
|
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
|
2
|
Tal T, Myhre O, Fritsche E, Rüegg J, Craenen K, Aiello-Holden K, Agrillo C, Babin PJ, Escher BI, Dirven H, Hellsten K, Dolva K, Hessel E, Heusinkveld HJ, Hadzhiev Y, Hurem S, Jagiello K, Judzinska B, Klüver N, Knoll-Gellida A, Kühne BA, Leist M, Lislien M, Lyche JL, Müller F, Colbourne JK, Neuhaus W, Pallocca G, Seeger B, Scharkin I, Scholz S, Spjuth O, Torres-Ruiz M, Bartmann K. New approach methods to assess developmental and adult neurotoxicity for regulatory use: a PARC work package 5 project. FRONTIERS IN TOXICOLOGY 2024; 6:1359507. [PMID: 38742231 PMCID: PMC11089904 DOI: 10.3389/ftox.2024.1359507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/18/2024] [Indexed: 05/16/2024] Open
Abstract
In the European regulatory context, rodent in vivo studies are the predominant source of neurotoxicity information. Although they form a cornerstone of neurotoxicological assessments, they are costly and the topic of ethical debate. While the public expects chemicals and products to be safe for the developing and mature nervous systems, considerable numbers of chemicals in commerce have not, or only to a limited extent, been assessed for their potential to cause neurotoxicity. As such, there is a societal push toward the replacement of animal models with in vitro or alternative methods. New approach methods (NAMs) can contribute to the regulatory knowledge base, increase chemical safety, and modernize chemical hazard and risk assessment. Provided they reach an acceptable level of regulatory relevance and reliability, NAMs may be considered as replacements for specific in vivo studies. The European Partnership for the Assessment of Risks from Chemicals (PARC) addresses challenges to the development and implementation of NAMs in chemical risk assessment. In collaboration with regulatory agencies, Project 5.2.1e (Neurotoxicity) aims to develop and evaluate NAMs for developmental neurotoxicity (DNT) and adult neurotoxicity (ANT) and to understand the applicability domain of specific NAMs for the detection of endocrine disruption and epigenetic perturbation. To speed up assay time and reduce costs, we identify early indicators of later-onset effects. Ultimately, we will assemble second-generation developmental neurotoxicity and first-generation adult neurotoxicity test batteries, both of which aim to provide regulatory hazard and risk assessors and industry stakeholders with robust, speedy, lower-cost, and informative next-generation hazard and risk assessment tools.
Collapse
Affiliation(s)
- Tamara Tal
- Helmholtz Centre for Environmental Research – UFZ, Chemicals in the Environment Research Section, Leipzig, Germany
- University of Leipzig, Medical Faculty, Leipzig, Germany
| | - Oddvar Myhre
- Norwegian Institute of Public Health – NIPH, Department of Chemical Toxicology, Oslo, Norway
| | - Ellen Fritsche
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
- DNTOX GmbH, Düsseldorf, Germany
- Swiss Centre for Applied Human Toxicology, University of Basel, Basel, Switzerland
| | - Joëlle Rüegg
- Uppsala University, Department of Organismal Biology, Uppsala, Sweden
| | - Kai Craenen
- European Chemicals Agency (ECHA), Helsinki, Finland
| | | | - Caroline Agrillo
- Uppsala University, Department of Organismal Biology, Uppsala, Sweden
| | - Patrick J. Babin
- Université de Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Maladies Rares: Génétique et Métabolisme (MRGM), Pessac, France
| | - Beate I. Escher
- Helmholtz Centre for Environmental Research – UFZ, Chemicals in the Environment Research Section, Leipzig, Germany
| | - Hubert Dirven
- Norwegian Institute of Public Health – NIPH, Department of Chemical Toxicology, Oslo, Norway
| | | | - Kristine Dolva
- University of Oslo, Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Olso, Norway
| | - Ellen Hessel
- Dutch Nation Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, Netherlands
| | - Harm J. Heusinkveld
- Dutch Nation Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, Netherlands
| | - Yavor Hadzhiev
- University of Birmingham, Centre for Environmental Research and Justice, Birmingham, UK
| | - Selma Hurem
- Norwegian University of Life Sciences (NMBU), Faculty of Veterinary Medicine, Ås, Norway
| | - Karolina Jagiello
- University of Gdansk, Laboratory of Environmental Chemoinformatics, Gdansk, Poland
| | - Beata Judzinska
- University of Gdansk, Laboratory of Environmental Chemoinformatics, Gdansk, Poland
| | - Nils Klüver
- Helmholtz Centre for Environmental Research – UFZ, Chemicals in the Environment Research Section, Leipzig, Germany
| | - Anja Knoll-Gellida
- Université de Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Maladies Rares: Génétique et Métabolisme (MRGM), Pessac, France
| | - Britta A. Kühne
- University of Veterinary Medicine Hannover, Foundation, Institute for Food Quality and Food Safety, Hannover, Germany
| | - Marcel Leist
- University of Konstanz, In Vitro Toxicology and Biomedicine/CAAT-Europe, Konstanz, Germany
| | - Malene Lislien
- Norwegian Institute of Public Health – NIPH, Department of Chemical Toxicology, Oslo, Norway
| | - Jan L. Lyche
- Norwegian University of Life Sciences (NMBU), Faculty of Veterinary Medicine, Ås, Norway
| | - Ferenc Müller
- University of Birmingham, Centre for Environmental Research and Justice, Birmingham, UK
| | - John K. Colbourne
- University of Birmingham, Centre for Environmental Research and Justice, Birmingham, UK
| | - Winfried Neuhaus
- AIT Austrian Institute of Technology GmbH, Competence Unit Molecular Diagnostics, Center Health and Bioresources, Vienna, Austria
- Danube Private University, Faculty of Dentistry and Medicine, Department of Medicine, Krems, Austria
| | - Giorgia Pallocca
- University of Konstanz, In Vitro Toxicology and Biomedicine/CAAT-Europe, Konstanz, Germany
| | - Bettina Seeger
- University of Veterinary Medicine Hannover, Foundation, Institute for Food Quality and Food Safety, Hannover, Germany
| | - Ilka Scharkin
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Stefan Scholz
- Helmholtz Centre for Environmental Research – UFZ, Chemicals in the Environment Research Section, Leipzig, Germany
| | - Ola Spjuth
- Uppsala University and Science for Life Laboratory, Department of Pharmaceutical Biosciences, Uppsala, Sweden
| | - Monica Torres-Ruiz
- Instituto de Salud Carlos III (ISCIII), Centro Nacional de Sanidad Ambiental (CNSA), Environmental Toxicology Unit, Majadahonda, Spain
| | - Kristina Bartmann
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
- DNTOX GmbH, Düsseldorf, Germany
| |
Collapse
|
3
|
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
|
4
|
Yang Y, Valdés-Rives SA, Liu Q, Li Y, Tan J, Tan Y, Koch CA, Rong Y, Houser SR, Wei S, Cai KQ, Cheng SY, Curran T, Wechsler-Reya R, Yang ZJ. Thyroid Hormone Suppresses Medulloblastoma Progression Through Promoting Terminal Differentiation of Tumor Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580111. [PMID: 38405864 PMCID: PMC10888774 DOI: 10.1101/2024.02.13.580111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Hypothyroidism is commonly detected in patients with medulloblastoma (MB). A possible link between thyroid hormone (TH) signaling and MB pathogenicity has not been reported. Here, we find that TH plays a critical role in promoting tumor cell differentiation. Reduction in TH levels frees the TH receptor, TRα1, to bind to EZH2 and repress expression of NeuroD1, a transcription factor that drives tumor cell differentiation. Increased TH reverses EZH2-mediated repression of NeuroD1 by abrogating the binding of EZH2 and TRα1, thereby stimulating tumor cell differentiation and reducing MB growth. Importantly, TH-induced differentiation of tumor cells is not restricted by the molecular subgroup of MB. These findings establish an unprecedented association between TH signaling and MB pathogenicity, providing solid evidence for TH as a promising modality for MB treatment.
Collapse
|
5
|
de Souza JS. Thyroid hormone biosynthesis and its role in brain development and maintenance. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 142:329-365. [PMID: 39059990 DOI: 10.1016/bs.apcsb.2023.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Thyroid hormones are critical modulators in the physiological processes necessary to virtually all tissues, with exceptionally fundamental roles in brain development and maintenance. These hormones regulate essential neurodevelopment events, including neuronal migration, synaptogenesis, and myelination. Additionally, thyroid hormones are crucial for maintaining brain homeostasis and cognitive function in adulthood. This chapter aims to offer a comprehensive understanding of thyroid hormone biosynthesis and its intricate role in brain physiology. Here, we described the mechanisms underlying the biosynthesis of thyroid hormones, their influence on various aspects of brain development and ongoing maintenance, and the proteins in the brain that are responsive to these hormones. This chapter was geared towards broadening our understanding of thyroid hormone action in the brain, shedding light on potential therapeutic targets for neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Janaina Sena de Souza
- Department of Pediatrics and Cellular & Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
6
|
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
|
7
|
Ren J, Flamant F. Thyroid hormone as a temporal switch in mouse development. Eur Thyroid J 2023; 12:e220225. [PMID: 36715693 PMCID: PMC10083660 DOI: 10.1530/etj-22-0225] [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: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 01/31/2023] Open
Abstract
Thyroid hormones are known to trigger metamorphosis in an amphibian. This review discusses the hypothesis according to which they act in a similar manner to synchronize the post-natal development of mice, using brain, brown adipose tissue, and heart as examples.
