1
|
Sane R, Seyffarth C, Kleissle S, Neuenschwander M, von Kries JP, Frädrich C, Renko K, Wirth EK, Köhrle J. Identification and Characterization of Highly Potent and Isoenzyme-Selective Inhibitors of Deiodinase Type I via a Nonradioactive High-Throughput Screening Method. Thyroid 2025; 35:576-589. [PMID: 40170637 DOI: 10.1089/thy.2025.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Objective: Deiodinase type I (DIO1) is crucial in maintaining thyroid hormone (TH) balance. It converts the prohormone thyroxine (T4) to the active triiodothyronine (T3) and degrades T3 to inactive 3,3'-diiodothyronine (3,3'-T2). It also acts on reverse T3 (rT3) and sulfated TH metabolites, thus contributing to TH elimination. Upregulation of DIO1 is linked to hyperthyroid conditions such as Graves' disease and autonomous thyroid adenoma, making it a promising target for pharmacological intervention. The adverse side effects of the antithyroid drug propylthiouracil (PTU), used in clinics to treat hyperthyroidism due to its thyroid peroxidase- and DIO1-blocking action, highlight the need for novel and potent DIO1-selective inhibitors. Methods: Using a semiautomatic high-throughput screening (HTS) assay based on the Sandell-Kolthoff (SK) reaction in 384-well plates, we screened 69,344 low-molecular-weight compounds for DIO1-inhibitory effects. Shortlisted hits underwent detailed manual characterization, where we evaluated the potency and isoenzyme specificity by assessing their DIO-inhibitory effects on enzyme preparations from all three DIO isoenzymes, over a wide concentration range (5 nM-20 µM). To evaluate the DIO1 inhibitory effects in intact cells, we applied a novel protocol based on the SK reaction to cell culture supernatants and assessed the intracellular deiodinase activity in DIO1 overexpressing HEK293 cells. Results: The robust HTS assay flagged 436 (<1%) of the screened compounds as hits, also including known DIO1 inhibitors such as PTU and genistein. Based on a validation screen of 298 compounds, we prioritized 26 compounds to comprehensively characterize their DIO1-selective inhibition. We identified 15 DIO1-selective compounds (IC50 < 1 µM), more potent than the bonafide DIO1-selective inhibitor PTU. Additionally, 8 of the 13 tested compounds were found capable of inhibiting DIO1 in intact cells. Conclusions: With a successful SK-reaction-based HTS application, we identified novel, potent, and selective inhibitors of DIO1 with nanomolar IC50 values. Furthermore, we successfully showed that some of these compounds were also capable of inhibiting intracellular DIO1 in intact cells. These novel compounds hold immense potential in studying TH modulation, deciphering DIO1 enzyme structure, and developing structure-activity relationships. Furthermore, our novel inhibitors act as lead compounds in developing strategies to combat hyperthyroidism.
Collapse
Affiliation(s)
- Rajas Sane
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institut für Experimentelle Endokrinologie, Berlin, Germany
| | - Carola Seyffarth
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Sabrina Kleissle
- Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtz-Gemeinschaft, Berlin, Germany
| | | | | | - Caroline Frädrich
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institut für Experimentelle Endokrinologie, Berlin, Germany
| | - Kostja Renko
- German Federal Institute for Risk Assessment, German Centre for the Protection of Laboratory Animals (Bf3R), Berlin, Germany
| | - Eva K Wirth
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, European Reference Network on Rare Endocrine Diseases (ENDO-ERN), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Josef Köhrle
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institut für Experimentelle Endokrinologie, Berlin, Germany
| |
Collapse
|
2
|
Normann SS, Ma Y, Andersen HR, Valente MJ, Renko K, Arnold S, Jensen RC, Andersen MS, Vinggaard AM. Pyrethroid exposure biomarker 3-phenoxybenzoic acid (3-PBA) binds to transthyretin and is positively associated with free T3 in pregnant women. Int J Hyg Environ Health 2025; 264:114495. [PMID: 39616831 DOI: 10.1016/j.ijheh.2024.114495] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 02/04/2025]
Abstract
Pyrethroids constitute a large group of insecticides widely used in agriculture, indoor environments, and in vector control. Structurally, pyrethroids resemble thyroid hormones, and have been suggested to be thyroid hormone disruptors based on experimental studies. During pregnancy, even minor disturbances in maternal levels can affect fetal brain development. Therefore, we aimed to investigate whether three commonly used pyrethroids and their common metabolite, 3-PBA, were able to trigger thyroid disrupting effects on thyroid hormone production and transport, activation or deactivation of thyroid hormones, recycling of iodine, or on iodide uptake into the thyroid. Furthermore, we investigated associations between urinary 3-PBA concentrations (as biomarker of pyrethroid exposure) and serum concentrations of thyroid hormones in early pregnancy in the large prospective Odense Child Cohort (OCC). We found that the generic metabolite, 3-PBA, was capable of binding to transthyretin (TTR) at low concentrations, comparable to those reported in human cord blood. Among pregnant women in OCC, we found urinary 3-PBA concentrations to be positively associated with free triiodothyronine (fT3) serum levels. Displacement of thyroid hormones from TTR by pyrethroid exposure in early pregnancy may disturb the transplacental transport of thyroid hormones to the fetus during a very vulnerable window of development, including neural maturation. We did not find any evidence for thyroid disrupting effects in vitro for the three pyrethroids: Deltamethrin, α-cypermethrin, and etofenprox.
Collapse
Affiliation(s)
- Stine Søgaard Normann
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark.
| | - Yanying Ma
- Cell Toxicology Team, National Food Institute, Technical University of Denmark, 2800, Kgs. Lyngby, Denmark
| | - Helle Raun Andersen
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Maria João Valente
- Cell Toxicology Team, National Food Institute, Technical University of Denmark, 2800, Kgs. Lyngby, Denmark
| | - Kostja Renko
- German Federal Institute for Risk Assessment, German Centre for the Protection of Laboratory Animals (Bf3R), Berlin, Germany
| | - Selina Arnold
- German Federal Institute for Risk Assessment, German Centre for the Protection of Laboratory Animals (Bf3R), Berlin, Germany
| | - Richard Christian Jensen
- Department of Clinical Pharmacology, Pharmacy and Environmental Medicine, Institute of Public Health, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark; Department of Endocrinology, Odense University Hospital, Søndre Blvd. 29, 5000, Odense C, Denmark
| | | | - Anne Marie Vinggaard
- Cell Toxicology Team, National Food Institute, Technical University of Denmark, 2800, Kgs. Lyngby, Denmark
| |
Collapse
|
3
|
Vergauwen L, Bajard L, Tait S, Langezaal I, Sosnowska A, Roncaglioni A, Hessel E, van den Brand AD, Haigis AC, Novák J, Hilscherová K, Buławska N, Papaioannou N, Renieri E, Spilioti E, Spyropoulou A, Gutleb AC, Holbech H, Nikolopoulou D, Jacobs MN, Knapen D. A 2024 inventory of test methods relevant to thyroid hormone system disruption for human health and environmental regulatory hazard assessment. OPEN RESEARCH EUROPE 2024; 4:242. [PMID: 39931575 PMCID: PMC11809485 DOI: 10.12688/openreseurope.18739.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 02/13/2025]
Abstract
Thyroid hormone system disruption (THSD) is a growing concern in chemical hazard assessment due to its impact on human and environmental health and the scarce methods available for assessing the THSD potential of chemicals. In particular, the general lack of validated in silico and in vitro methods for assessing THS activity is of high concern. This manuscript provides an inventory of test methods relevant to THSD. Building on the Organisation for Economic Co-operation and Development (OECD) Guidance Document 150 and recent international developments, we highlight progress in in silico and in vitro methods, as well as in vivo assays. The provided inventory categorizes available methods according to the levels of the OECD Conceptual Framework, with an assessment of the validation status of each method. At Level 1, 12 in silico models that have been statistically validated and are directly related to THSD have been identified. At Level 2, 67 in vitro methods have been listed including those assessed in key initiatives such as the European Union Network of Laboratories for the Validation of Alternative Methods (EU-NETVAL) validation study to identify potential thyroid disruptors. At Levels 3-5, THSD-sensitive endpoints are being included in existing fish-based OECD Test Guidelines to complement amphibian assays. In total, the inventory counts 108 entries comprising established methods (e.g., OECD Test Guidelines) as well as citable methods that are under further development and in some cases are ready for validation or in the initial stages of validation. This work aims to support the ongoing development of strategies for regulatory hazard assessment, such as integrated approaches to testing and assessment (IATAs), for endocrine disruptors, addressing critical gaps in the current testing landscape for THSD in both human and environmental health contexts.
