1
|
Polyzos SA, Targher G. Hepatic thyroid hormone receptor-β signalling: Mechanisms and recent advancements in the treatment of metabolic dysfunction-associated steatohepatitis. Diabetes Obes Metab 2024. [PMID: 39658733 DOI: 10.1111/dom.16117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/23/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024]
Abstract
The pharmacotherapy of metabolic dysfunction-associated steatotic liver disease (MASLD) and its progressive form, the metabolic dysfunction-associated steatohepatitis (MASH), remains a hot topic in research and a largely unmet need in clinical practice. As the first approval of a disease-specific drug, resmetirom, was regarded as a milestone for the management of this common liver disease, this comprehensive and updated review aimed to highlight the importance of the hepatic thyroid hormone (TH) receptor (THR)-β signalling for the treatment of MASH, with a special focus on resmetirom. First, the genomic and non-genomic actions of the liver-directed THR-β mediated mechanisms are summarized. THR-β has a key role in hepatic lipid and carbohydrate metabolism; disruption of THR-β signalling leads to dysmetabolism, thus promoting MASLD and possibly its progression to MASH and cirrhosis. In the clinical setting, this is translated into a significant association between primary hypothyroidism and MASLD, as confirmed by recent meta-analyses. An association between MASLD and subclinical intrahepatic hypothyroidism (i.e. a state of relatively low hepatic triiodothyronine concentrations, with circulating TH concentrations within the normal range) is also emerging and under investigation. In line with this, the favourable results of the phase 3 placebo-controlled MAESTRO trials led to the recent conditional approval of resmetirom by the US FDA for treating adults with MASH and moderate-to-advanced fibrosis. This conditional approval of resmetirom opened a new window to the management of this common and burdensome liver disease, thus bringing the global scientific community in front of new perspectives and challenges.
Collapse
Affiliation(s)
- Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar di Valpolicella (VR), Italy
| |
Collapse
|
2
|
Hönes GS, Geist D, Wenzek C, Pfluger PT, Müller TD, Aguilar-Pimentel JA, Amarie OV, Becker L, Dragano N, Garrett L, Hölter SM, Rathkolb B, Rozman J, Spielmann N, Treise I, Wolf E, Wurst W, Fuchs H, Gailus-Durner V, Hrabe de Angelis M, Führer D, Moeller LC. Comparative Phenotyping of Mice Reveals Canonical and Noncanonical Physiological Functions of TRα and TRβ. Endocrinology 2024; 165:bqae067. [PMID: 38889231 DOI: 10.1210/endocr/bqae067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/14/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024]
Abstract
Thyroid hormone (TH) effects are mediated through TH receptors (TRs), TRα1, TRβ1, and TRβ2. The TRs bind to the DNA and regulate expression of TH target genes (canonical signaling). In addition, they mediate activation of signaling pathways (noncanonical signaling). Whether noncanonical TR action contributes to the spectrum of TH effects is largely unknown. The aim of this study was to attribute physiological effects to the TR isoforms and their canonical and noncanonical signaling. We conducted multiparameter phenotyping in male and female TR knockout mice (TRαKO, TRβKO), mice with disrupted canonical signaling due to mutations in the TR DNA binding domain (TRαGS, TRβGS), and their wild-type littermates. Perturbations in senses, especially hearing (mainly TRβ with a lesser impact of TRα), visual acuity, retinal thickness (TRα and TRβ), and in muscle metabolism (TRα) highlighted the role of canonical TR action. Strikingly, selective abrogation of canonical TR action often had little phenotypic consequence, suggesting that noncanonical TR action sufficed to maintain the wild-type phenotype for specific effects. For instance, macrocytic anemia, reduced retinal vascularization, or increased anxiety-related behavior were only observed in TRαKO but not TRαGS mice. Noncanonical TRα action improved energy utilization and prevented hyperphagia observed in female TRαKO mice. In summary, by examining the phenotypes of TRα and TRβ knockout models alongside their DNA binding-deficient mutants and wild-type counterparts, we could establish that the noncanonical actions of TRα and TRβ play a crucial role in modulating sensory, behavioral, and metabolic functions and, thus, contribute to the spectrum of physiological TH effects.