Collapse
Affiliation(s)
- Juan Ren
- ENS de Lyon, INRAE, CNRS, Institut de Génomique Fonctionnelle de Lyon, Lyon, France
| | - Frédéric Flamant
- ENS de Lyon, INRAE, CNRS, Institut de Génomique Fonctionnelle de Lyon, Lyon, France
| |
Collapse
|
8
|
Murolo M, Di Vincenzo O, Cicatiello AG, Scalfi L, Dentice M. Cardiovascular and Neuronal Consequences of Thyroid Hormones Alterations in the Ischemic Stroke. Metabolites 2022; 13:metabo13010022. [PMID: 36676947 PMCID: PMC9863748 DOI: 10.3390/metabo13010022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Ischemic stroke is one of the leading global causes of neurological morbidity and decease. Its etiology depends on multiple events such as cardiac embolism, brain capillaries occlusion and atherosclerosis, which ultimately culminate in blood flow interruption, incurring hypoxia and nutrient deprivation. Thyroid hormones (THs) are pleiotropic modulators of several metabolic pathways, and critically influence different aspects of tissues development. The brain is a key TH target tissue and both hypo- and hyperthyroidism, during embryonic and adult life, are associated with deranged neuronal formation and cognitive functions. Accordingly, increasing pieces of evidence are drawing attention on the consistent relationship between the THs status and the acute cerebral and cardiac diseases. However, the concrete contribution of THs systemic or local alteration to the pathology outcome still needs to be fully addressed. In this review, we aim to summarize the multiple influences that THs exert on the brain and heart patho-physiology, to deepen the reasons for the harmful effects of hypo- and hyperthyroidism on these organs and to provide insights on the intricate relationship between the THs variations and the pathological alterations that take place after the ischemic injury.
Collapse
Affiliation(s)
- Melania Murolo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
- Correspondence:
| | - Olivia Di Vincenzo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy
| | | | - Luca Scalfi
- Department of Public Health, University of Naples “Federico II”, 80131 Naples, Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
- CEINGE-Biotecnologie Avanzate S.c.a.r.l., 80131 Naples, Italy
| |
Collapse
|
9
|
Dubois E, Galindo AN, Dayon L, Cominetti O. Assessing normalization methods in mass spectrometry-based proteome profiling of clinical samples. Biosystems 2022; 215-216:104661. [PMID: 35247480 DOI: 10.1016/j.biosystems.2022.104661] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 02/21/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Large-scale proteomic studies have to deal with unwanted variability, especially when samples originate from different centers and multiple analytical batches are needed. Such variability is typically added throughout all the steps of a clinical research study, from human biological sample collection and storage, sample preparation, spectral data acquisition, to peptide and protein quantification. In order to remove such diverse and unwanted variability, normalization of the protein data is performed. There have been already several published reviews comparing normalization methods in the -omics field, but reports focusing on proteomic data generated with mass spectrometry (MS) are much fewer. Additionally, most of these reports have only dealt with small datasets. RESULTS As a case study, here we focused on the normalization of a large MS-based proteomic dataset obtained from an overweight and obese pan-European cohort, where different normalization methods were evaluated, namely: center standardize, quantile protein, quantile sample, global standardization, ComBat, median centering, mean centering, single standard and removal of unwanted variation (RUV); some of these are generic normalization methods while others have been specifically created to deal with genomic or metabolomic data. We checked how relationships between proteins and clinical variables (e.g., gender, levels of triglycerides or cholesterol) were improved after normalizing the data with the different methods. CONCLUSIONS Some normalization methods were better adapted for this particular large-scale shotgun proteomic dataset of human plasma samples labeled with isobaric tags and analyzed with liquid chromatography-tandem MS. In particular, quantile sample normalization, RUV, mean and median centering showed very good performance, while quantile protein normalization provided worse results than those obtained with unnormalized data.
Collapse
Affiliation(s)
- Etienne Dubois
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, EPFL Innovation Park, 1015, Lausanne, Switzerland
| | - Antonio Núñez Galindo
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, EPFL Innovation Park, 1015, Lausanne, Switzerland
| | - Loïc Dayon
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, EPFL Innovation Park, 1015, Lausanne, Switzerland; Chemistry and Chemical Engineering Section, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Ornella Cominetti
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, EPFL Innovation Park, 1015, Lausanne, Switzerland.
| |
Collapse
|
10
|
Tarai A, Li Y, Liu B, Zhang D, Li J, Yan W, Zhang J, Qu J, Yang Z. A review on recognition of tri-/tetra-analyte by using simple organic colorimetric and fluorometric probes. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
11
|
Martinez ME, Hernandez A. The Type 3 Deiodinase Is a Critical Modulator of Thyroid Hormone Sensitivity in the Fetal Brain. Front Neurosci 2021; 15:703730. [PMID: 34248495 PMCID: PMC8265566 DOI: 10.3389/fnins.2021.703730] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/01/2021] [Indexed: 11/13/2022] Open
Abstract
Thyroid hormones (TH) are critical for the development and function of the central nervous system (CNS). Although their effects on the rodent brain peak within 2-3 weeks postnatally, the fetal brain has been found largely insensitive to exogenously administrated TH. To address this issue, here we examined gene expression in brains from mouse fetuses deficient in the type 3 deiodinase (DIO3), the selenoenzyme responsible for clearing TH. At embryonic day E18.5 qPCR determinations indicated a marked increase in the mRNA expression of T3-responsive genes Klf9 and Nrgn. The increased expression of these genes was confirmed by in situ hydridization in multiple areas of the cortex and in the striatum. RNA sequencing revealed 246 genes differentially expressed (70% up-regulated) in the brain of E18.5 Dio3-/- male fetuses. Differential expression of 13 of these genes was confirmed in an extended set of samples that included females. Pathway analyses of differentially expressed genes indicated enrichment in glycolysis and signaling related to axonal guidance, synaptogenesis and hypoxia inducible factor alpha. Additional RNA sequencing identified 588 genes differentially expressed (35% up-regulated) in the brain of E13.5 Dio3-/- male fetuses. Differential expression of 13 of these genes, including Klf9, Hr, and Mgp, was confirmed in an extended set of samples including females. Although pathway analyses of differentially expressed genes at E13.5 also revealed significant enrichment in axonal guidance and synaptogenesis signaling, top enrichment was found for functions related to the cell cycle, aryl hydrocarbon receptor signaling, PCP and kinetochore metaphase signaling pathways and mitotic roles of polo-like kinase. Differential expression at E13.5 was confirmed by qPCR for additional genes related to collagen and extracellular matrix and for selected transcription factors. Overall, our results demonstrate that the rodent fetal brain is sensitive to TH as early as E13.5 of gestational age, and suggest that TH distinctly affects brain developmental programs in early and late gestation. We conclude that DIO3 function is critical to ensure an adequate timing for TH action in the developing brain and is probably the main factor underlying the lack of effects on the fetal brain observed in previous studies after TH administration.
Collapse
Affiliation(s)
- Maria Elena Martinez
- Center for Molecular Medicine, Maine Medical Center Research Institute, MaineHealth, Scarborough, ME, United States
| | - Arturo Hernandez
- Center for Molecular Medicine, Maine Medical Center Research Institute, MaineHealth, Scarborough, ME, United States.,Graduate School for Biomedical Science and Engineering, University of Maine, Orono, ME, United States.,Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
12
|
Marty S, Beekhuijzen M, Charlton A, Hallmark N, Hannas BR, Jacobi S, Melching-Kollmuss S, Sauer UG, Sheets LP, Strauss V, Urbisch D, Botham PA, van Ravenzwaay B. Towards a science-based testing strategy to identify maternal thyroid hormone imbalance and neurodevelopmental effects in the progeny - part II: how can key events of relevant adverse outcome pathways be addressed in toxicological assessments? Crit Rev Toxicol 2021; 51:328-358. [PMID: 34074207 DOI: 10.1080/10408444.2021.1910625] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The current understanding of thyroid-related adverse outcome pathways (AOPs) with adverse neurodevelopmental outcomes in mammals has been reviewed. This served to establish if standard rodent toxicity test methods and in vitro assays allow identifying thyroid-related modes-of-action potentially leading to adverse neurodevelopmental outcomes, and the human relevance of effects - in line with the European Commission's Endocrine Disruptor Criteria. The underlying hypothesis is that an understanding of the key events of relevant AOPs provides insight into differences in incidence, magnitude, or species sensitivity of adverse outcomes. The rodent studies include measurements of serum thyroid hormones, thyroid gland pathology and neurodevelopmental assessments, but do not directly inform on specific modes-of-action. Opportunities to address additional non-routine parameters reflecting critical events of AOPs in toxicological assessments are presented. These parameters appear relevant to support the identification of specific thyroid-related modes-of-action, provided that prevailing technical limitations are overcome. Current understanding of quantitative key event relationships is often weak, but would be needed to determine if the triggering of a molecular initiating event will ultimately result in an adverse outcome. Also, significant species differences in all processes related to thyroid hormone signalling are evident, but the biological implications thereof (including human relevance) are often unknown. In conclusion, careful consideration of the measurement (e.g. timing, method) and interpretation of additional non-routine parameters is warranted. These findings will be used in a subsequent paper to propose a testing strategy to identify if a substance may elicit maternal thyroid hormone imbalance and potentially also neurodevelopmental effects in the progeny.