Collapse
Affiliation(s)
- Lucia Vergauwen
- Zebrafishlab, Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, 2610, Belgium
| | - Lola Bajard
- RECETOX, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic
| | - Sabrina Tait
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, 00161, Italy
| | - Ingrid Langezaal
- European Commission Joint Research Centre Ispra, Ispra, Lombardy, 21027, Italy
| | - Anita Sosnowska
- Faculty of Chemistry, University of Gdansk, Gdańsk, 80-308, Poland
| | - Alessandra Roncaglioni
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Lombardy, 20156, Italy
| | - Ellen Hessel
- National Institute for Public Health and the Environment, Bilthoven, Utrecht, 3721, The Netherlands
| | - Annick D van den Brand
- National Institute for Public Health and the Environment, Bilthoven, Utrecht, 3721, The Netherlands
| | - Ann-Cathrin Haigis
- Zebrafishlab, Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, 2610, Belgium
| | - Jiří Novák
- RECETOX, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic
| | - Klára Hilscherová
- RECETOX, Faculty of Science, Masaryk University, Brno, 611 37, Czech Republic
| | - Natalia Buławska
- Faculty of Chemistry, University of Gdansk, Gdańsk, 80-308, Poland
| | - Nafsika Papaioannou
- HERACLES Research Center on the Exposome and Health, Center for Interdisciplinary Research and Innovation, Aristotle University of Thessaloniki, Thessaloniki, 570 01, Greece
| | - Elisavet Renieri
- HERACLES Research Center on the Exposome and Health, Center for Interdisciplinary Research and Innovation, Aristotle University of Thessaloniki, Thessaloniki, 570 01, Greece
| | - Eliana Spilioti
- Laboratory of Toxicological Control of Pesticides, Scientific Directorate of Pesticides’ Control and Phytopharmacy, Benaki Phytopathological Institute, Athens, Attica, 145 61, Greece
| | - Anastasia Spyropoulou
- Laboratory of Toxicological Control of Pesticides, Scientific Directorate of Pesticides’ Control and Phytopharmacy, Benaki Phytopathological Institute, Athens, Attica, 145 61, Greece
| | - Arno C Gutleb
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology, Belvaux, 4422, Luxembourg
| | - Henrik Holbech
- Department of Biology, University of Southern Denmark, Odense, 5230, Denmark
| | - Dimitra Nikolopoulou
- Laboratory of Toxicological Control of Pesticides, Scientific Directorate of Pesticides’ Control and Phytopharmacy, Benaki Phytopathological Institute, Athens, Attica, 145 61, Greece
| | - Miriam N Jacobs
- Radiation, Chemical and Environmental Hazards, Harwell Innovation Campus, UK Health Security Agency, Chilton, OX11 0RQ, UK
| | - Dries Knapen
- Zebrafishlab, Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, 2610, Belgium
| |
Collapse
|
4
|
Giri D, Govindaraj V, Kumar S, Ungati H, Mugesh G. A Highly Selective Fluorescent Probe for Monitoring the Thyroid Hormone Transporter Activity in Mammalian Cells. Chemistry 2024; 30:e202401719. [PMID: 38995511 DOI: 10.1002/chem.202401719] [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: 05/01/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/13/2024]
Abstract
Monocarboxylate transporter 8 (MCT8) is a trans-membrane transporter, which mediates the cellular delivery of thyroid hormones, L-thyroxine (T4) and 3,5,3'-triiodo-L-thyronine (T3). In humans, the MCT8 protein is encoded by the SLC16A2 gene and mutations in the transporter cause a genetic neurological disorder known as Allan-Herndon-Dudley Syndrome (AHDS). MCT8 deficiency leads to impaired transport of thyroid hormones in the brain. Radiolabelled T4 and T3 or LC/MS-MS methods have been used to monitor the thyroid hormone uptake through MCT8. Herein, we developed a fluorescent based assay to monitor the thyroid hormone uptake through MCT8. A dansyl-based fluorescent probe having L-thyroxine moiety is found to be highly selective towards MCT8 in living cells. The high selectivity of the probe towards MCT8 can be attributed to the halogen bond-mediated recognition by the transporter protein. The presence of a free carboxylic acid group is essential for the specificity of the probe towards MCT8. Additionally, the selectivity of the probe for MCT8 is abolished upon esterification of the carboxylic group. Similarly, MCT8 does not recognize the probe when it contains a free amine group.
Collapse
Affiliation(s)
- Debasish Giri
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Vijayakumar Govindaraj
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
- Department of Biochemistry, School of Allied Health Sciences, REVA University, Bangalore, India
| | - Sagar Kumar
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Harinarayana Ungati
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
5
|
Wagenaars F, Cenijn P, Chen Z, Meima M, Scholze M, Hamers T. Two novel in vitro assays to screen chemicals for their capacity to inhibit thyroid hormone transmembrane transporter proteins OATP1C1 and OAT4. Arch Toxicol 2024; 98:3019-3034. [PMID: 38761188 PMCID: PMC11324666 DOI: 10.1007/s00204-024-03787-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024]
Abstract
Early brain development depends on adequate transport of thyroid hormones (THs) from the maternal circulation to the fetus. To reach the fetal brain, THs have to cross several physiological barriers, including the placenta, blood-brain-barrier and blood-cerebrospinal fluid-barrier. Transport across these barriers is facilitated by thyroid hormone transmembrane transporters (THTMTs). Some endocrine disrupting chemicals (EDCs) can interfere with the transport of THs by THTMTs. To screen chemicals for their capacity to disrupt THTMT facilitated TH transport, in vitro screening assays are required. In this study, we developed assays for two THTMTs, organic anion transporter polypeptide 1C1 (OATP1C1) and organic anion transporter 4 (OAT4), both known to play a role in the transport of THs across barriers. We used overexpressing cell models for both OATP1C1 and OAT4, which showed an increased uptake of radiolabeled T4 compared to control cell lines. Using these models, we screened various reference and environmental chemicals for their ability to inhibit T4 uptake by OATP1C1 and OAT4. Tetrabromobisphenol A (TBBPA) was identified as an OATP1C1 inhibitor, more potent than any of the reference chemicals tested. Additionally perfluorooctanesulfonic acid (PFOS), perfluoroctanic acid (PFOA), pentachlorophenol and quercetin were identified as OATP1C1 inhibitors in a similar range of potency to the reference chemicals tested. Bromosulfophthalein, TBBPA, PFOA and PFOS were identified as potent OAT4 inhibitors. These results demonstrate that EDCs commonly found in our environment can disrupt TH transport by THTMTs, and contribute to the identification of molecular mechanisms underlying TH system disruption chemicals.