Collapse
Affiliation(s)
- Georg Sebastian Hönes
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Daniela Geist
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Christina Wenzek
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Paul Thomas Pfluger
- Research Unit NeuroBiology of Diabetes, Helmholtz Zentrum München, Neuherberg 85764, Germany
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg 85764, Germany
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich 80333, Germany
| | - Timo Dirk Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Neuherberg 85764, Germany
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University (LMU) Munich, Munich 80336, Germany
| | - Juan Antonio Aguilar-Pimentel
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Oana Veronica Amarie
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Natalia Dragano
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Lillian Garrett
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Sabine Maria Hölter
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Birgit Rathkolb
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians University (LMU) Munich, Munich 81377, Germany
| | - Jan Rozman
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Nadine Spielmann
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Irina Treise
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians University (LMU) Munich, Munich 81377, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich 80336, Germany
- Chair of Developmental Genetics, TUM School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Martin Hrabe de Angelis
- German Center for Diabetes Research, Neuherberg 85764, Germany
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
- Chair of Experimental Genetics, TUM School of Life Science Weihenstephan, Technical University of Munich, Freising 85354, Germany
| | - Dagmar Führer
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Lars Christian Moeller
- Department of Endocrinology, Diabetes and Metabolism and Division of Laboratory Research, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| |
Collapse
|
3
|
Sinha RA, Yen PM. Metabolic Messengers: Thyroid Hormones. Nat Metab 2024; 6:639-650. [PMID: 38671149 PMCID: PMC7615975 DOI: 10.1038/s42255-024-00986-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 01/15/2024] [Indexed: 04/28/2024]
Abstract
Thyroid hormones (THs) are key hormones that regulate development and metabolism in mammals. In man, the major target tissues for TH action are the brain, liver, muscle, heart, and adipose tissue. Defects in TH synthesis, transport, metabolism, and nuclear action have been associated with genetic and endocrine diseases in man. Over the past few years, there has been renewed interest in TH action and the therapeutic potential of THs and thyromimetics to treat several metabolic disorders such as hypercholesterolemia, dyslipidaemia, non-alcoholic fatty liver disease (NAFLD), and TH transporter defects. Recent advances in the development of tissue and TH receptor isoform-targeted thyromimetics have kindled new hope for translating our fundamental understanding of TH action into an effective therapy. This review provides a concise overview of the historical development of our understanding of TH action, its physiological and pathophysiological effects on metabolism, and future therapeutic applications to treat metabolic dysfunction.
Collapse
Affiliation(s)
- Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
| | - Paul M Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore.
- Div. Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
4
|
Zhao X, Sun Y, Zhang H, Zhang Y, Zhao H, Yao X, Zhang W. Effect of different iodide intake during pregnancy and lactation on thyroid and cardiovascular function in maternal and offspring rats. J Trace Elem Med Biol 2023; 79:127267. [PMID: 37506535 DOI: 10.1016/j.jtemb.2023.127267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