Collapse
Affiliation(s)
- Sue Marty
- The Dow Chemical Company, Midland, MI, USA
| | | | | | | | | | | | | | - Ursula G Sauer
- Scientific Consultancy - Animal Welfare, Neubiberg, Germany
| | | | | | | | | | | |
Collapse
|
13
|
Abstract
Thyroid hormone is essential for brain development and brain function in the adult. During development, thyroid hormone acts in a spatial and temporal-specific manner to regulate the expression of genes essential for normal neural cell differentiation, migration, and myelination. In the adult brain, thyroid hormone is important for maintaining normal brain function. Thyroid hormone excess, hyperthyroidism, and thyroid hormone deficiency, hypothyroidism, are associated with disordered brain function, including depression, memory loss, impaired cognitive function, irritability, and anxiety. Adequate thyroid hormone levels are required for normal brain function. Thyroid hormone acts through a cascade of signaling components: activation and inactivation by deiodinase enzymes, thyroid hormone membrane transporters, and nuclear thyroid hormone receptors. Additionally, the hypothalamic-pituitary-thyroid axis, with negative feedback of thyroid hormone on thyrotropin-releasing hormone (TRH) and thyroid-stimulating hormone (TSH) secretion, regulates serum thyroid hormone levels in a narrow range. Animal and human studies have shown both systemic and local reduction in thyroid hormone availability in neurologic disease and after brain trauma. Treatment with thyroid hormone and selective thyroid hormone analogs has resulted in a reduction in injury and improved recovery. This article will describe the thyroid hormone signal transduction pathway in the brain and the role of thyroid hormone in the aging brain, neurologic diseases, and the protective role when administered after traumatic brain injury. © 2021 American Physiological Society. Compr Physiol 11:1-21, 2021.
Collapse
Affiliation(s)
- Yan-Yun Liu
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA.,Departments of Medicine and Physiology, Endocrinology, Diabetes and Metabolism Division, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Gregory A Brent
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA.,Departments of Medicine and Physiology, Endocrinology, Diabetes and Metabolism Division, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
14
|
Natural Autoimmunity to the Thyroid Hormone Monocarboxylate Transporters MCT8 and MCT10. Biomedicines 2021; 9:biomedicines9050496. [PMID: 33946552 PMCID: PMC8147215 DOI: 10.3390/biomedicines9050496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/24/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
The monocarboxylate transporters 8 (MCT8) and 10 (MCT10) are important for thyroid hormone (TH) uptake and signaling. Reduced TH activity is associated with impaired development, weight gain and discomfort. We hypothesized that autoantibodies (aAb) to MCT8 or MCT10 are prevalent in thyroid disease and obesity. Analytical tests for MCT8-aAb and MCT10-aAb were developed and characterized with commercial antiserum. Serum samples from healthy controls, thyroid patients and young overweight subjects were analyzed, and prevalence of the aAb was compared. MCT8-aAb were additionally tested for biological effects on thyroid hormone uptake in cell culture. Positive MCT8-aAb and MCT10-aAb were detected in all three clinical cohorts analyzed. MCT8-aAb were most prevalent in thyroid patients (11.9%) as compared to healthy controls (3.8%) and overweight adolescents (4.2%). MCT8-aAb positive serum reduced T4 uptake in cell culture in comparison to MCT8-aAb negative control serum. Prevalence of MCT10-aAb was highest in the group of thyroid patients as compared to healthy subjects or overweight adolescents (9.0% versus 4.5% and 6.3%, respectively). We conclude that MCT8 and MCT10 represent autoantigens in humans, and that MCT8-aAb may interfere with regular TH uptake and signaling. The increased prevalence of MCT8-aAb and MCT10-aAb in thyroid disease suggests that their presence may be of pathophysiological relevance. This hypothesis deserves an analysis in large prospective studies.
Collapse
|
15
|
Pagnin M, Kondos-Devcic D, Chincarini G, Cumberland A, Richardson SJ, Tolcos M. Role of thyroid hormones in normal and abnormal central nervous system myelination in humans and rodents. Front Neuroendocrinol 2021; 61:100901. [PMID: 33493504 DOI: 10.1016/j.yfrne.2021.100901] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/07/2021] [Accepted: 01/16/2021] [Indexed: 12/13/2022]
Abstract
Thyroid hormones (THs) are instrumental in promoting the molecular mechanisms which underlie the complex nature of neural development and function within the central nervous system (CNS) in vertebrates. The key neurodevelopmental process of myelination is conserved between humans and rodents, of which both experience peak fetal TH concentrations concomitant with onset of myelination. The importance of supplying adequate levels of THs to the myelin producing cells, the oligodendrocytes, for promoting their maturation is crucial for proper neural function. In this review we examine the key TH distributor and transport proteins, including transthyretin (TTR) and monocarboxylate transporter 8 (MCT8), essential for supporting proper oligodendrocyte and myelin health; and discuss disorders with impaired TH signalling in relation to abnormal CNS myelination in humans and rodents. Furthermore, we explore the importance of using novel TH analogues in the treatment of myelination disorders associated with abnormal TH signalling.
Collapse
Affiliation(s)
- Maurice Pagnin
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Delphi Kondos-Devcic
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Ginevra Chincarini
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Angela Cumberland
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | | | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia.
| |
Collapse
|
16
|
McNerney C, Johnston RJ. Thyroid hormone signaling specifies cone photoreceptor subtypes during eye development: Insights from model organisms and human stem cell-derived retinal organoids. VITAMINS AND HORMONES 2021; 116:51-90. [PMID: 33752828 DOI: 10.1016/bs.vh.2021.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Cones are the color-detecting photoreceptors of the vertebrate eye. Cones are specialized into subtypes whose functions are determined by the expression of color-sensitive opsin proteins. Organisms differ greatly in the number and patterning of cone subtypes. Despite these differences, thyroid hormone is an important regulator of opsin expression in most vertebrates. In this chapter, we outline how the timing of thyroid hormone signaling controls cone subtype fates during retinal development. We first examine our current understanding of cone subtype specification in model organisms and then describe advances in human stem cell-derived organoid technology that identified mechanisms controlling development of the human retina.
Collapse
Affiliation(s)
- Christina McNerney
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
17
|
Jonklaas J, Bianco AC, Cappola AR, Celi FS, Fliers E, Heuer H, McAninch EA, Moeller LC, Nygaard B, Sawka AM, Watt T, Dayan CM. Evidence-Based Use of Levothyroxine/Liothyronine Combinations in Treating Hypothyroidism: A Consensus Document. Eur Thyroid J 2021; 10:10-38. [PMID: 33777817 PMCID: PMC7983670 DOI: 10.1159/000512970] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Fourteen clinical trials have not shown a consistent benefit of combination therapy with levothyroxine (LT4) and liothyronine (LT3). Despite the publication of these trials, combination therapy is widely used and patients reporting benefit continue to generate patient and physician interest in this area. Recent scientific developments may provide insight into this inconsistency and guide future studies. METHODS The American Thyroid Association (ATA), British Thyroid Association (BTA), and European Thyroid Association (ETA) held a joint conference on November 3, 2019 (live-streamed between Chicago and London) to review new basic science and clinical evidence regarding combination therapy with presentations and input from 12 content experts. After the presentations, the material was synthesized and used to develop Summary Statements of the current state of knowledge. After review and revision of the material and Summary Statements, there was agreement that there was equipoise for a new clinical trial of combination therapy. Consensus Statements encapsulating the implications of the material discussed with respect to the design of future clinical trials of LT4/LT3 combination therapy were generated. Authors voted upon the Consensus Statements. Iterative changes were made in several rounds of voting and after comments from ATA/BTA/ETA members. RESULTS Of 34 Consensus Statements available for voting, 28 received at least 75% agreement, with 13 receiving 100% agreement. Those with 100% agreement included studies being powered to study the effect of deiodinase and thyroid hormone transporter polymorphisms on study outcomes, inclusion of patients dissatisfied with their current therapy and requiring at least 1.2 µg/kg of LT4 daily, use of twice daily LT3 or preferably a slow-release preparation if available, use of patient-reported outcomes as a primary outcome (measured by a tool with both relevant content validity and responsiveness) and patient preference as a secondary outcome, and utilization of a randomized placebo-controlled adequately powered double-blinded parallel design. The remaining statements are presented as potential additional considerations. DISCUSSION This article summarizes the areas discussed and presents Consensus Statements to guide development of future clinical trials of LT4/LT3 combination therapy. The results of such redesigned trials are expected to be of benefit to patients and of value to inform future thyroid hormone replacement clinical practice guidelines treatment recommendations.
Collapse
Affiliation(s)
- Jacqueline Jonklaas
- Division of Endocrinology, Georgetown University, Washington, District of Columbia, USA
- *Jacqueline Jonklaas, Division of Endocrinology, Georgetown University, 4000 Reservoir Road, NW, Washington, DC 20007 (USA),
| | - Antonio C. Bianco
- Section of Adult and Pediatric Endocrinology and Metabolism, University of Chicago, Chicago, Illinois, USA
| | - Anne R. Cappola
- Division of Endocrinology, Diabetes, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Francesco S. Celi
- Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Duisburg-Essen, Essen, Germany
| | | | - Lars C. Moeller
- Department of Endocrinology, Diabetes and Metabolism, University Duisburg-Essen, Essen, Germany
| | - Birte Nygaard
- Center for Endocrinology and Metabolism, Department of Internal Medicine, Herlev and Gentofte Hospitals, Herlev, Denmark
| | - Anna M. Sawka
- Division of Endocrinology, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Torquil Watt
- Department of Endocrinology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Colin M. Dayan
- Thyroid Research Group, School of Medicine, Cardiff University, Cardiff, United Kingdom
- **Colin M. Dayan, Thyroid Research Group, School of Medicine, Cardiff University, C2 Link, Heath Park, Cardiff CF14 4XN (UK),
| |
Collapse
|
18
|
Aksoy O, Pencik J, Hartenbach M, Moazzami AA, Schlederer M, Balber T, Varady A, Philippe C, Baltzer PA, Mazumder B, Whitchurch JB, Roberts CJ, Haitel A, Herac M, Susani M, Mitterhauser M, Marculescu R, Stangl‐Kremser J, Hassler MR, Kramer G, Shariat SF, Turner SD, Tichy B, Oppelt J, Pospisilova S, Hartenbach S, Tangermann S, Egger G, Neubauer HA, Moriggl R, Culig Z, Greiner G, Hoermann G, Hacker M, Heery DM, Merkel O, Kenner L. Thyroid and androgen receptor signaling are antagonized by μ-Crystallin in prostate cancer. Int J Cancer 2021; 148:731-747. [PMID: 33034050 PMCID: PMC7756625 DOI: 10.1002/ijc.33332] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/02/2020] [Indexed: 12/29/2022]
Abstract
Androgen deprivation therapy (ADT) remains a key approach in the treatment of prostate cancer (PCa). However, PCa inevitably relapses and becomes ADT resistant. Besides androgens, there is evidence that thyroid hormone thyroxine (T4) and its active form 3,5,3'-triiodo-L-thyronine (T3) are involved in the progression of PCa. Epidemiologic evidences show a higher incidence of PCa in men with elevated thyroid hormone levels. The thyroid hormone binding protein μ-Crystallin (CRYM) mediates intracellular thyroid hormone action by sequestering T3 and blocks its binding to cognate receptors (TRα/TRβ) in target tissues. We show in our study that low CRYM expression levels in PCa patients are associated with early biochemical recurrence and poor prognosis. Moreover, we found a disease stage-specific expression of CRYM in PCa. CRYM counteracted thyroid and androgen signaling and blocked intracellular choline uptake. CRYM inversely correlated with [18F]fluoromethylcholine (FMC) levels in positron emission tomography/magnetic resonance imaging of PCa patients. Our data suggest CRYM as a novel antagonist of T3- and androgen-mediated signaling in PCa. The role of CRYM could therefore be an essential control mechanism for the prevention of aggressive PCa growth.