Collapse
Affiliation(s)
- Fabian Wagenaars
- Amsterdam Institute for Life and Environment (A-Life), Vrije Universiteit Amsterdam (VU), De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands.
| | - Peter Cenijn
- Amsterdam Institute for Life and Environment (A-Life), Vrije Universiteit Amsterdam (VU), De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - Zhongli Chen
- Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
| | - Marcel Meima
- Academic Centre for Thyroid Diseases, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, 3015 GD, Rotterdam, The Netherlands
| | - Martin Scholze
- Centre for Pollution Research and Policy, College of Health, Medicine and Life Sciences, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK
| | - Timo Hamers
- Amsterdam Institute for Life and Environment (A-Life), Vrije Universiteit Amsterdam (VU), De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Kadic A, Oles P, Fischer BC, Reetz AE, Sylla BS, Feiertag K, Ritz V, Heise T, Marx-Stoelting P, Tralau T, Renko K, Solano MDLM. In vitro and in vivo investigation of a thyroid hormone system-specific interaction with triazoles. Sci Rep 2024; 14:6503. [PMID: 38499550 PMCID: PMC10948911 DOI: 10.1038/s41598-024-55019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
Alterations in thyroid hormones (TH) and thyroid-stimulating hormone levels are frequently found following exposure to chemicals of concern. Dysregulation of TH levels can severely perturb physiological growth, metabolism, differentiation, homeostasis in the adult and developmental processes in utero. A frequently identified mode of action for this interaction is the induction of hepatic detoxification mechanisms (e.g. SULTs and UGTs), which lead to TH conjugation and elimination and therefore interfere with hormonal homeostasis, fulfilling the endocrine disruptors (EDs) definition. A short-term study in rats with dietary exposure to cyproconazole, epoxiconazole and prochloraz was conducted and hepatocyte hypertrophy, hepatic UGT activity and Phase 1/2 gene expression inductions were observed together with changes in TH levels and thyroid follicular hypertrophy and hyperplasia. To test for specific interaction with the thyroid hormone system, in vitro assays were conducted covering thyroidal I-uptake (NIS), TH transmembranal transport via MCT8 and thyroid peroxidase (TPO) function. Assays for iodothyronine deiodinases (DIO1-DIO3) and iodotyrosine deiodinase (DEHAL1) were included, and from the animal experiment, Dio1 and Dehal1 activities were measured in kidney and liver as relevant local indicators and endpoints. The fungicides did not affect any TH-specific KEs, in vitro and in vivo, thereby suggesting hepatic conjugation as the dominant MoA.
Collapse
Affiliation(s)
- Asya Kadic
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Patricia Oles
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R), Diedersdorfer Weg 1, 12277, Berlin, Germany
| | - Benjamin Christian Fischer
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Anne Elisabeth Reetz
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
- Institute of Veterinary Pathology (WE12), Freie University Berlin, Berlin, Germany
| | - Boubacar Sidiki Sylla
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Katreece Feiertag
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Vera Ritz
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Tanja Heise
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Philip Marx-Stoelting
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Tewes Tralau
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Kostja Renko
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R), Diedersdorfer Weg 1, 12277, Berlin, Germany.
| | - Marize de Lourdes Marzo Solano
- Department of Pesticides Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| |
Collapse
|
7
|
Escher BI, Lamoree M, Antignac JP, Scholze M, Herzler M, Hamers T, Jensen TK, Audebert M, Busquet F, Maier D, Oelgeschläger M, Valente MJ, Boye H, Schmeisser S, Dervilly G, Piumatti M, Motteau S, König M, Renko K, Margalef M, Cariou R, Ma Y, Treschow AF, Kortenkamp A, Vinggaard AM. Mixture Risk Assessment of Complex Real-Life Mixtures-The PANORAMIX Project. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:12990. [PMID: 36293571 PMCID: PMC9602166 DOI: 10.3390/ijerph192012990] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 05/06/2023]
Abstract
Humans are involuntarily exposed to hundreds of chemicals that either contaminate our environment and food or are added intentionally to our daily products. These complex mixtures of chemicals may pose a risk to human health. One of the goals of the European Union's Green Deal and zero-pollution ambition for a toxic-free environment is to tackle the existent gaps in chemical mixture risk assessment by providing scientific grounds that support the implementation of adequate regulatory measures within the EU. We suggest dealing with this challenge by: (1) characterising 'real-life' chemical mixtures and determining to what extent they are transferred from the environment to humans via food and water, and from the mother to the foetus; (2) establishing a high-throughput whole-mixture-based in vitro strategy for screening of real-life complex mixtures of organic chemicals extracted from humans using integrated chemical profiling (suspect screening) together with effect-directed analysis; (3) evaluating which human blood levels of chemical mixtures might be of concern for children's development; and (4) developing a web-based, ready-to-use interface that integrates hazard and exposure data to enable component-based mixture risk estimation. These concepts form the basis of the Green Deal project PANORAMIX, whose ultimate goal is to progress mixture risk assessment of chemicals.
Collapse
Affiliation(s)
- Beate I. Escher
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research—UFZ, DE-04318 Leipzig, Germany
- Environmental Toxicology, Department of Geoscience, Eberhard Karls University Tübingen, DE-72076 Tübingen, Germany
| | - Marja Lamoree
- Department Environment & Health, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | | | - Martin Scholze
- Centre for Pollution Research and Policy, Environmental Sciences Division, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
| | - Matthias Herzler
- German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Timo Hamers
- Department Environment & Health, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Tina Kold Jensen
- Department of Environmental Medicine, University of Southern Denmark, DK-5000 Odense, Denmark
| | - Marc Audebert
- Toxalim, UMR1331, INRAE, 31027 Toulouse, France
- PrediTox, 31100 Toulouse, France
| | | | | | | | - Maria João Valente
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Henriette Boye
- Odense Child Cohort, Hans Christian Andersen Hospital for Children, Odense University Hospital, DK-5000 Odense, Denmark
| | | | | | | | | | - Maria König
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research—UFZ, DE-04318 Leipzig, Germany
- Environmental Toxicology, Department of Geoscience, Eberhard Karls University Tübingen, DE-72076 Tübingen, Germany
| | - Kostja Renko
- German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Maria Margalef
- Department Environment & Health, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | | | - Yanying Ma
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | | | - Andreas Kortenkamp
- Centre for Pollution Research and Policy, Environmental Sciences Division, Brunel University London, Kingston Lane, Uxbridge UB8 3PH, UK
| | - Anne Marie Vinggaard
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
8
|
Olker JH, Korte JJ, Haselman JT, Hornung MW, Degitz SJ. Cross-species comparison of chemical inhibition of human and Xenopus iodotyrosine deiodinase. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 249:106227. [PMID: 35767922 PMCID: PMC9887787 DOI: 10.1016/j.aquatox.2022.106227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 06/15/2023]
Abstract
The transition to include in vitro-based data in chemical hazard assessment has resulted in the development and implementation of screening assays to cover a diversity of biological pathways, including recently added assays to interrogate chemical disruption of proteins relevant to thyroid signaling pathways. Iodotyrosine deiodinase (IYD), the iodide recycling enzyme, is one such thyroid-relevant endpoint for which a human-based screening assay has recently been developed and used to screen large libraries of chemicals. Presented here is the development of an amphibian IYD inhibition assay and its implementation to conduct a cross-species comparison between chemical inhibition of mammalian and non-mammalian IYD enzyme activity. The successful development of an amphibian IYD inhibition assay was based on demonstration of sufficient IYD enzyme activity in several tissues collected from larval Xenopus laevis. With this new assay, 154 chemicals were tested in concentration-response to provide a basis for comparison of relative chemical potency to results obtained from the human IYD assay. Most chemicals exhibited similar inhibition in both assays, with less than 25% variation in median inhibition for 120 of 154 chemicals and 85% concordance in categorization of "active" (potential IYD inhibitor) versus "inactive". For chemicals that produced 50% or greater inhibition in both assays, rank-order potency was similar, with the majority of the IC50s varying by less than 2-fold (and all within an order of magnitude). Most differences resulted from greater maximum inhibition or higher chemical potency observed with human IYD. This strong cross-species agreement suggests that results from the human-based assay would be conservatively predictive of chemical effects on amphibian IYD.