OBJECTIVE We aimed to investigate the impact of different iodide intake during pregnancy and lactation on iodine concentration in urine and serum, fatty acid metabolism, thyroid and cardiovascular function in maternal and offspring rats. METHODS Pregnant rats were randomly assigned to four groups: normal adult iodide intake (NAI, 7.5 μg/d), normal pregnant iodide intake (NPI, 12.5 μg/d), 5 times (5 HI, 62.5 μg/d) and 10 times higher-than-normal pregnant iodide intake (10 HI, 125 μg/d). The maternal rats were continuously administered potassium iodide until postnatal day 16 (PN16). Thyroid function was measured by enzyme-linked immunosorbent assay (ELISA). The iodine concentration in urine and serum were detected by inductively coupled plasma mass spectrometry (ICP-MS). The messenger ribonucleic acid (mRNA) expressions of Krüppel-like factor 9 (KLF9) and thioredoxin reductase 2 (Txnrd2) were measured using quantitative real-time polymerase chain reaction (RT-qPCR). Characteristic distribution of KLF9 expression and its interaction with TRβ was assessed by immunohistochemical and immunofluorescence staining. Serum fatty acids were analyzed by Liquid Chromatography-Mass Spectrometry (LC-MS). Cardiac function and blood pressure were measured by echocardiography and a non-invasive tail-cuff system. RESULTS High iodide intake (5 HI and 10 HI) during pregnancy and lactation results in increased urinary iodine concentration (UIC), serum total iodine concentration (STIC) and serum non-protein-bound iodine concentration (SNBIC) in both maternal and offspring rats, along with significantly increased FT3 and its target gene expression of KLF9. In maternal rats of both 5 HI and 10 HI groups, systolic blood pressure (SBP) was significantly higher, the increased SBP was significantly correlated with the increased UIC (r = 0.968, p = 0.002; r = 0.844, p = 0.035), KLF9 (r = 0.935, p = 0.006; r = 0.954, p = 0.003) and the decreased Txnrd2 (r = -0.909, p = 0.012; r = -0.912, p = 0.011). In maternal rats of 10 HI group, cardiac hyperfunction with increased LVEF, LVFS and decreased LVESD were observed. The increased LVEF and decreased LVESD were significantly correlated with UIC, STIC and SNBIC (r = 0.976, p = 0.001; r = 0.945, p = 0.005; r = 0.953, p = 0.003; r = -0.917, p = 0.01; r = -0.859, p = 0.028; r = -0.847, p = 0.033), LVEF, LVFS and LVESD were significant correlated with KLF9 (r = 0.950, p = 0.004; r = 0.963, p = 0.002; r = -0.990, p = 0.0002) and Txnrd2 expression (r = -0.979, p = 0.001; r = -0.915, p = 0.01; r = 0.933, p = 0.007), and the decreased LVESD was correlated with decreased epoxyeicosatrienoic acid (EET) metabolites: 5,6-EET, 8,9-DHET and 11,12-DHET (r = 0.999, p = 0.034; r = 1.000, p = 0.017; r = 1.000, p = 0.017). While in offspring rats, no significant change in SBP and cardiac function was found. STIC and SNBIC were much lower than those in maternal rats, and eicosapentaenoic acid (EPA) metabolites (9-HEPE, 15-HEPE and 14,15 DiHETE) were significantly increased. CONCLUSION In addition to thyroid hormones, STIC, SNBIC, KLF9, Txnrd2, EET and EPA metabolites might be promising biomarkers in high iodide intake-induced thyroid and cardiovascular function.
Collapse
Affiliation(s)
- Xiuxiu Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yue Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hexi Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Yue Zhang
- Tianjin Key Laboratory of Ionic-Molecular of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Hailing Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiaomei Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| | - Wanqi Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
5
|
Mourouzis I, Apostolaki V, Trikas A, Kokkinos L, Alexandrou N, Avdikou M, Giannoulopoulou M, Vassi A, Tseti I, Pantos C. The Potential of Thyroid Hormone Therapy in Severe COVID-19: Rationale and Preliminary Evidence. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19138063. [PMID: 35805716 PMCID: PMC9265958 DOI: 10.3390/ijerph19138063] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023]
Abstract
Tissue hypoxia is one of the main pathophysiologic mechanisms in sepsis and particularly in COVID-19. Microvascular dysfunction, endothelialitis and alterations in red blood cell hemorheology are all implicated in severe COVID-19 hypoxia and multiorgan dysfunction. Tissue hypoxia results in tissue injury and remodeling with re-emergence of fetal programming via hypoxia-inducible factor-1α (HIF-1a)-dependent and -independent pathways. In this context, thyroid hormone (TH), a critical regulator of organ maturation, may be of relevance in preventing fetal-like hypoxia-induced remodeling in COVID-19 sepsis. Acute triiodothyronine (T3) treatment can prevent cardiac remodeling and improve recovery of function in clinical settings of hypoxic injury as acute myocardial infarction and by-pass cardiac surgery. Furthermore, T3 administration prevents tissue hypoxia in experimental sepsis. On the basis of this evidence, the use of T3 treatment was proposed for ICU (Intensive Care Unit) COVID-19 patients (Thy-Support, NCT04348513). The rationale for T3 therapy in severe COVID-19 and preliminary experimental and clinical evidence are discussed in this review.