Collapse
Affiliation(s)
- Osman Aksoy
- Department of PathologyMedical University ViennaViennaAustria
| | - Jan Pencik
- Department of PathologyMedical University ViennaViennaAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
- Present address: Jan Pencik, Molecular and Cell Biology LaboratoryThe Salk Institute for Biological StudiesLa JollaCaliforniaUSA
| | - Markus Hartenbach
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
| | - Ali A. Moazzami
- Department of Molecular Sciences, Uppsala BioCenterSwedish University of Agricultural SciencesUppsalaSweden
| | | | - Theresa Balber
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
- Ludwig Boltzmann Institute Applied DiagnosticsViennaAustria
- Department for Pharmaceutical Technology and BiopharmaceuticsUniversity of ViennaViennaAustria
| | - Adam Varady
- Department of PathologyMedical University ViennaViennaAustria
| | - Cecile Philippe
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
| | - Pascal A. Baltzer
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
| | | | | | | | - Andrea Haitel
- Department of PathologyMedical University ViennaViennaAustria
| | - Merima Herac
- Department of PathologyMedical University ViennaViennaAustria
| | - Martin Susani
- Department of PathologyMedical University ViennaViennaAustria
| | - Markus Mitterhauser
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
- Ludwig Boltzmann Institute Applied DiagnosticsViennaAustria
| | - Rodrig Marculescu
- Department of Laboratory MedicineMedical University ViennaViennaAustria
| | | | | | - Gero Kramer
- Department of UrologyMedical University ViennaViennaAustria
| | - Shahrokh F. Shariat
- Department of UrologyMedical University ViennaViennaAustria
- Division of Urology, Department of Special SurgeryJordan University Hospital, The University of JordanAmmanJordan
- Institute for Urology and Reproductive HealthSechenov UniversityMoscowRussia
- Departments of UrologyWeill Cornell Medical CollegeNew YorkNew YorkUSA
- Department of UrologyUniversity of Texas SouthwesternDallasTexasUSA
- Department of Urology, Second Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Suzanne D. Turner
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of CambridgeCambridgeUK
- Center of Molecular Medicine, Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Boris Tichy
- Center of Molecular Medicine, Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Jan Oppelt
- Center of Molecular Medicine, Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Sarka Pospisilova
- Center of Molecular Medicine, Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Sabrina Hartenbach
- Histo Consulting Inc.UlmGermany
- Department of PathologyRudolfinerhaus Privatklinik GmbhViennaAustria
| | - Simone Tangermann
- Unit for Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
| | - Gerda Egger
- Department of PathologyMedical University ViennaViennaAustria
- Ludwig Boltzmann Institute Applied DiagnosticsViennaAustria
| | - Heidi A. Neubauer
- Institute of Animal Breeding and GeneticsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Richard Moriggl
- Institute of Animal Breeding and GeneticsUniversity of Veterinary Medicine ViennaViennaAustria
| | - Zoran Culig
- Department of UrologyInnsbruck Medical UniversityInnsbruckAustria
| | - Georg Greiner
- Department of Laboratory MedicineMedical University ViennaViennaAustria
| | - Gregor Hoermann
- Department of Laboratory MedicineMedical University ViennaViennaAustria
- MLL Munich Leukemia LaboratoryMunichGermany
| | - Marcus Hacker
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
- Department of Biomedical Imaging and Image Guided TherapyMedical University ViennaViennaAustria
| | | | - Olaf Merkel
- Department of PathologyMedical University ViennaViennaAustria
| | - Lukas Kenner
- Department of PathologyMedical University ViennaViennaAustria
- Center for Biomarker Research in Medicine (CBmed)GrazAustria
- Unit for Laboratory Animal PathologyUniversity of Veterinary Medicine ViennaViennaAustria
- Christian Doppler Laboratory for Applied Metabolomics (CDL‐AM)Medical University of ViennaViennaAustria
| |
Collapse
|
19
|
Jonklaas J, Bianco AC, Cappola AR, Celi FS, Fliers E, Heuer H, McAninch EA, Moeller LC, Nygaard B, Sawka AM, Watt T, Dayan CM. Evidence-Based Use of Levothyroxine/Liothyronine Combinations in Treating Hypothyroidism: A Consensus Document. Thyroid 2021; 31:156-182. [PMID: 33276704 PMCID: PMC8035928 DOI: 10.1089/thy.2020.0720] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: Fourteen clinical trials have not shown a consistent benefit of combination therapy with levothyroxine (LT4) and liothyronine (LT3). Despite the publication of these trials, combination therapy is widely used and patients reporting benefit continue to generate patient and physician interest in this area. Recent scientific developments may provide insight into this inconsistency and guide future studies. Methods: The American Thyroid Association (ATA), British Thyroid Association (BTA), and European Thyroid Association (ETA) held a joint conference on November 3, 2019 (live-streamed between Chicago and London) to review new basic science and clinical evidence regarding combination therapy with presentations and input from 12 content experts. After the presentations, the material was synthesized and used to develop Summary Statements of the current state of knowledge. After review and revision of the material and Summary Statements, there was agreement that there was equipoise for a new clinical trial of combination therapy. Consensus Statements encapsulating the implications of the material discussed with respect to the design of future clinical trials of LT4/LT3 combination therapy were generated. Authors voted upon the Consensus Statements. Iterative changes were made in several rounds of voting and after comments from ATA/BTA/ETA members. Results: Of 34 Consensus Statements available for voting, 28 received at least 75% agreement, with 13 receiving 100% agreement. Those with 100% agreement included studies being powered to study the effect of deiodinase and thyroid hormone transporter polymorphisms on study outcomes, inclusion of patients dissatisfied with their current therapy and requiring at least 1.2 μg/kg of LT4 daily, use of twice daily LT3 or preferably a slow-release preparation if available, use of patient-reported outcomes as a primary outcome (measured by a tool with both relevant content validity and responsiveness) and patient preference as a secondary outcome, and utilization of a randomized placebo-controlled adequately powered double-blinded parallel design. The remaining statements are presented as potential additional considerations. Discussion: This article summarizes the areas discussed and presents Consensus Statements to guide development of future clinical trials of LT4/LT3 combination therapy. The results of such redesigned trials are expected to be of benefit to patients and of value to inform future thyroid hormone replacement clinical practice guidelines treatment recommendations.
Collapse
Affiliation(s)
- Jacqueline Jonklaas
- Division of Endocrinology, Georgetown University, Washington, District of Columbia, USA
- Address correspondence to: Jacqueline Jonklaas, MD, PhD, Division of Endocrinology, Georgetown University, 4000 Reservoir Road, NW, Washington, DC 20007, USA
| | - Antonio C. Bianco
- Section of Adult and Pediatric Endocrinology and Metabolism, University of Chicago, Chicago, Illinois, USA
| | - Anne R. Cappola
- Division of Endocrinology, Diabetes, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Francesco S. Celi
- Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Duisburg-Essen, Essen, Germany
| | | | - Lars C. Moeller
- Department of Endocrinology, Diabetes and Metabolism, University Duisburg-Essen, Essen, Germany
| | - Birte Nygaard
- Center for Endocrinology and Metabolism, Department Internal Medicine, Herlev and Gentofte Hospitals, Herlev, Denmark
| | - Anna M. Sawka
- Division of Endocrinology, University Health Network and University of Toronto, Toronto, Canada
| | - Torquil Watt
- Department of Endocrinology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Colin M. Dayan
- Thyroid Research Group, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Address correspondence to: Colin M. Dayan, MD, PhD, Thyroid Research Group, School of Medicine, Cardiff University, C2 Link, Heath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
20
|
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
|
21
|
Bae HS, Jin YK, Ham S, Kim HK, Shin H, Cho GB, Lee KJ, Lee H, Kim KM, Koo OJ, Jang G, Lee JM, Lee JY. CRISRP/Cas9-mediated knockout of Mct8 reveals a functional involvement of Mct8 in testis and sperm development in a rat. Sci Rep 2020; 10:11148. [PMID: 32636400 PMCID: PMC7341756 DOI: 10.1038/s41598-020-67594-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 06/07/2020] [Indexed: 11/17/2022] Open
Abstract
Thyroid hormone (TH) has long been believed to play a minor role in male reproduction. However, evidences from experimental model of thyrotoxicosis or hypothyroidism suggests its role in spermatogenesis. Cellular action of TH requires membrane transport via specific transporters such as monocarboxylate transporter 8 (MCT8). SLC16A2 (encodes for MCT8) inactivating mutation in humans can lead to Allan-Herndon Dudley-syndrome, a X-linked psychomotor and growth retardation. These patients present cryptorchidism which suggests a role of MCT8 during spermatogenesis. In this study, we found that Mct8 is highly expressed during early postnatal development and decreases its expression in the adulthood of testis of wild-type male rats. Histological analysis revealed that spermatogonia largely lacks MCT8 expression while spermatocytes and maturing spermatids highly express MCT8. To further understand the role of Mct8 during spermatogenesis, we generated Slc16a2 (encodes MCT8) knockout rats using CRISPR/Cas9. Serum THs (T3 and T4) level were significantly altered in Slc16a2 knockout rats when compared to wild-type littermates during early to late postnatal development. Unlike Slc16a2 knockout mice, Slc16a2 knockout rats showed growth delay during early to late postnatal development. In adult Slc16a2 knockout rats, we observed reduced sperm motility and viability. Collectively, our data unveil a functional involvement of MCT8 in spermatogenesis, underscoring the importance of TH signaling and action during spermatogenesis.