Collapse
Affiliation(s)
- Jennifer H Olker
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, 6201 Congdon Blvd, Duluth, MN 55804, USA.
| | - Joseph J Korte
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, 6201 Congdon Blvd, Duluth, MN 55804, USA
| | - Jonathan T Haselman
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, 6201 Congdon Blvd, Duluth, MN 55804, USA
| | - Michael W Hornung
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, 6201 Congdon Blvd, Duluth, MN 55804, USA
| | - Sigmund J Degitz
- Great Lakes Toxicology and Ecology Division, Office of Research and Development, Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, 6201 Congdon Blvd, Duluth, MN 55804, USA
| |
Collapse
|
9
|
Renko K, Kerp H, Pape J, Rijntjes E, Burgdorf T, Führer D, Köhrle J. Tentative Application of a Streamlined Protocol to Determine Organ-Specific Regulations of Deiodinase 1 and Dehalogenase Activities as Readouts of the Hypothalamus-Pituitary-Thyroid-Periphery-Axis. FRONTIERS IN TOXICOLOGY 2022; 4:822993. [PMID: 35387426 PMCID: PMC8978789 DOI: 10.3389/ftox.2022.822993] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/24/2022] [Indexed: 11/22/2022] Open
Abstract
In animal studies, both in basic science and in toxicological assessment of potential endocrine disruptors, the state of the thyroid hormone (TH) axis is often described and defined exclusively by the concentrations of circulating THs and TSH. Although it is known that the local, organ-specific effects of THs are also substantially regulated by local mechanisms such as TH transmembrane transport and metabolism of TH by deiodinases, such endpoint parameters of the axis are rarely assessed in these experiments. Currently developed in vitro assays utilize the Sandell-Kolthoff reaction, a photometric method of iodide determination, to test the effect of chemicals on iodotyrosine and iodothyronine deiodinases. Furthermore, this technology offers the possibility to determine the iodine content of various sample types (e.g., urine, ex vivo tissue) in a simple way. Here, we measured deiodinase type 1 and iodotyrosine dehalogenase activity by means of the Sandell-Kolthoff reaction in ex vivo samples of hypo- and hyperthyroid mice of two age groups (young; 3 months and old; 20 months). In thyroid, liver and kidney, organ-specific regulation patterns emerged across both age groups, which, based on this pilot study, may serve as a starting point for a deeper characterization of the TH system in relevant studies in the future and support the development of Integrated Approach for Testing and Assessment (IATA).
Collapse
Affiliation(s)
- Kostja Renko
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R), Berlin, Germany
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany
- *Correspondence: Kostja Renko,
| | - Helena Kerp
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Janina Pape
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Eddy Rijntjes
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany
| | - Tanja Burgdorf
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R), Berlin, Germany
| | - Dagmar Führer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Josef Köhrle
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany
| |
Collapse
|
10
|
Venugopalan V, Al-Hashimi A, Rehders M, Golchert J, Reinecke V, Homuth G, Völker U, Manirajah M, Touzani A, Weber J, Bogyo MS, Verrey F, Wirth EK, Schweizer U, Heuer H, Kirstein J, Brix K. The Thyroid Hormone Transporter Mct8 Restricts Cathepsin-Mediated Thyroglobulin Processing in Male Mice through Thyroid Auto-Regulatory Mechanisms That Encompass Autophagy. Int J Mol Sci 2021; 22:ijms22010462. [PMID: 33466458 PMCID: PMC7796480 DOI: 10.3390/ijms22010462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
The thyroid gland is both a thyroid hormone (TH) generating as well as a TH responsive organ. It is hence crucial that cathepsin-mediated proteolytic cleavage of the precursor thyroglobulin is regulated and integrated with the subsequent export of TH into the blood circulation, which is enabled by TH transporters such as monocarboxylate transporters Mct8 and Mct10. Previously, we showed that cathepsin K-deficient mice exhibit the phenomenon of functional compensation through cathepsin L upregulation, which is independent of the canonical hypothalamus-pituitary-thyroid axis, thus, due to auto-regulation. Since these animals also feature enhanced Mct8 expression, we aimed to understand if TH transporters are part of the thyroid auto-regulatory mechanisms. Therefore, we analyzed phenotypic differences in thyroid function arising from combined cathepsin K and TH transporter deficiencies, i.e., in Ctsk-/-/Mct10-/-, Ctsk-/-/Mct8-/y, and Ctsk-/-/Mct8-/y/Mct10-/-. Despite the impaired TH export, thyroglobulin degradation was enhanced in the mice lacking Mct8, particularly in the triple-deficient genotype, due to increased cathepsin amounts and enhanced cysteine peptidase activities, leading to ongoing thyroglobulin proteolysis for TH liberation, eventually causing self-thyrotoxic thyroid states. The increased cathepsin amounts were a consequence of autophagy-mediated lysosomal biogenesis that is possibly triggered due to the stress accompanying intrathyroidal TH accumulation, in particular in the Ctsk-/-/Mct8-/y/Mct10-/- animals. Collectively, our data points to the notion that the absence of cathepsin K and Mct8 leads to excessive thyroglobulin degradation and TH liberation in a non-classical pathway of thyroid auto-regulation.
Collapse
Affiliation(s)
- Vaishnavi Venugopalan
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Alaa Al-Hashimi
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Maren Rehders
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Janine Golchert
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Vivien Reinecke
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475 Greifswald, Germany; (J.G.); (V.R.); (G.H.); (U.V.)
| | - Mythili Manirajah
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Adam Touzani
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Jonas Weber
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
| | - Matthew S. Bogyo
- Department of Pathology, School of Medicine, Stanford University, 300 Pasteur Dr., Stanford, CA 94305-5324, USA;
| | - Francois Verrey
- Physiologisches Institut, Universität Zürich, Winterthurerstr. 190, CH-8057 Zürich, Switzerland;
| | - Eva K. Wirth
- Berlin Institute of Health, Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Hessische Str. 3-4, Germany and DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, D-10115 Berlin, Germany;
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Medizinische Fakultät, Universität Bonn, Nußallee 11, D-53115 Bonn, Germany;
| | - Heike Heuer
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen (AöR), Universität Duisburg-Essen, Hufelandstr. 55, D-45147 Essen, Germany;
| | - Janine Kirstein
- Fachbereich 2 Biologie/Chemie, Faculty of Cell Biology, Universität Bremen, Leobener Straße 5, D-28359 Bremen, Germany;
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Focus Area HEALTH, Jacobs University Bremen, Campus Ring 1, D-29759 Bremen, Germany; (V.V.); (A.A.-H.); (M.R.); (M.M.); (A.T.); (J.W.)
- Correspondence: ; Tel.: +49-421-200-3246
| |
Collapse
|
11
|
Olker JH, Korte JJ, Denny JS, Haselman JT, Hartig PC, Cardon MC, Hornung MW, Degitz SJ. In vitro screening for chemical inhibition of the iodide recycling enzyme, iodotyrosine deiodinase. Toxicol In Vitro 2020; 71:105073. [PMID: 33352258 DOI: 10.1016/j.tiv.2020.105073] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/25/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023]
Abstract
The iodide recycling enzyme, iodotyrosine deiodinase (IYD), is a largely unstudied molecular mechanism through which environmental chemicals can potentially cause thyroid disruption. This highly conserved enzyme plays an essential role in maintaining adequate levels of free iodide for thyroid hormone synthesis. Thyroid disruption following in vivo IYD inhibition has been documented in mammalian and amphibian models; however, few chemicals have been tested for IYD inhibition in either in vivo or in vitro assays. Presented here are the development and application of a screening assay to assess susceptibility of IYD to chemical inhibition. With recombinant human IYD enzyme, a 96-well plate in vitro assay was developed and then used to screen over 1800 unique substances from the U.S. EPA ToxCast screening library. Through a tiered screening approach, 194 IYD inhibitors were identified (inhibited IYD enzyme activity by 20% or greater at target concentration of 200 μM). 154 chemicals were further tested in concentration-response (0.032-200 μM) to determine IC50 and rank-order potency. This work broadens the coverage of thyroid-relevant molecular targets for chemical screening, provides the largest set of chemicals tested for IYD inhibition, and aids in prioritizing chemicals for targeted in vivo testing to evaluate thyroid-related adverse outcomes.