Collapse
Affiliation(s)
- Iordanis Mourouzis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Ave., Goudi, 11527 Athens, Greece; (I.M.); (V.A.); (A.T.); (I.T.)
| | - Vassiliki Apostolaki
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Ave., Goudi, 11527 Athens, Greece; (I.M.); (V.A.); (A.T.); (I.T.)
| | - Athanasios Trikas
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Ave., Goudi, 11527 Athens, Greece; (I.M.); (V.A.); (A.T.); (I.T.)
| | - Leonidas Kokkinos
- Department of Anesthesiology, ELPIS General Hospital of Athens, 11522 Athens, Greece; (L.K.); (N.A.); (M.A.); (M.G.); (A.V.)
| | - Natassa Alexandrou
- Department of Anesthesiology, ELPIS General Hospital of Athens, 11522 Athens, Greece; (L.K.); (N.A.); (M.A.); (M.G.); (A.V.)
| | - Maria Avdikou
- Department of Anesthesiology, ELPIS General Hospital of Athens, 11522 Athens, Greece; (L.K.); (N.A.); (M.A.); (M.G.); (A.V.)
| | - Myrto Giannoulopoulou
- Department of Anesthesiology, ELPIS General Hospital of Athens, 11522 Athens, Greece; (L.K.); (N.A.); (M.A.); (M.G.); (A.V.)
| | - Aimilia Vassi
- Department of Anesthesiology, ELPIS General Hospital of Athens, 11522 Athens, Greece; (L.K.); (N.A.); (M.A.); (M.G.); (A.V.)
| | - Ioulia Tseti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Ave., Goudi, 11527 Athens, Greece; (I.M.); (V.A.); (A.T.); (I.T.)
| | - Constantinos Pantos
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Ave., Goudi, 11527 Athens, Greece; (I.M.); (V.A.); (A.T.); (I.T.)
- Correspondence: ; Tel.: +30-210-746-2560; Fax: +30-210-746-2562
| |
Collapse
|
6
|
Hönes GS, Härting N, Mittag J, Kaiser FJ. TRα2—An Untuned Second Fiddle or Fine-Tuning Thyroid Hormone Action? Int J Mol Sci 2022; 23:ijms23136998. [PMID: 35806002 PMCID: PMC9266318 DOI: 10.3390/ijms23136998] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Thyroid hormones (THs) control a wide range of physiological functions essential for metabolism, growth, and differentiation. On a molecular level, TH action is exerted by nuclear receptors (TRs), which function as ligand-dependent transcription factors. Among several TR isoforms, the function of TRα2 remains poorly understood as it is a splice variant of TRα with an altered C-terminus that is unable to bind T3. This review highlights the molecular characteristics of TRα2, proposed mechanisms that regulate alternative splicing and indications pointing towards an antagonistic function of this TR isoform in vitro and in vivo. Moreover, remaining knowledge gaps and major challenges that complicate TRα2 characterization, as well as future strategies to fully uncover its physiological relevance, are discussed.
Collapse
Affiliation(s)
- Georg Sebastian Hönes
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
- Correspondence:
| | - Nina Härting
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (N.H.); (F.J.K.)
| | - Jens Mittag
- Institute for Endocrinology and Diabetes-Molecular Endocrinology, Center of Brain Behavior and Metabolism CBBM, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany;
| | - Frank J. Kaiser
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (N.H.); (F.J.K.)
| |
Collapse
|
7
|
3,5-T2-an Endogenous Thyroid Hormone Metabolite as Promising Lead Substance in Anti-Steatotic Drug Development? Metabolites 2022; 12:metabo12070582. [PMID: 35888706 PMCID: PMC9322486 DOI: 10.3390/metabo12070582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/10/2022] Open
Abstract
Thyroid hormones, their metabolites, and synthetic analogues are potential anti-steatotic drug candidates considering that subclinical and manifest hypothyroidism is associated with hepatic lipid accumulation, non-alcoholic fatty liver disease, and its pandemic sequelae. Thyromimetically active compounds stimulate hepatic lipogenesis, fatty acid beta-oxidation, cholesterol metabolism, and metabolic pathways of glucose homeostasis. Many of these effects are mediated by T3 receptor β1-dependent modulation of transcription. However, rapid non-canonical mitochondrial effects have also been reported, especially for the metabolite 3,5-diiodothyronine (3,5-T2), which does not elicit the full spectrum of “thyromimetic” actions inherent to T3. Most preclinical studies in rodent models of obesity and first human clinical trials are promising with respect to the antisteatotic hepatic effects, but potent agents exhibit unwanted thyromimetic effects on the heart and/or suppress feedback regulation of the hypothalamus-pituitary-thyroid-periphery axis and the fine-tuned thyroid hormone system. This narrative review focuses on 3,5-T2 effects on hepatic lipid and glucose metabolism and (non-)canonical mechanisms of action including its mitochondrial targets. Various high fat diet animal models with distinct thyroid hormone status indicate species- and dose-dependent efficiency of 3,5-T2 and its synthetic analogue TRC150094. No convincing evidence has been presented for their clinical use in the prevention or treatment of obesity and related metabolic conditions.