Collapse
Affiliation(s)
- Hee Sook Bae
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Yun-Kyeong Jin
- Laboratory of Theriogenology, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Sangwoo Ham
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Hee Kyoung Kim
- Laboratory of Theriogenology, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyejung Shin
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Gyu-Bon Cho
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Kyu Jun Lee
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Hohyeon Lee
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Kyeong-Min Kim
- Laboratory of Theriogenology, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Ok-Jae Koo
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Goo Jang
- Laboratory of Theriogenology, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Jung Min Lee
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea.,School of Life Science, Handong Global University, Pohang, 37554, South Korea
| | - Jae Young Lee
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea.
| |
Collapse
|
22
|
Skitchenko RK, Usoltsev D, Uspenskaya M, Kajava AV, Guskov A. Census of halide-binding sites in protein structures. Bioinformatics 2020; 36:3064-3071. [PMID: 32022861 PMCID: PMC7214031 DOI: 10.1093/bioinformatics/btaa079] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 12/02/2022] Open
Abstract
Motivation Halides are negatively charged ions of halogens, forming fluorides (F−), chlorides (Cl−), bromides (Br−) and iodides (I−). These anions are quite reactive and interact both specifically and non-specifically with proteins. Despite their ubiquitous presence and important roles in protein function, little is known about the preferences of halides binding to proteins. To address this problem, we performed the analysis of halide–protein interactions, based on the entries in the Protein Data Bank. Results We have compiled a pipeline for the quick analysis of halide-binding sites in proteins using the available software. Our analysis revealed that all of halides are strongly attracted by the guanidinium moiety of arginine side chains, however, there are also certain preferences among halides for other partners. Furthermore, there is a certain preference for coordination numbers in the binding sites, with a correlation between coordination numbers and amino acid composition. This pipeline can be used as a tool for the analysis of specific halide–protein interactions and assist phasing experiments relying on halides as anomalous scatters. Availability and implementation All data described in this article can be reproduced via complied pipeline published at https://github.com/rostkick/Halide_sites/blob/master/README.md. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Dmitrii Usoltsev
- Institute BioEngineering, ITMO University, Saint-Petersburg 197101, Russia
| | - Mayya Uspenskaya
- Institute BioEngineering, ITMO University, Saint-Petersburg 197101, Russia
| | - Andrey V Kajava
- Institute BioEngineering, ITMO University, Saint-Petersburg 197101, Russia.,Centre de Recherche en Biologie cellulaire de Montpellier (CRBM), UMR 5237 CNRS, Universite Montpellier, Montpellier 34293, France
| | - Albert Guskov
- Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, Groningen 9747 AG, the Netherlands
| |
Collapse
|
23
|
Okamoto N, Yamanaka N. Steroid Hormone Entry into the Brain Requires a Membrane Transporter in Drosophila. Curr Biol 2020; 30:359-366.e3. [PMID: 31928869 DOI: 10.1016/j.cub.2019.11.085] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 02/08/2023]
Abstract
Steroid hormones control various aspects of brain development and behavior in metazoans, but how they enter the central nervous system (CNS) through the blood-brain barrier (BBB) remains poorly understood. It is generally believed that steroid hormones freely diffuse through the plasma membrane of the BBB cells to reach the brain [1], because of the predominant "simple diffusion" model of steroid hormone transport across cell membranes. Recently, however, we challenged the simple diffusion model by showing that a Drosophila organic anion-transporting polypeptide (OATP), which we named Ecdysone Importer (EcI), is required for cellular uptake of the primary insect steroid hormone ecdysone [2]. As ecdysone is first secreted into the hemolymph before reaching the CNS [3], our finding raised the question of how ecdysone enters the CNS through the BBB to exert its diverse role in Drosophila brain development. Here, we demonstrate in the Drosophila BBB that EcI is indispensable for ecdysone entry into the CNS to facilitate brain development. EcI is highly expressed in surface glial cells that form the BBB, and EcI knockdown in the BBB suppresses ecdysone signaling within the CNS and blocks ecdysone-mediated neuronal events during development. In an ex vivo culture system, the CNS requires EcI in the BBB to incorporate ecdysone from the culture medium. Our results suggest a transporter-mediated mechanism of steroid hormone entry into the CNS, which may provide important implications in controlling brain development and behavior by regulating steroid hormone permeability across the BBB.
Collapse
Affiliation(s)
- Naoki Okamoto
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA.
| |
Collapse
|
24
|
La Merrill MA, Vandenberg LN, Smith MT, Goodson W, Browne P, Patisaul HB, Guyton KZ, Kortenkamp A, Cogliano VJ, Woodruff TJ, Rieswijk L, Sone H, Korach KS, Gore AC, Zeise L, Zoeller RT. Consensus on the key characteristics of endocrine-disrupting chemicals as a basis for hazard identification. Nat Rev Endocrinol 2020; 16:45-57. [PMID: 31719706 PMCID: PMC6902641 DOI: 10.1038/s41574-019-0273-8] [Citation(s) in RCA: 490] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/02/2019] [Indexed: 12/11/2022]
Abstract
Endocrine-disrupting chemicals (EDCs) are exogenous chemicals that interfere with hormone action, thereby increasing the risk of adverse health outcomes, including cancer, reproductive impairment, cognitive deficits and obesity. A complex literature of mechanistic studies provides evidence on the hazards of EDC exposure, yet there is no widely accepted systematic method to integrate these data to help identify EDC hazards. Inspired by work to improve hazard identification of carcinogens using key characteristics (KCs), we have developed ten KCs of EDCs based on our knowledge of hormone actions and EDC effects. In this Expert Consensus Statement, we describe the logic by which these KCs are identified and the assays that could be used to assess several of these KCs. We reflect on how these ten KCs can be used to identify, organize and utilize mechanistic data when evaluating chemicals as EDCs, and we use diethylstilbestrol, bisphenol A and perchlorate as examples to illustrate this approach.
Collapse
Affiliation(s)
- Michele A La Merrill
- Department of Environmental Toxicology, University of California, Davis, CA, USA.
| | - Laura N Vandenberg
- Department of Environmental Health Science, School of Public Health and Health Sciences, University of Masschusetts, Amherst, MA, USA
| | - Martyn T Smith
- School of Public Health, University of California, Berkeley, CA, USA
| | - William Goodson
- California Pacific Medical Center Research Institute, Sutter Hospital, San Francisco, CA, USA
| | - Patience Browne
- Environmental Directorate, Organisation for Economic Co-operation and Development, Paris, France
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Kathryn Z Guyton
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | | | - Vincent J Cogliano
- Office of the Science Advisor, United States Environmental Protection Agency, Washington, DC, USA
| | - Tracey J Woodruff
- Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Linda Rieswijk
- School of Public Health, University of California, Berkeley, CA, USA
- Institute of Data Science, Maastricht University, Maastricht, Netherlands
| | - Hideko Sone
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Ibaraki, Japan
| | - Kenneth S Korach
- Receptor Biology, Section Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Science, Durham, NC, USA
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Lauren Zeise
- Office of the Director, Office of Environmental Health Hazard Assessment of the California Environmental Protection Agency, Sacramento, CA, USA
| | - R Thomas Zoeller
- Biology Department, University of Masschusetts, Amherst, MA, USA
| |
Collapse
|
25
|
Walter KM, Dach K, Hayakawa K, Giersiefer S, Heuer H, Lein PJ, Fritsche E. Ontogenetic expression of thyroid hormone signaling genes: An in vitro and in vivo species comparison. PLoS One 2019; 14:e0221230. [PMID: 31513589 PMCID: PMC6742404 DOI: 10.1371/journal.pone.0221230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Thyroid hormone (TH) is essential for brain development. While disruption of TH signaling by environmental chemicals has been discussed as a mechanism of developmental neurotoxicity (DNT) for more than a decade, there remains a paucity of information linking specific TH disrupting chemicals to adverse neurodevelopmental outcomes. This data gap reflects, in part, the fact that the molecular machinery of TH signaling is complex and varies according to cell type and developmental time. Thus, establishing a baseline of the ontogenetic profile of expression of TH signaling molecules in relevant cell types is critical for developing in vitro and alternative systems-based models for screening TH disrupting chemicals for DNT. Here, we characterize the transcriptomic profile of molecules critical to TH signaling across three species-human, rat, and zebrafish-in vitro and in vivo across different stages of neurodevelopment. Our data indicate that while cultured human and rat neural progenitor cells, primary cultures of rat cortical cells, and larval zebrafish all express a fairly comprehensive transcriptome of TH signaling molecules, the spatiotemporal expression profiles as well as the responses to TH vary across species and developmental stages. The data presented here provides a roadmap for identifying appropriate in vitro and in simpler systems-based models for mechanistic studies and screening of chemicals that alter neurodevelopment via interference with TH action.