Collapse
Affiliation(s)
- Jennifer H Olker
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, Duluth, MN 55804, USA.
| | - Joseph J Korte
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, Duluth, MN 55804, USA
| | - Jeffrey S Denny
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, Duluth, MN 55804, USA
| | - Jonathan T Haselman
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, Duluth, MN 55804, USA
| | - Phillip C Hartig
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Public Health and Environmental Assessment, Research Triangle Park, North Carolina 27709, USA
| | - Mary C Cardon
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Public Health and Environmental Assessment, Research Triangle Park, North Carolina 27709, USA
| | - Michael W Hornung
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, Duluth, MN 55804, USA
| | - Sigmund J Degitz
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, Duluth, MN 55804, USA
| |
Collapse
|
12
|
Lossow K, Kopp JF, Schwarz M, Finke H, Winkelbeiner N, Renko K, Meçi X, Ott C, Alker W, Hackler J, Grune T, Schomburg L, Haase H, Schwerdtle T, Kipp AP. Aging affects sex- and organ-specific trace element profiles in mice. Aging (Albany NY) 2020; 12:13762-13790. [PMID: 32620712 PMCID: PMC7377894 DOI: 10.18632/aging.103572] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/13/2020] [Indexed: 12/18/2022]
Abstract
A decline of immune responses and dynamic modulation of the redox status are observed during aging and are influenced by trace elements such as copper, iodine, iron, manganese, selenium, and zinc. So far, analytical studies have focused mainly on single trace elements. Therefore, we aimed to characterize age-specific profiles of several trace elements simultaneously in serum and organs of adult and old mice. This allows for correlating multiple trace element levels and to identify potential patterns of age-dependent alterations. In serum, copper and iodine concentrations were increased and zinc concentration was decreased in old as compared to adult mice. In parallel, decreased copper and elevated iron concentrations were observed in liver. The age-related reduction of hepatic copper levels was associated with reduced expression of copper transporters, whereas the increased hepatic iron concentrations correlated positively with proinflammatory mediators and Nrf2-induced ferritin H levels. Interestingly, the age-dependent inverse regulation of copper and iron was unique for the liver and not observed in any other organ. The physiological importance of alterations in the iron/copper ratio for liver function and the aging process needs to be addressed in further studies.
Collapse
Affiliation(s)
- Kristina Lossow
- Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany.,Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,German Institute of Human Nutrition, Nuthetal, Germany.,TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany
| | - Johannes F Kopp
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany
| | - Maria Schwarz
- Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany.,TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany
| | - Hannah Finke
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Nicola Winkelbeiner
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany
| | - Kostja Renko
- Institute for Experimental Endocrinology, Charité University Medical School Berlin, Berlin, Germany.,German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Xheni Meçi
- Institute for Experimental Endocrinology, Charité University Medical School Berlin, Berlin, Germany
| | - Christiane Ott
- German Institute of Human Nutrition, Nuthetal, Germany.,DZHK German Centre for Cardiovascular Research, Berlin, Germany
| | - Wiebke Alker
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany.,Department of Food Chemistry and Toxicology, Technische Universität Berlin, Berlin, Germany
| | - Julian Hackler
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany.,Institute for Experimental Endocrinology, Charité University Medical School Berlin, Berlin, Germany
| | - Tilman Grune
- German Institute of Human Nutrition, Nuthetal, Germany
| | - Lutz Schomburg
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany.,Institute for Experimental Endocrinology, Charité University Medical School Berlin, Berlin, Germany
| | - Hajo Haase
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany.,Department of Food Chemistry and Toxicology, Technische Universität Berlin, Berlin, Germany
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany.,German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Anna P Kipp
- Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany.,TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Germany
| |
Collapse
|
13
|
Kortenkamp A, Axelstad M, Baig AH, Bergman Å, Bornehag CG, Cenijn P, Christiansen S, Demeneix B, Derakhshan A, Fini JB, Frädrich C, Hamers T, Hellwig L, Köhrle J, Korevaar TI, Lindberg J, Martin O, Meima ME, Mergenthaler P, Nikolov N, Du Pasquier D, Peeters RP, Platzack B, Ramhøj L, Remaud S, Renko K, Scholze M, Stachelscheid H, Svingen T, Wagenaars F, Wedebye EB, Zoeller RT. Removing Critical Gaps in Chemical Test Methods by Developing New Assays for the Identification of Thyroid Hormone System-Disrupting Chemicals-The ATHENA Project. Int J Mol Sci 2020; 21:E3123. [PMID: 32354186 PMCID: PMC7247692 DOI: 10.3390/ijms21093123] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/20/2020] [Accepted: 04/24/2020] [Indexed: 11/30/2022] Open
Abstract
The test methods that currently exist for the identification of thyroid hormone system-disrupting chemicals are woefully inadequate. There are currently no internationally validated in vitro assays, and test methods that can capture the consequences of diminished or enhanced thyroid hormone action on the developing brain are missing entirely. These gaps put the public at risk and risk assessors in a difficult position. Decisions about the status of chemicals as thyroid hormone system disruptors currently are based on inadequate toxicity data. The ATHENA project (Assays for the identification of Thyroid Hormone axis-disrupting chemicals: Elaborating Novel Assessment strategies) has been conceived to address these gaps. The project will develop new test methods for the disruption of thyroid hormone transport across biological barriers such as the blood-brain and blood-placenta barriers. It will also devise methods for the disruption of the downstream effects on the brain. ATHENA will deliver a testing strategy based on those elements of the thyroid hormone system that, when disrupted, could have the greatest impact on diminished or enhanced thyroid hormone action and therefore should be targeted through effective testing. To further enhance the impact of the ATHENA test method developments, the project will develop concepts for better international collaboration and development in the area of thyroid hormone system disruptor identification and regulation.
Collapse
Affiliation(s)
- Andreas Kortenkamp
- Institute of Environment, Health and Societies, Brunel University London, Uxbridge UB8 3PH, UK
| | - Marta Axelstad
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Asma H. Baig
- Institute of Environment, Health and Societies, Brunel University London, Uxbridge UB8 3PH, UK
| | - Åke Bergman
- School of Science and Technology, Orebro University, SE-701 82 Orebro, Sweden
| | | | - Peter Cenijn
- Department of Environment and Health, Vrije Universiteit Amsterdam, VUA, 1081 HV Amsterdam, The Netherlands
| | - Sofie Christiansen
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Barbara Demeneix
- Unité PhyMA Laboratory, Adaptation du Vivant, Muséum national d’Histoire naturelle, Centre National de la Recherche Scientifique CNRS 7, rue Cuvier, F-75005 Paris, France
| | - Arash Derakhshan
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Centre, 3000 CA Rotterdam, The Netherlands
| | - Jean-Baptiste Fini
- Unité PhyMA Laboratory, Adaptation du Vivant, Muséum national d’Histoire naturelle, Centre National de la Recherche Scientifique CNRS 7, rue Cuvier, F-75005 Paris, France
| | - Caroline Frädrich
- Department of Experimental Endocrinology, Charitė - Universitätsmedizin Berlin, D-13353 Berlin, Germany
| | - Timo Hamers
- Department of Environment and Health, Vrije Universiteit Amsterdam, VUA, 1081 HV Amsterdam, The Netherlands
| | - Lina Hellwig
- Dept. of Experimental Neurology, Dept. of Neurology, Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, D-10117 Berlin, Germany