Collapse
|
8
|
Gaynullina DK, Sofronova SI, Selivanova EK, Shvetsova AA, Borzykh AA, Tarasova OS. Region-specific effects of antenatal/early postnatal hypothyroidism on endothelial NO-pathway activity in systemic circulation. Curr Res Physiol 2022; 5:8-15. [PMID: 34984343 PMCID: PMC8692882 DOI: 10.1016/j.crphys.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/01/2021] [Accepted: 12/09/2021] [Indexed: 11/17/2022] Open
Abstract
Background Antenatal/early postnatal hypothyroidism weakens NO-mediated anticontractile influence of endothelium in coronary arteries of adult rats, but it remains unclear whether this occurs in other vascular regions. We hypothesized that developmental thyroid deficiency is followed by region-specific changes in the endothelial NO-pathway activity in systemic vasculature. To explore this, we estimated the effects of antenatal/early postnatal hypothyroidism on NO-pathway activity and its potential local control mechanisms in rat mesenteric and skeletal muscle (sural) arteries. Methods Dams were treated with 6-propyl-2-thiouracil (PTU) in drinking water (0.0007%) during pregnancy and 2 weeks postpartum; control (CON) females received PTU-free water. Adult offspring (10–12-weeks) arteries were studied by wire myography, qPCR, and Western blotting. Results Endothelium removal or inhibition of NO-synthase with L-NNA augmented contractile responses to α1-adrenoceptor agonist methoxamine. In PTU compared to CON group, these effects were stronger in sural arteries, but did not differ in mesenteric arteries. The responses of both arteries to NO-donor DEA/NO were similar in CON and PTU rats. mRNA contents of deiodinase 2 and thyroid hormone receptor α were similar in mesenteric arteries of two groups but were elevated in sural arteries of PTU group compared to CON. The abundance of eNOS protein was higher in sural arteries of PTU compared to CON rats. Conclusion Antenatal/early postnatal hypothyroidism is followed by an increase in NO-mediated anticontractile influence in sural, but not in mesenteric arteries of adult animals. The diversity of hypothyroidism effects may be due to different alterations of local T3 synthesis/reception in different vascular beds. Antenatal hypothyroidism increases anticontractile NO-effect in sural arteries. Antenatal hypothyroidism doesn't change anticontractile effect in mesenteric arteries. Diverse hypothyroidism effects may be due to the differences in local T3 signaling.
Collapse
Affiliation(s)
- Dina K Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Svetlana I Sofronova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Ekaterina K Selivanova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Anastasia A Shvetsova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Anna A Borzykh
- Laboratory of Exercise Physiology, State Research Center of the Russian Federation-Institute for Biomedical Problems, Russian Academy of Sciences, 123007, Moscow, Russia
| | - Olga S Tarasova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia.,Laboratory of Exercise Physiology, State Research Center of the Russian Federation-Institute for Biomedical Problems, Russian Academy of Sciences, 123007, Moscow, Russia
| |
Collapse
|
9
|
Chen S, Wohlford GF, Vecchie’ A, Carbone S, Yavuz S, Van Tassell B, Abbate A, Celi FS. Acute Effects of Liothyronine Administration on Cardiovascular System and Energy Metabolism in Healthy Volunteers. Front Endocrinol (Lausanne) 2022; 13:843539. [PMID: 35295986 PMCID: PMC8919711 DOI: 10.3389/fendo.2022.843539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 02/02/2022] [Indexed: 11/26/2022] Open
Abstract
CONTEXT The pharmacokinetics of liothyronine causes concerns for cardiovascular toxicity. While the effects of sustained increase in serum T3 concentrations are well described, little is known on the effects of acute changes in T3 concentrations due to rapid action of thyroid hormone. OBJECTIVE To assess the clinical relevance of transient increase of T3 levels on cardiovascular system and energy metabolism. SETTING Double-blind, three arms, placebo controlled, cross-over study (ClinicalTrials.gov Identifier: NCT03098433). STUDY PARTICIPANTS Twelve volunteers (3 females, 9 males), age 27.7 ± 5.1 years. INTERVENTION Oral administration of liothyronine 0.7 mcg/kg, equimolar dose of levothyroxine (0.86 mcg/kg), or placebo in three identical study visits. Blood samples for total T3, free T4 were collected at times 0', 60' 120' 180' 240'. Continuous recording of heart rate, blood pressure, and hemodynamic data was performed using the volume clamp method. Resting energy expenditure was measured by indirect calorimetry. An echocardiogram was performed on each study visit at baseline and after the last blood sampling. MAIN OUTCOME MEASURES Changes in cardiovascular function and energy expenditure. RESULTS Following the administration of liothyronine, serum T3 reached a Cmax of 421 ± 57 ng/dL with an estimated Tmax of 120 ± 26 minutes. No differences between study arms were observed in heart rate, blood pressure, hemodynamics parameters, energy expenditure, and in echocardiogram parameters. CONCLUSIONS The absence of measurable rapid effects on the cardiovascular system following a high dose of liothyronine supports the rationale to perform long-term studies to assess its safety and effectiveness in patients affected by hypothyroidism.