Collapse
Affiliation(s)
- Kyla M. Walter
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, Davis, CA, United States of America
| | - Katharina Dach
- IUF–Leibniz Research Institute for Environmental Medicine, Dusseldorf, Germany
| | - Keri Hayakawa
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, Davis, CA, United States of America
| | - Susanne Giersiefer
- IUF–Leibniz Research Institute for Environmental Medicine, Dusseldorf, Germany
| | - Heike Heuer
- IUF–Leibniz Research Institute for Environmental Medicine, Dusseldorf, Germany
- Dept. Endocrinology, University Hospital Essen, Essen, Germany
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California-Davis, School of Veterinary Medicine, Davis, CA, United States of America
- * E-mail: (PJL); (EF)
| | - Ellen Fritsche
- IUF–Leibniz Research Institute for Environmental Medicine, Dusseldorf, Germany
- * E-mail: (PJL); (EF)
| |
Collapse
|
26
|
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
|
27
|
Malagola E, Chen R, Bombardo M, Saponara E, Dentice M, Salvatore D, Reding T, Myers S, Hills AP, Graf R, Sonda S. Local hyperthyroidism promotes pancreatic acinar cell proliferation during acute pancreatitis. J Pathol 2019; 248:217-229. [PMID: 30714146 DOI: 10.1002/path.5247] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/17/2018] [Accepted: 01/08/2019] [Indexed: 01/01/2023]
Abstract
Proliferation of pancreatic acinar cells is a critical process in the pathophysiology of pancreatic diseases, because limited or defective proliferation is associated with organ dysfunction and patient morbidity. In this context, elucidating the signalling pathways that trigger and sustain acinar proliferation is pivotal to develop therapeutic interventions promoting the regenerative process of the organ. In this study we used genetic and pharmacological approaches to manipulate both local and systemic levels of thyroid hormones to elucidate their role in acinar proliferation following caerulein-mediated acute pancreatitis in mice. In addition, molecular mechanisms mediating the effects of thyroid hormones were identified by genetic and pharmacological inactivation of selected signalling pathways.In this study we demonstrated that levels of the thyroid hormone 3,3',5-triiodo-l-thyronine (T3) transiently increased in the pancreas during acute pancreatitis. Moreover, by using genetic and pharmacological approaches to manipulate both local and systemic levels of thyroid hormones, we showed that T3 was required to promote proliferation of pancreatic acinar cells, without affecting the extent of tissue damage or inflammatory infiltration.Finally, upon genetic and pharmacological inactivation of selected signalling pathways, we demonstrated that T3 exerted its mitogenic effect on acinar cells via a tightly controlled action on different molecular effectors, including histone deacetylase, AKT, and TGFβ signalling.In conclusion, our data suggest that local availability of T3 in the pancreas is required to promote acinar cell proliferation and provide the rationale to exploit thyroid hormone signalling to enhance pancreatic regeneration. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ermanno Malagola
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, Switzerland
| | - Rong Chen
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, Switzerland
| | - Marta Bombardo
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, Switzerland
| | - Enrica Saponara
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, Switzerland
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Domenico Salvatore
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Theresia Reding
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, Switzerland
| | - Stephen Myers
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Andrew P Hills
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Rolf Graf
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, Switzerland.,Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Sabrina Sonda
- Swiss Hepato-Pancreato-Biliary Center, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, Switzerland.,School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia.,Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
A transient window of hypothyroidism alters neural progenitor cells and results in abnormal brain development. Sci Rep 2019; 9:4662. [PMID: 30874585 PMCID: PMC6420655 DOI: 10.1038/s41598-019-40249-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022] Open
Abstract
Cortical heterotopias are clusters of ectopic neurons in the brain and are associated with neurodevelopmental disorders like epilepsy and learning disabilities. We have previously characterized the robust penetrance of a heterotopia in a rat model, induced by thyroid hormone (TH) disruption during gestation. However, the specific mechanism by which maternal TH insufficiency results in this birth defect remains unknown. Here we first determined the developmental window susceptible to endocrine disruption and describe a cellular mechanism responsible for heterotopia formation. We show that five days of maternal goitrogen treatment (10 ppm propylthiouracil) during the perinatal period (GD19-PN2) induces a periventricular heterotopia in 100% of the offspring. Beginning in the early postnatal brain, neurons begin to aggregate near the ventricles of treated animals. In parallel, transcriptional and architectural changes of this region were observed including decreased Sonic hedgehog (Shh) expression, abnormal cell adhesion, and altered radial glia morphology. As the ventricular epithelium is juxtaposed to two sources of brain THs, the cerebrospinal fluid and vasculature, this progenitor niche may be especially susceptible to TH disruption. This work highlights the spatiotemporal vulnerabilities of the developing brain and demonstrates that a transient period of TH perturbation is sufficient to induce a congenital abnormality.
Collapse
|
29
|
Montesinos MDM, Pellizas CG. Thyroid Hormone Action on Innate Immunity. Front Endocrinol (Lausanne) 2019; 10:350. [PMID: 31214123 PMCID: PMC6558108 DOI: 10.3389/fendo.2019.00350] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022] Open
Abstract
The interplay between thyroid hormone action and the immune system has been established in physiological and pathological settings. However, their connection is complex and still not completely understood. The thyroid hormones (THs), 3,3',5,5' tetraiodo-L-thyroxine (T4) and 3,3',5-triiodo-L-thyronine (T3) play essential roles in both the innate and adaptive immune responses. Despite much research having been carried out on this topic, the available data are sometimes difficult to interpret or even contradictory. Innate immune cells act as the first line of defense, mainly involving granulocytes and natural killer cells. In turn, antigen presenting cells, macrophages and dendritic cells capture, process and present antigens (self and foreign) to naïve T lymphocytes in secondary lymphoid tissues for the development of adaptive immunity. Here, we review the cellular and molecular mechanisms involved in T4 and T3 effects on innate immune cells. An overview of the state-of-the-art of TH transport across the target cell membrane, TH metabolism inside these cells, and the genomic and non-genomic mechanisms involved in the action of THs in the different innate immune cell subsets is included. The present knowledge of TH effects as well as the thyroid status on innate immunity helps to understand the complex adaptive responses achieved with profound implications in immunopathology, which include inflammation, cancer and autoimmunity, at the crossroads of the immune and endocrine systems.
Collapse
|
30
|
Hüser S, Guth S, Joost HG, Soukup ST, Köhrle J, Kreienbrock L, Diel P, Lachenmeier DW, Eisenbrand G, Vollmer G, Nöthlings U, Marko D, Mally A, Grune T, Lehmann L, Steinberg P, Kulling SE. Effects of isoflavones on breast tissue and the thyroid hormone system in humans: a comprehensive safety evaluation. Arch Toxicol 2018; 92:2703-2748. [PMID: 30132047 PMCID: PMC6132702 DOI: 10.1007/s00204-018-2279-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023]
Abstract
Isoflavones are secondary plant constituents of certain foods and feeds such as soy, linseeds, and red clover. Furthermore, isoflavone-containing preparations are marketed as food supplements and so-called dietary food for special medical purposes to alleviate health complaints of peri- and postmenopausal women. Based on the bioactivity of isoflavones, especially their hormonal properties, there is an ongoing discussion regarding their potential adverse effects on human health. This review evaluates and summarises the evidence from interventional and observational studies addressing potential unintended effects of isoflavones on the female breast in healthy women as well as in breast cancer patients and on the thyroid hormone system. In addition, evidence from animal and in vitro studies considered relevant in this context was taken into account along with their strengths and limitations. Key factors influencing the biological effects of isoflavones, e.g., bioavailability, plasma and tissue concentrations, metabolism, temporality (pre- vs. postmenopausal women), and duration of isoflavone exposure, were also addressed. Final conclusions on the safety of isoflavones are guided by the aim of precautionary consumer protection.
Collapse
Affiliation(s)
- S Hüser
- Institute for Food Toxicology, Senate Commission on Food Safety, University of Veterinary Medicine Hannover, Hannover, Germany
| | - S Guth
- Institute for Food Toxicology, Senate Commission on Food Safety, University of Veterinary Medicine Hannover, Hannover, Germany
| | - H G Joost
- Department of Experimental Diabetology, German Institute of Human Nutrition (DIfE), Nuthetal, Germany
| | - S T Soukup
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Haid-und-Neu-Str. 9, 76131, Karlsruhe, Germany
| | - J Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, CVK, Berlin, Germany
| | - L Kreienbrock
- Department of Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Hannover, Germany
| | - P Diel
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - D W Lachenmeier
- Chemisches und Veterinäruntersuchungsamt Karlsruhe, Karlsruhe, Germany
| | - G Eisenbrand
- Division of Food Chemistry and Toxicology, Molecular Nutrition, Department of Chemistry, Technische Universität Kaiserslautern, Kaiserslautern, Germany
| | - G Vollmer
- Department of Biology, Molecular Cell Physiology and Endocrinology, Technische Universität Dresden, Dresden, Germany
| | - U Nöthlings
- Department of Nutrition and Food Sciences, Nutritional Epidemiology, Rheinische Friedrich-Wilhelms University Bonn, Bonn, Germany
| | - D Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - A Mally
- Department of Toxicology, University of Würzburg, Würzburg, Germany
| | - T Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition (DIfE), Nuthetal, Germany
| | - L Lehmann
- Department of Food Chemistry, Institute for Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| | - P Steinberg
- Institute for Food Toxicology, University of Veterinary Medicine Hannover, Hannover, Germany
- Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Haid-und-Neu-Str. 9, 76131, Karlsruhe, Germany
| | - S E Kulling
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Haid-und-Neu-Str. 9, 76131, Karlsruhe, Germany.