- Charité-BIH Centrum Therapy and Research, BIH Stem Cell Core Facility, Charité – Universitätsmedizin Berlin, D-13353 Berlin, Germany
| | - Josef Köhrle
- Department of Experimental Endocrinology, Charitė - Universitätsmedizin Berlin, D-13353 Berlin, Germany
| | - Tim I.M. Korevaar
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Centre, 3000 CA Rotterdam, The Netherlands
| | - Johan Lindberg
- Department of C4hemical Process and Pharmaceutical Development, Research Institutes Sweden, RISE, SE-151 36 Sodertalje, Sweden
| | - Olwenn Martin
- Institute of Environment, Health and Societies, Brunel University London, Uxbridge UB8 3PH, UK
| | - Marcel E. Meima
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Centre, 3000 CA Rotterdam, The Netherlands
| | - Philipp Mergenthaler
- Dept. of Experimental Neurology, Dept. of Neurology, Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, D-10117 Berlin, Germany
- Berlin Institute of Health, D-10178 Berlin, Germany
| | - Nikolai Nikolov
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | | | - Robin P. Peeters
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus Medical Centre, 3000 CA Rotterdam, The Netherlands
| | - Bjorn Platzack
- Department of C4hemical Process and Pharmaceutical Development, Research Institutes Sweden, RISE, SE-151 36 Sodertalje, Sweden
| | - Louise Ramhøj
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Sylvie Remaud
- Unité PhyMA Laboratory, Adaptation du Vivant, Muséum national d’Histoire naturelle, Centre National de la Recherche Scientifique CNRS 7, rue Cuvier, F-75005 Paris, France
| | - Kostja Renko
- Department of Experimental Endocrinology, Charitė - Universitätsmedizin Berlin, D-13353 Berlin, Germany
| | - Martin Scholze
- Institute of Environment, Health and Societies, Brunel University London, Uxbridge UB8 3PH, UK
| | - Harald Stachelscheid
- Charité-BIH Centrum Therapy and Research, BIH Stem Cell Core Facility, Charité – Universitätsmedizin Berlin, D-13353 Berlin, Germany
- Berlin Institute of Health, D-10178 Berlin, Germany
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - Fabian Wagenaars
- Department of Environment and Health, Vrije Universiteit Amsterdam, VUA, 1081 HV Amsterdam, The Netherlands
| | - Eva Bay Wedebye
- National Food Institute, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
| | - R. Thomas Zoeller
- School of Science and Technology, Orebro University, SE-701 82 Orebro, Sweden
| |
Collapse
|
14
|
Groeneweg S, van Geest FS, Peeters RP, Heuer H, Visser WE. Thyroid Hormone Transporters. Endocr Rev 2020; 41:5637505. [PMID: 31754699 DOI: 10.1210/endrev/bnz008] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ferdy S van Geest
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - W Edward Visser
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
15
|
Lademann F, Tsourdi E, Rijntjes E, Köhrle J, Hofbauer LC, Heuer H, Rauner M. Lack of the Thyroid Hormone Transporter Mct8 in Osteoblast and Osteoclast Progenitors Increases Trabecular Bone in Male Mice. Thyroid 2020; 30:329-342. [PMID: 31910109 DOI: 10.1089/thy.2019.0271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Bone is an important target of thyroid hormones (THs), which require transport into target cells to exert their actions. Recently, the TH-specific monocarboxylate transporter 8 (Mct8) was reported as a regulator of bone mass in male mice. However, its global deletion leads to high 3,3',5-L-triiodothyronine (T3) serum concentrations that may mask direct effects of Mct8-deficiency on bone. In this study, we assessed the bone cell intrinsic function of Mct8 ex vivo and in vivo using conditional Mct8-knockout lines specifically targeting osteoclast and osteoblast progenitors, as well as mature osteoblasts and osteocytes. Materials and Methods: Twelve-week-old male mice with a global Mct8-deficiency or a conditional Mct8-knockout in osteoclast precursors, osteoprogenitors, or mature osteoblasts/osteocytes were analyzed regarding their bone microarchitecture, turnover, and strength. Furthermore, ex vivo studies were conducted to investigate the role of Mct8 in bone cell differentiation and functionality, as well as TH uptake. Results: Global Mct8-knockout mice demonstrated 1.7-fold higher T3 serum concentrations and trabecular bone loss (-28%) likely due to an increased bone turnover as shown by increased osteoblast (+45%) and osteoclast numbers (+41%). However, cortical bone mineral density was increased. Ex vivo cultures of bone marrow-derived osteoblasts and osteoclasts revealed highest expression of Mct8 in mature bone cells. In addition, Mct8-deficiency resulted in a lower mRNA expression of osteoblast and osteoclast differentiation markers, as well as a reduced mineralization capacity and osteoclast numbers, respectively, indicating a bone cell intrinsic role of Mct8. In fact, conditional Mct8-knockout and inhibition of Mct8 in osteoblasts led to an attenuated T3 uptake ex vivo. In vivo, osteoprogenitor-specific Mct8-knockout enhanced trabecular bone volume (+16%) with osteoblast numbers being increased 3.7 fold. Interestingly, Mct8-deficiency in osteoprogenitors and late osteoblasts/osteocytes both resulted in cortical bone loss. Finally, Mct8-deletion in osteoclast progenitors increased trabecular bone volume (+20%) due to reduced osteoclast numbers (-32%), whereas osteoblast numbers were enhanced (+25%). Conclusions: This study confirms that high systemic T3 in global Mct8-knockout mice masks the direct effect of Mct8. Moreover, it identifies Mct8 as a critical regulator of trabecular vs. cortical bone by regulating T3 uptake and highlights its cell intrinsic role in osteoclast and osteoblast progenitors.
Collapse
Affiliation(s)
- Franziska Lademann
- Department of Medicine III, Universitätsklinikum Dresden, Dresden, Germany
- Center for Healthy Aging, Universitätsklinikum Dresden, Dresden, Germany
| | - Elena Tsourdi
- Department of Medicine III, Universitätsklinikum Dresden, Dresden, Germany
- Center for Healthy Aging, Universitätsklinikum Dresden, Dresden, Germany
| | - Eddy Rijntjes
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Universitätsklinikum Dresden, Dresden, Germany
- Center for Healthy Aging, Universitätsklinikum Dresden, Dresden, Germany
| | - Heike Heuer
- Klinik für Endokrinologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Martina Rauner
- Department of Medicine III, Universitätsklinikum Dresden, Dresden, Germany
- Center for Healthy Aging, Universitätsklinikum Dresden, Dresden, Germany
| |
Collapse
|
16
|
Olker JH, Korte JJ, Denny JS, Hartig PC, Cardon MC, Knutsen CN, Kent PM, Christensen JP, Degitz SJ, Hornung MW. Screening the ToxCast Phase 1, Phase 2, and e1k Chemical Libraries for Inhibitors of Iodothyronine Deiodinases. Toxicol Sci 2019; 168:430-442. [PMID: 30561685 PMCID: PMC6520049 DOI: 10.1093/toxsci/kfy302] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Deiodinase enzymes play an essential role in converting thyroid hormones between active and inactive forms by deiodinating the pro-hormone thyroxine (T4) to the active hormone triiodothyronine (T3) and modifying T4 and T3 to inactive forms. Chemical inhibition of deiodinase activity has been identified as an important endpoint to include in screening chemicals for thyroid hormone disruption. To address the lack of data regarding chemicals that inhibit the deiodinase enzymes, we developed robust in vitro assays that utilized human deiodinase types 1, 2, and 3 and screened over 1800 unique chemicals from the U.S. EPA's ToxCast phase 1_v2, phase 2, and e1k libraries. Initial testing at a single concentration identified 411 putative deiodinase inhibitors that produced inhibition of 20% or greater in at least 1 of the 3 deiodinase assays, including chemicals that have not previously been shown to inhibit deiodinases. Of these, 228 chemicals produced enzyme inhibition of 50% or greater; these chemicals were further tested in concentration-response to determine relative potency. Comparisons across these deiodinase assays identified 81 chemicals that produced selective inhibition, with 50% inhibition or greater of only 1 of the deiodinases. This set of 3 deiodinase inhibition assays provides a significant contribution toward expanding the limited number of in vitro assays used to identify chemicals with the potential to interfere with thyroid hormone homeostasis. In addition, these results set the groundwork for development and evaluation of structure-activity relationships for deiodinase inhibition, and inform targeted selection of chemicals for further testing to identify adverse outcomes of deiodinase inhibition.