Collapse
Affiliation(s)
- Shanshan Chen
- Division of Endocrinology Diabetes and Metabolism, Virginia Commonwealth University, Richmond, VA, United States
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, United States
| | - George F. Wohlford
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, VA, United States
| | - Alessandra Vecchie’
- Division of Cardiology, Virginia Commonwealth University, Richmond, VA, United States
- Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | - Salvatore Carbone
- Department of Kinesiology and Health Sciences, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA, United States
| | - Sahzene Yavuz
- Division of Endocrinology Diabetes and Metabolism, Virginia Commonwealth University, Richmond, VA, United States
| | - Benjamin Van Tassell
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University, Richmond, VA, United States
| | - Antonio Abbate
- Division of Cardiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Francesco S. Celi
- Division of Endocrinology Diabetes and Metabolism, Virginia Commonwealth University, Richmond, VA, United States
- *Correspondence: Francesco S. Celi,
| |
Collapse
|
10
|
Selivanova EK, Gaynullina DK, Tarasova OS. Thyroxine Induces Acute Relaxation of Rat Skeletal Muscle Arteries via Integrin αvβ3, ERK1/2 and Integrin-Linked Kinase. Front Physiol 2021; 12:726354. [PMID: 34594239 PMCID: PMC8477044 DOI: 10.3389/fphys.2021.726354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
Aim: Hyperthyroidism is associated with a decreased peripheral vascular resistance, which could be caused by the vasodilator genomic or non-genomic effects of thyroid hormones (TH). Non-genomic, or acute, effects develop within several minutes and involve a wide tissue-specific spectrum of molecular pathways poorly studied in vasculature. We aimed to investigate the mechanisms of acute effects of TH on rat skeletal muscle arteries. Methods: Sural arteries from male Wistar rats were used for isometric force recording (wire myography) and phosphorylated protein content measurement (Western blotting). Results: Both triiodothyronine (T3) and thyroxine (T4) reduced contractile response of sural arteries to α1-adrenoceptor agonist methoxamine. The effect of T4 was more prominent than T3 and not affected by iopanoic acid, an inhibitor of deiodinase 2. Endothelium denudation abolished the effect of T3, but not T4. Integrin αvβ3 inhibitor tetrac abolished the effect of T4 in endothelium-denuded arteries. T4 weakened methoxamine-induced elevation of phospho-MLC2 (Ser19) content in arterial samples. The effect of T4 in endothelium-denuded arteries was abolished by inhibiting ERK1/2 activation with U0126 as well as by ILK inhibitor Cpd22 but persisted in the presence of Src- or Rho-kinase inhibitors (PP2 and Y27632, respectively). Conclusion: Acute non-genomic relaxation of sural arteries induced by T3 is endothelium-dependent and that induced by T4 is endothelium-independent. The effect of T4 on α1-adrenergic contraction is stronger compared to T3 and involves the suppression of extracellular matrix signaling via integrin αvβ3, ERK1/2 and ILK with subsequent decrease of MLC2 (Ser19) phosphorylation.
Collapse
Affiliation(s)
- Ekaterina K Selivanova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dina K Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.,Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Olga S Tarasova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.,Laboratory of Exercise Physiology, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|