| |
Collapse
|
31
|
Meyer Zu Schwabedissen HE, Ferreira C, Schaefer AM, Oufir M, Seibert I, Hamburger M, Tirona RG. Thyroid Hormones Are Transport Substrates and Transcriptional Regulators of Organic Anion Transporting Polypeptide 2B1. Mol Pharmacol 2018; 94:700-712. [PMID: 29735582 DOI: 10.1124/mol.117.111161] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/02/2018] [Indexed: 01/06/2025] Open
Abstract
Levothyroxine replacement therapy forms the cornerstone of hypothyroidism management. Variability in levothyroxine oral absorption may contribute to the well-recognized large interpatient differences in required dose. Moreover, levothyroxine-drug pharmacokinetic interactions are thought to be caused by altered oral bioavailability. Interestingly, little is known regarding the mechanisms contributing to levothyroxine absorption in the gastrointestinal tract. Here, we aimed to determine whether the intestinal drug uptake transporter organic anion transporting polypeptide 2B1 (OATP2B1) may be involved in facilitating intestinal absorption of thyroid hormones. We also explored whether thyroid hormones regulate OATP2B1 gene expression. In cultured Madin-Darby Canine Kidney II/OATP2B1 cells and in OATP2B1-transfected Caco-2 cells, thyroid hormones were found to inhibit OATP2B1-mediated uptake of estrone-3-sulfate. Competitive counter-flow experiments evaluating the influence on the cellular accumulation of estrone-3-sulfate in the steady state indicated that thyroid hormones were substrates of OATP2B1. Additional evidence that thyroid hormones were OATP2B1 substrates was provided by OATP2B1-dependent stimulation of thyroid hormone receptor activation in cell-based reporter assays. Bidirectional transport studies in intestinal Caco-2 cells showed net absorptive flux of thyroid hormones, which was attenuated by the presence of the OATP2B1 inhibitor, atorvastatin. In intestinal Caco-2 and LS180 cells, but not in liver Huh-7 or HepG2 cells, OATP2B1 expression was induced by treatment with thyroid hormones. Reporter gene assays revealed thyroid hormone receptor α-mediated transactivation of the SLCO2B1 1b and the SLCO2B1 1e promoters. We conclude that thyroid hormones are substrates and transcriptional regulators of OATP2B1. These insights provide a potential mechanistic basis for oral levothyroxine dose variability and drug interactions.
Collapse
Affiliation(s)
- Henriette E Meyer Zu Schwabedissen
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Celio Ferreira
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Anima M Schaefer
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Mouhssin Oufir
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Isabell Seibert
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Matthias Hamburger
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| | - Rommel G Tirona
- Biopharmacy (H.E.M.z.S., C.F., A.M.S., I.S.), and Pharmaceutical Biology (M.O., M.H.), Department Pharmaceutical Sciences, University of Basel, Basel, Switzerland; and Departments of Physiology and Pharmacology and Medicine, University of Western Ontario, London, Ontario, Canada (A.M.S., R.G.T.)
| |
Collapse
|
32
|
Prevot V, Dehouck B, Sharif A, Ciofi P, Giacobini P, Clasadonte J. The Versatile Tanycyte: A Hypothalamic Integrator of Reproduction and Energy Metabolism. Endocr Rev 2018; 39:333-368. [PMID: 29351662 DOI: 10.1210/er.2017-00235] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
Abstract
The fertility and survival of an individual rely on the ability of the periphery to promptly, effectively, and reproducibly communicate with brain neural networks that control reproduction, food intake, and energy homeostasis. Tanycytes, a specialized glial cell type lining the wall of the third ventricle in the median eminence of the hypothalamus, appear to act as the linchpin of these processes by dynamically controlling the secretion of neuropeptides into the portal vasculature by hypothalamic neurons and regulating blood-brain and blood-cerebrospinal fluid exchanges, both processes that depend on the ability of these cells to adapt their morphology to the physiological state of the individual. In addition to their barrier properties, tanycytes possess the ability to sense blood glucose levels, and play a fundamental and active role in shuttling circulating metabolic signals to hypothalamic neurons that control food intake. Moreover, accumulating data suggest that, in keeping with their putative descent from radial glial cells, tanycytes are endowed with neural stem cell properties and may respond to dietary or reproductive cues by modulating hypothalamic neurogenesis. Tanycytes could thus constitute the missing link in the loop connecting behavior, hormonal changes, signal transduction, central neuronal activation and, finally, behavior again. In this article, we will examine these recent advances in the understanding of tanycytic plasticity and function in the hypothalamus and the underlying molecular mechanisms. We will also discuss the putative involvement and therapeutic potential of hypothalamic tanycytes in metabolic and fertility disorders.
Collapse
Affiliation(s)
- Vincent Prevot
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Bénédicte Dehouck
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Ariane Sharif
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Philippe Ciofi
- Inserm, Neurocentre Magendie, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Paolo Giacobini
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| | - Jerome Clasadonte
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Center, Lille, France.,University of Lille, FHU 1000 Days for Health, School of Medicine, Lille, France
| |
Collapse
|
33
|
Gałecka E, Talarowska M, Maes M, Su KP, Górski P, Kumor-Kisielewska A, Szemraj J. Expression levels of interferon-ɣ and type 2 deiodinase in patients diagnosed with recurrent depressive disorders. Pharmacol Rep 2018; 70:133-138. [PMID: 29367100 DOI: 10.1016/j.pharep.2017.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/03/2017] [Accepted: 08/23/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Thyroid hormones (TH) are involved in modulation of the immune system and inflammation. TH dysregulation is associated with depressive disorders. The iodothyronine deiodinases (DIOs), the key enzymes for TH synthesis, can be affected and induced by pro-inflammatory cytokines. We aimed to investigate the levels of and correlation between type 2 DIO (DIO2) and interferon-gamma (IFN-ɣ) in patients with recurrent depressive disorders (rDD). METHODS Data from 91 rDD patients and 105 healthy controls were analyzed. The diagnoses are based on the ICD-10 criteria (F33.0-F33.8). Expression levels of DIO2 and IFN-ɣ were estimated using the method based on the polymerase chain reaction and the enzyme-linked immunosorbent assay (ELISA). RESULTS The DIO2 expression on mRNA/protein levels in rDD patients (both female and males) was reduced as compared with the control subjects. No correlation between DIO2 and IFN-ɣ expression was observed. CONCLUSION This is the first study to reveal that one may cautiously suggest that DIO2 may be involved in the development and/or progression of rDD. The mechanisms of TH regulation on depression, however, need further investigation.
Collapse
Affiliation(s)
- Elżbieta Gałecka
- Department of Pneumology and Allergy, Medical University of Łódź, Łódź, Poland.
| | - Monika Talarowska
- Department of Adult Psychiatry, Medical University of Łódź, Łódź, Poland
| | - Michael Maes
- Deakin University IMPACT Strategic Research Centre, Deakin University, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand; Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Brazil
| | - Kuan-Pin Su
- Graduate Institute of Neural and Cognitive Sciences, School of Medicine, China Medical University, Taichung, Taiwan; Department of Psychiatry and Mind-Body Research Center (MBI-Lab), China Medical University Hospital, 404, Taichung, Taiwan
| | - Paweł Górski
- Department of Pneumology and Allergy, Medical University of Łódź, Łódź, Poland
| | | | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Łódź, Łódź Poland
| |
Collapse
|
34
|
Richard S, Flamant F. Regulation of T3 Availability in the Developing Brain: The Mouse Genetics Contribution. Front Endocrinol (Lausanne) 2018; 9:265. [PMID: 29892264 PMCID: PMC5985302 DOI: 10.3389/fendo.2018.00265] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/07/2018] [Indexed: 12/11/2022] Open
Abstract
Alterations in maternal thyroid physiology may have deleterious consequences on the development of the fetal brain, but the underlying mechanisms remain elusive, hampering the development of appropriate therapeutic strategies. The present review sums up the contribution of genetically modified mouse models to this field. In particular, knocking out genes involved in thyroid hormone (TH) deiodination, transport, and storage has significantly improved the picture that we have of the economy of TH in the fetal brain and the underlying genetic program. These data pave the way for future studies to bridge the gap in knowledge between thyroid physiology and brain development.
Collapse
|
35
|
Hasel P, Dando O, Jiwaji Z, Baxter P, Todd AC, Heron S, Márkus NM, McQueen J, Hampton DW, Torvell M, Tiwari SS, McKay S, Eraso-Pichot A, Zorzano A, Masgrau R, Galea E, Chandran S, Wyllie DJA, Simpson TI, Hardingham GE. Neurons and neuronal activity control gene expression in astrocytes to regulate their development and metabolism. Nat Commun 2017; 8:15132. [PMID: 28462931 PMCID: PMC5418577 DOI: 10.1038/ncomms15132] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 03/02/2017] [Indexed: 12/17/2022] Open
Abstract
The influence that neurons exert on astrocytic function is poorly understood. To investigate this, we first developed a system combining cortical neurons and astrocytes from closely related species, followed by RNA-seq and in silico species separation. This approach uncovers a wide programme of neuron-induced astrocytic gene expression, involving Notch signalling, which drives and maintains astrocytic maturity and neurotransmitter uptake function, is conserved in human development, and is disrupted by neurodegeneration. Separately, hundreds of astrocytic genes are acutely regulated by synaptic activity via mechanisms involving cAMP/PKA-dependent CREB activation. This includes the coordinated activity-dependent upregulation of major astrocytic components of the astrocyte-neuron lactate shuttle, leading to a CREB-dependent increase in astrocytic glucose metabolism and elevated lactate export. Moreover, the groups of astrocytic genes induced by neurons or neuronal activity both show age-dependent decline in humans. Thus, neurons and neuronal activity regulate the astrocytic transcriptome with the potential to shape astrocyte-neuron metabolic cooperation.