Collapse
Affiliation(s)
- Jennifer H. Olker
- U.S. Environmental Protection Agency, Duluth, Minnesota, 55804
- Office of Research and Development, Duluth, Minnesota, 55804
- National Health and Environmental Effects Research Laboratory, Duluth, Minnesota, 55804
- Mid-Continent Ecology Division, Duluth, Minnesota, 55804
| | - Joseph J. Korte
- U.S. Environmental Protection Agency, Duluth, Minnesota, 55804
- Office of Research and Development, Duluth, Minnesota, 55804
- National Health and Environmental Effects Research Laboratory, Duluth, Minnesota, 55804
- Mid-Continent Ecology Division, Duluth, Minnesota, 55804
| | - Jeffrey S. Denny
- U.S. Environmental Protection Agency, Duluth, Minnesota, 55804
- Office of Research and Development, Duluth, Minnesota, 55804
- National Health and Environmental Effects Research Laboratory, Duluth, Minnesota, 55804
- Mid-Continent Ecology Division, Duluth, Minnesota, 55804
| | - Phillip C. Hartig
- U.S. Environmental Protection Agency, Duluth, Minnesota, 55804
- Office of Research and Development, Duluth, Minnesota, 55804
- National Health and Environmental Effects Research Laboratory, Duluth, Minnesota, 55804
- Toxicity Assessment Division, Research Triangle Park, North Carolina 27709
| | - Mary C. Cardon
- U.S. Environmental Protection Agency, Duluth, Minnesota, 55804
- Office of Research and Development, Duluth, Minnesota, 55804
- National Health and Environmental Effects Research Laboratory, Duluth, Minnesota, 55804
- Toxicity Assessment Division, Research Triangle Park, North Carolina 27709
| | - Carsten N. Knutsen
- Student Services Contractor to the U.S. EPA, NHEERL, Mid-Continent Ecology Division, Duluth, Minnesota, 55804
| | - Paige M. Kent
- ORAU Student Services Contractor to the U.S. EPA, NHEERL, Mid-Continent Ecology Division, Duluth, Minnesota, 55804
| | - Jessica P. Christensen
- ORAU Student Services Contractor to the U.S. EPA, NHEERL, Mid-Continent Ecology Division, Duluth, Minnesota, 55804
| | - Sigmund J. Degitz
- U.S. Environmental Protection Agency, Duluth, Minnesota, 55804
- Office of Research and Development, Duluth, Minnesota, 55804
- National Health and Environmental Effects Research Laboratory, Duluth, Minnesota, 55804
- Mid-Continent Ecology Division, Duluth, Minnesota, 55804
| | - Michae1 W. Hornung
- U.S. Environmental Protection Agency, Duluth, Minnesota, 55804
- Office of Research and Development, Duluth, Minnesota, 55804
- National Health and Environmental Effects Research Laboratory, Duluth, Minnesota, 55804
- Mid-Continent Ecology Division, Duluth, Minnesota, 55804
| |
Collapse
|
17
|
Characterization of non-radiolabeled Thyroxine (T 4) uptake in cryopreserved rat hepatocyte suspensions: Pharmacokinetic implications for PFOA and PFOS chemical exposure. Toxicol In Vitro 2019; 58:230-238. [PMID: 30930230 DOI: 10.1016/j.tiv.2019.03.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/28/2019] [Accepted: 03/18/2019] [Indexed: 11/22/2022]
Abstract
The alteration of thyroxine (T4) cellular uptake by an environmental chemical can serve as a contributing factor in thyroid hormone (TH) disruption. Herein, we describe a non-radiolabeled (LC-MS/MS) oil-filtration technique designed to characterize the mechanism(s) responsible for T4 cellular uptake in cryopreserved rat hepatocyte suspensions. The environmental chemicals perfluorooctanoate (PFOA) and perfluorooctane sulfonate (PFOS) were evaluated for their effect on T4 hepatic uptake. At 37 °C, hepatic assays demonstrated saturable kinetics with increasing T4 concentrations, while a linear uptake rate consistent with passive diffusion was detected at 4 °C. Carrier-mediated (37-4 °C) transport of T4 was the predominant hepatic uptake process versus passive diffusion. Cyclosporin A (CsA) chemically inhibited T4 hepatic uptake, whereas PFOA/PFOS displayed no inhibition of T4 translocation. Increasing PFOA/PFOS concentration levels with the T4 serum carrier-protein transthyretin (TTR) present resulted in a dose-response increase in T4 hepatic uptake rates, correlating with increased T4 free fraction values. Hepatic assays conducted in the presence of PFOA/PFOS and TTR displayed an enhanced first-order T4 hepatic uptake rate consistent with carrier-mediated transport. These in vitro findings characterizing increased T4 hepatic uptake provides mechanistic insight regarding decreased T4 serum levels (hypothyroxinemia) previously observed within in vivo rodent studies following perfluorinated chemical exposure.
Collapse
|
18
|
Comparative Analysis of Zearalenone Effects on Thyroid Receptor Alpha (TRα) and Beta (TRβ) Expression in Rat Primary Cerebellar Cell Cultures. Int J Mol Sci 2018; 19:ijms19051440. [PMID: 29751674 PMCID: PMC5983839 DOI: 10.3390/ijms19051440] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/20/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022] Open
Abstract
Thyroid receptors play an important role in postnatal brain development. Zearalenone (ZEN), a major mycotoxin of Fusarium fungi, is well known to cause serious health problems in animals and humans through various mechanisms, including the physiological pathways of thyroid hormone (TH). In the present study, we aimed to investigate the expression of thyroid receptors α (TRα) and β (TRβ) in primary cerebellar neurons in the presence or absence of glia and following ZEN treatment, using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot. Primary cerebellar granule cells were treated with low doses of ZEN (0.1 nM) in combination with physiologically relevant concentrations of l-thyroxine (T4), 3,3′,5-triiodo-l-thyronine (T3) and 17β-estradiol (E2). Expression levels of TRα and TRβ at mRNA and protein levels were slightly modified by ZEN administered alone; however, along with thyroid and steroid hormones, modelling the physiological conditions, expression levels of TRs varied highly depending on the given treatment. Gene expression levels were also highly modulated by the presence or absence of glial cells, with mostly contrasting effects. Our results demonstrate divergent transcriptional and translational mechanisms involved in the expression of TRs implied by ZEN and hormonal milieu, as well as culturing conditions.
Collapse
|
19
|
Hornung MW, Korte JJ, Olker JH, Denny JS, Knutsen C, Hartig PC, Cardon MC, Degitz SJ. Screening the ToxCast Phase 1 Chemical Library for Inhibition of Deiodinase Type 1 Activity. Toxicol Sci 2018; 162:570-581. [PMID: 29228274 PMCID: PMC6639810 DOI: 10.1093/toxsci/kfx279] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Thyroid hormone (TH) homeostasis is dependent upon coordination of multiple key events including iodide uptake, hormone synthesis, metabolism, and elimination, to maintain proper TH signaling. Deiodinase enzymes catalyze iodide release from THs to interconvert THs between active and inactive forms, and are integral to hormone metabolism. The activity of deiodinases has been identified as an important endpoint to include in the context of screening chemicals for TH disruption. To begin to address the potential for chemicals to inhibit these enzymes an adenovirus expression system was used to produce human deiodinase type 1 (DIO1) enzyme, established robust assay parameters for nonradioactive determination of iodide release by the Sandell-Kolthoff method, and employed a 96-well plate format for screening chemical libraries. An initial set of 18 chemicals was used to establish the assay, along with the known DIO1 inhibitor 6-propylthiouracil as a positive control. An additional 292 unique chemicals from the EPA's ToxCast phase 1_v2 chemical library were screened. Chemicals were initially screened at a single high concentration of 200 µM to identify potential DIO1 inhibitors. There were 50 chemicals, or 17% of the TCp1_v2 chemicals tested, that produced >20% inhibition of DIO1 activity. Eighteen of these inhibited DIO1 activity >50% and were further tested in concentration-response mode to determine IC50s. This work presents an initial effort toward identifying chemicals with potential for affecting THs via inhibition of deiodinases and sets the foundation for further testing of large chemical libraries against DIO1 and the other deiodinase enzymes involved in TH function.