Collapse
Affiliation(s)
- Philip Hasel
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Owen Dando
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, UK
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore 560065, India
| | - Zoeb Jiwaji
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Paul Baxter
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Alison C. Todd
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Samuel Heron
- School of Informatics, University of Edinburgh, Edinburgh EH8 9AB, UK
| | - Nóra M. Márkus
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Jamie McQueen
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - David W. Hampton
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Megan Torvell
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Sachin S. Tiwari
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Sean McKay
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Abel Eraso-Pichot
- Institut de Neurociències and Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Medicina, Edifici M, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine, Barcelona 08028, Spain
- Department of Biochemistry and Molecular Biology, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid 28029, Spain
| | - Roser Masgrau
- Institut de Neurociències and Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Medicina, Edifici M, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Elena Galea
- Institut de Neurociències and Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Medicina, Edifici M, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- Institució Catalana De Recerca I Estudis Avançats (ICREA), Passeig Lluís Companys 23, Barcelona, Catalonia, 08010, Spain
| | - Siddharthan Chandran
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, UK
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore 560065, India
| | - David J. A. Wyllie
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - T. Ian Simpson
- School of Informatics, University of Edinburgh, Edinburgh EH8 9AB, UK
| | - Giles E. Hardingham
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9XD, UK
- 10UK Dementia Research Institute at The University of Edinburgh, Edinburgh Medical School, 47 Little France Crescent, Edinburgh EH16 4TJ, , UK
| |
Collapse
|
36
|
Milanesi A, Brent GA. Beam Me In: Thyroid Hormone Analog Targets Alternative Transporter in Mouse Model of X-Linked Adrenoleukodystrophy. Endocrinology 2017; 158:1116-1119. [PMID: 28609836 PMCID: PMC5460838 DOI: 10.1210/en.2017-00206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 02/25/2017] [Indexed: 01/04/2023]
Affiliation(s)
- Anna Milanesi
- Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, and Endocrinology Division, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90073
| | - Gregory A Brent
- Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, and Endocrinology Division, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California 90073
| |
Collapse
|
37
|
Delbaere J, Vancamp P, Van Herck SLJ, Bourgeois NMA, Green MJ, Wingate RJT, Darras VM. MCT8 deficiency in Purkinje cells disrupts embryonic chicken cerebellar development. J Endocrinol 2017; 232:259-272. [PMID: 27879339 DOI: 10.1530/joe-16-0323] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 11/21/2016] [Indexed: 01/17/2023]
Abstract
Inactivating mutations in the human SLC16A2 gene encoding the thyroid hormone transporter monocarboxylate transporter 8 (MCT8) result in the Allan-Herndon-Dudley syndrome accompanied by severe locomotor deficits. The underlying mechanisms of the associated cerebellar maldevelopment were studied using the chicken as a model. Electroporation of an MCT8-RNAi vector into the cerebellar anlage of a 3-day-old embryo allowed knockdown of MCT8 in Purkinje cell precursors. This resulted in the downregulation of the thyroid hormone-responsive gene RORα and the Purkinje cell-specific differentiation marker LHX1/5 at day 6. MCT8 knockdown also results in a smaller and less complex dendritic tree at day 18 suggesting a pivotal role of MCT8 for cell-autonomous Purkinje cell maturation. Early administration of the thyroid hormone analogue 3,5,3'-triiodothyroacetic acid partially rescued early Purkinje cell differentiation. MCT8-deficient Purkinje cells also induced non-autonomous effects as they led to a reduced granule cell precursor proliferation, a thinner external germinal layer and a loss of PAX6 expression. By contrast, at day 18, the external germinal layer thickness was increased, with an increase in presence of Axonin-1-positive post-mitotic granule cells in the initial stage of radial migration. The concomitant accumulation of presumptive migrating granule cells in the molecular layer, suggests that inward radial migration to the internal granular layer is stalled. In conclusion, early MCT8 deficiency in Purkinje cells results in both cell-autonomous and non-autonomous effects on cerebellar development and indicates that MCT8 expression is essential from very early stages of development, providing a novel insight into the ontogenesis of the Allan-Herndon-Dudley syndrome.
Collapse
Affiliation(s)
- Joke Delbaere
- Laboratory of Comparative EndocrinologyDepartment of Biology, KU Leuven, Leuven, Belgium
| | - Pieter Vancamp
- Laboratory of Comparative EndocrinologyDepartment of Biology, KU Leuven, Leuven, Belgium
| | - Stijn L J Van Herck
- Laboratory of Comparative EndocrinologyDepartment of Biology, KU Leuven, Leuven, Belgium
| | - Nele M A Bourgeois
- Laboratory of Comparative EndocrinologyDepartment of Biology, KU Leuven, Leuven, Belgium
| | - Mary J Green
- Medical Research Council Centre for Developmental NeurobiologyKing's College London, London, UK
| | - Richard J T Wingate
- Medical Research Council Centre for Developmental NeurobiologyKing's College London, London, UK
| | - Veerle M Darras
- Laboratory of Comparative EndocrinologyDepartment of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Gigena N, Alamino VA, Montesinos MDM, Nazar M, Louzada RA, Wajner SM, Maia AL, Masini-Repiso AM, Carvalho DP, Cremaschi GA, Pellizas CG. Dissecting thyroid hormone transport and metabolism in dendritic cells. J Endocrinol 2017; 232:337-350. [PMID: 28052998 DOI: 10.1530/joe-16-0423] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 12/15/2016] [Indexed: 01/23/2023]
Abstract
We reported thyroid hormone (TH) receptor expression in murine dendritic cells (DCs) and 3,5,3'-triiodothyronine (T3)-dependent stimulation of DC maturation and ability to develop a Th1-type adaptive response. Moreover, an increased DC capacity to promote antigen-specific cytotoxic T-cell activity, exploited in a DC-based antitumor vaccination protocol, was revealed. However, putative effects of the main circulating TH, l-thyroxine (T4) and the mechanisms of TH transport and metabolism at DC level, crucial events for TH action at target cell level, were not known. Herein, we show that T4 did not reproduce those registered T3-dependent effects, finding that may reflect a homoeostatic control to prevent unspecific systemic activation of DCs. Besides, DCs express MCT10 and LAT2 TH transporters, and these cells mainly transport T3 with a favored involvement of MCT10 as its inhibition almost prevented T3 saturable uptake mechanism and reduced T3-induced IL-12 production. In turn, DCs express iodothyronine deiodonases type 2 and 3 (D2, D3) and exhibit both enzymatic activities with a prevalence towards TH inactivation. Moreover, T3 increased MCT10 and LAT2 expression and T3 efflux from DCs but not T3 uptake, whereas it induced a robust induction of D3 with a parallel slight reduction in D2. These findings disclose pivotal events involved in the mechanism of action of THs on DCs, providing valuable tools for manipulating the immunogenic potential of these cells. Furthermore, they broaden the knowledge of the TH mechanism of action at the immune system network.
Collapse
Affiliation(s)
- Nicolás Gigena
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Vanina A Alamino
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Del Mar Montesinos
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Magalí Nazar
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ruy A Louzada
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - Simone M Wajner
- Thyroid UnitEndocrinology Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana L Maia
- Thyroid UnitEndocrinology Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana M Masini-Repiso
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Denise P Carvalho
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - Graciela A Cremaschi
- Laboratorio de Neuroinmunomodulación y Oncología MolecularInstituto de Investigaciones Biomédicas (BIOMED-CONICET), Universidad Católica Argentina, Ciudad Autónoma de Buenos Aires, Argentina
| | - Claudia G Pellizas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
39
|
Salazar P, Cisternas P, Codocedo JF, Inestrosa NC. Induction of hypothyroidism during early postnatal stages triggers a decrease in cognitive performance by decreasing hippocampal synaptic plasticity. Biochim Biophys Acta Mol Basis Dis 2017; 1863:870-883. [PMID: 28088629 DOI: 10.1016/j.bbadis.2017.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/25/2016] [Accepted: 01/04/2017] [Indexed: 01/27/2023]
Abstract
Thyroid hormones are vital in the control of multiple body functions, including the correct performance of the brain. Multiple diseases are associated with thyroid gland functioning, including hypothyroidism. To date, little is known regarding the effects of the establishment of this condition at a young age on brain function. Here, we evaluated the effect of hypothyroidism in an early postnatal stage in cognitive abilities with focus on the hippocampus. In our model, hypothyroidism was induced in young rats at 21days of age using 0.05% 6-propyl-2-thiouracil (PTU) for 4weeks reaching significantly lower levels of fT4 (control: 1.337ng/dL±0.115, PTU: 0.050ng/dL±0.001). Following the induction of hypothyroidism, several cognitive tasks were assessed to investigate the effects of hypothyroidism on cognition performance. We determined that hypothyroidism triggers a significant dysfunction in learning and memory processes observed in the Morris Water Maze were the latency times were higher in PTU rats (controls: 37s; PTU: 57s). The cognitive impairment was correlated with a reduction in hippocampal plasticity with respect to both long-term potentiation (LTP) (control: 1.45, PTU: 1.00) and depression (LTD) (control: 0.71, PTU: 1.01). Furthermore, a decrease in the rate of glucose utilization (control: 223nmol∗mg of protein, PTU:148nmol∗mg of protein) was observed, along with an increase in oxidative stress and a decrease in MAP2 marker in the hippocampus. Our findings suggest that the induction of hypothyroidism in a young rat model alters numerous functions at the level of the hippocampus.
Collapse
Affiliation(s)
- Paulina Salazar
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pedro Cisternas
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Universidad de Atacama, Facultad de Ciencias Naturales, Departamento de Química y Biología, Copayapu 485, Copiapó, Chile
| | - Juan Francisco Codocedo
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|