Collapse
Affiliation(s)
- Michael W. Hornung
- US Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, Duluth, MN, 55804, USA
| | - Joseph J. Korte
- US Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, Duluth, MN, 55804, USA
| | - Jennifer H. Olker
- US Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, Duluth, MN, 55804, USA
| | - Jeffrey S. Denny
- US Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, Duluth, MN, 55804, USA
| | - Carsten Knutsen
- US Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, Duluth, MN, 55804, USA
| | - Phillip C. Hartig
- US Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Toxicity Assessment Division, Research Triangle Park, 27709, USA
| | - Mary C. Cardon
- US Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Toxicity Assessment Division, Research Triangle Park, 27709, USA
| | - Sigmund J. Degitz
- US Environmental Protection Agency, Office of Research and Development, National Health and Environmental Effects Research Laboratory, Mid-Continent Ecology Division, Duluth, MN, 55804, USA
| |
Collapse
|
20
|
Saha S, Chakraborty S, Bhattacharya A, Biswas A, Ain R. MicroRNA regulation of Transthyretin in trophoblast differentiation and Intra-Uterine Growth Restriction. Sci Rep 2017; 7:16548. [PMID: 29185488 PMCID: PMC5707432 DOI: 10.1038/s41598-017-16566-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023] Open
Abstract
Placental trophoblast cells produce various cytokines, transporters vital to normal embryogenesis. Transthyretin (TTR) aids trans-placental passage of maternal thyroxin (TH) to fetal circulation. Inadequate TH delivery leads to developmental abnormality. Regulation of TTR biosynthesis in placenta is critical for normal embryo development. We showed here that TTR transcripts were expressed more in fetal placenta. Using bioinformatic analysis and confirmation with dual-luciferase reporter assays, we found that miR-200a-3p and miR-141-3p inhibited TTR expression by directly binding to the 3'UTR of TTR, which is reversed by mutation in the microRNA binding site. Differentiation of human trophoblast BeWo cells was associated with decreased TTR transcript and protein levels with concomitant increase in the levels of both microRNAs. Interestingly, ectopic overexpression of the microRNA mimics abrogated thyroxin uptake by BeWo cells, which was reversed by the corresponding inhibitors. Furthermore, in a rat model of intra-uterine growth restriction (IUGR), TTR expression decreased significantly in placenta with reciprocal rise in miR-141-3p but not 200a-3p. In human IUGR placenta, TTR transcript and protein levels were significantly lower associated with high expression of miR-141-3p but not 200a-3p. These data provides new insight into physiological role of miR-141-3p in regulating TTR during trophoblast differentiation and IUGR.
Collapse
Affiliation(s)
- Sarbani Saha
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Shreeta Chakraborty
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | | | - Arati Biswas
- Calcutta National Medical College, Kolkata, 700014, India
| | - Rupasri Ain
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, 700032, India.
| |
Collapse
|
21
|
Dong H, Wade MG. Application of a nonradioactive assay for high throughput screening for inhibition of thyroid hormone uptake via the transmembrane transporter MCT8. Toxicol In Vitro 2017; 40:234-242. [DOI: 10.1016/j.tiv.2017.01.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 12/11/2022]
|
22
|
Abstract
Thyroid hormones (TH) are endocrine messengers essential for normal development and function of virtually every vertebrate. The hypothalamic-pituitary-thyroid axis is exquisitely modulated to maintain nearly constant TH (T4 and T3) levels in circulation. However peripheral tissues and the CNS control the intracellular availability of TH, suggesting that circulating concentrations of TH are not fully representative of what each cell type sees. Indeed, recent work in the field has identified that TH transporters, deiodinases and thyroid hormone receptor coregulators can strongly control tissue-specific sensitivity to a set amount of TH. Furthermore, the mechanism by which the thyroid hormone receptors regulate target gene expression can vary by gene, tissue and cellular context. This review will highlight novel insights into the machinery that controls the cellular response to TH, which include unique signaling cascades. These findings shed new light into the pathophysiology of human diseases caused by abnormal TH signaling.
Collapse
Affiliation(s)
- Arturo Mendoza
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Johannes J, Jayarama-Naidu R, Meyer F, Wirth EK, Schweizer U, Schomburg L, Köhrle J, Renko K. Silychristin, a Flavonolignan Derived From the Milk Thistle, Is a Potent Inhibitor of the Thyroid Hormone Transporter MCT8. Endocrinology 2016; 157:1694-701. [PMID: 26910310 DOI: 10.1210/en.2015-1933] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Thyroid hormones (THs) are charged and iodinated amino acid derivatives that need to pass the cell membrane facilitated by thyroid hormone transmembrane transporters (THTT) to exert their biological function. The importance of functional THTT is affirmed by the devastating effects of mutations in the human monocarboxylate transporter (MCT) 8, leading to a severe form of psychomotor retardation. Modulation of THTT function by pharmacological or environmental compounds might disturb TH action on a tissue-specific level. Therefore, it is important to identify compounds with relevant environmental exposure and THTT-modulating activity. Based on a nonradioactive TH uptake assay, we performed a screening of 13 chemicals, suspicious for TH receptor interaction, to test their potential effects on THTT in MCT8-overexpressing MDCK1-cells. We identified silymarin, an extract of the milk thistle, to be a potent inhibitor of T3 uptake by MCT8. Because silymarin is a complex mixture of flavonolignan substances, we further tested its individual components and identified silychristin as the most effective one with an IC50 of approximately 100 nM. The measured IC50 value is at least 1 order of magnitude below those of other known THTT inhibitors. This finding was confirmed by T3 uptake in primary murine astrocytes expressing endogenous Mct8 but not in MCT10-overexpressing MDCK1-cells, indicating a remarkable specificity of the inhibitor toward MCT8. Because silymarin is a frequently used adjuvant therapeutic for hepatitis C infection and chronic liver disease, our observations raise questions regarding its safety with respect to unwanted effects on the TH axis.
Collapse
Affiliation(s)
- Jörg Johannes
- Institut für Experimentelle Endokrinologie (R.J.-N., F.M., E.K.W., L.S., J.K., K.R.), Charité-Universitätsmedizin Berlin, D-13353 Berlin, Germany; and Institut für Biochemie und Molekularbiologie (J.J., U.S.), Rheinische Friedrich-Wilhelms-Universität, D-53115 Bonn, Germany
| | - Roopa Jayarama-Naidu
- Institut für Experimentelle Endokrinologie (R.J.-N., F.M., E.K.W., L.S., J.K., K.R.), Charité-Universitätsmedizin Berlin, D-13353 Berlin, Germany; and Institut für Biochemie und Molekularbiologie (J.J., U.S.), Rheinische Friedrich-Wilhelms-Universität, D-53115 Bonn, Germany
| | - Franziska Meyer
- Institut für Experimentelle Endokrinologie (R.J.-N., F.M., E.K.W., L.S., J.K., K.R.), Charité-Universitätsmedizin Berlin, D-13353 Berlin, Germany; and Institut für Biochemie und Molekularbiologie (J.J., U.S.), Rheinische Friedrich-Wilhelms-Universität, D-53115 Bonn, Germany
| | - Eva Katrin Wirth
- Institut für Experimentelle Endokrinologie (R.J.-N., F.M., E.K.W., L.S., J.K., K.R.), Charité-Universitätsmedizin Berlin, D-13353 Berlin, Germany; and Institut für Biochemie und Molekularbiologie (J.J., U.S.), Rheinische Friedrich-Wilhelms-Universität, D-53115 Bonn, Germany
| | - Ulrich Schweizer
- Institut für Experimentelle Endokrinologie (R.J.-N., F.M., E.K.W., L.S., J.K., K.R.), Charité-Universitätsmedizin Berlin, D-13353 Berlin, Germany; and Institut für Biochemie und Molekularbiologie (J.J., U.S.), Rheinische Friedrich-Wilhelms-Universität, D-53115 Bonn, Germany
| | - Lutz Schomburg
- Institut für Experimentelle Endokrinologie (R.J.-N., F.M., E.K.W., L.S., J.K., K.R.), Charité-Universitätsmedizin Berlin, D-13353 Berlin, Germany; and Institut für Biochemie und Molekularbiologie (J.J., U.S.), Rheinische Friedrich-Wilhelms-Universität, D-53115 Bonn, Germany
| | - Josef Köhrle
- Institut für Experimentelle Endokrinologie (R.J.-N., F.M., E.K.W., L.S., J.K., K.R.), Charité-Universitätsmedizin Berlin, D-13353 Berlin, Germany; and Institut für Biochemie und Molekularbiologie (J.J., U.S.), Rheinische Friedrich-Wilhelms-Universität, D-53115 Bonn, Germany
| | - Kostja Renko
- Institut für Experimentelle Endokrinologie (R.J.-N., F.M., E.K.W., L.S., J.K., K.R.), Charité-Universitätsmedizin Berlin, D-13353 Berlin, Germany; and Institut für Biochemie und Molekularbiologie (J.J., U.S.), Rheinische Friedrich-Wilhelms-Universität, D-53115 Bonn, Germany
| |
Collapse
|