1
|
Martin JV, Sarkar PK. Nongenomic roles of thyroid hormones and their derivatives in adult brain: are these compounds putative neurotransmitters? Front Endocrinol (Lausanne) 2023; 14:1210540. [PMID: 37701902 PMCID: PMC10494427 DOI: 10.3389/fendo.2023.1210540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/02/2023] [Indexed: 09/14/2023] Open
Abstract
We review the evidence regarding the nongenomic (or non-canonical) actions of thyroid hormones (thyronines) and their derivatives (including thyronamines and thyroacetic acids) in the adult brain. The paper seeks to evaluate these compounds for consideration as candidate neurotransmitters. Neurotransmitters are defined by their (a) presence in the neural tissue, (b) release from neural tissue or cell, (c) binding to high-affinity and saturable recognition sites, (d) triggering of a specific effector mechanism and (e) inactivation mechanism. Thyronines and thyronamines are concentrated in brain tissue and show distinctive patterns of distribution within the brain. Nerve terminals accumulate a large amount of thyroid hormones in mature brain, suggesting a synaptic function. However, surprisingly little is known about the potential release of thyroid hormones at synapses. There are specific binding sites for thyroid hormones in nerve-terminal fractions (synaptosomes). A notable cell-membrane binding site for thyroid hormones is integrin αvβ3. Furthermore, thyronines bind specifically to other defined neurotransmitter receptors, including GABAergic, catecholaminergic, glutamatergic, serotonergic and cholinergic systems. Here, the thyronines tend to bind to sites other than the primary sites and have allosteric effects. Thyronamines also bind to specific membrane receptors, including the trace amine associated receptors (TAARs), especially TAAR1. The thyronines and thyronamines activate specific effector mechanisms that are short in latency and often occur in subcellular fractions lacking nuclei, suggesting nongenomic actions. Some of the effector mechanisms for thyronines include effects on protein phosphorylation, Na+/K+ ATPase, and behavioral measures such as sleep regulation and measures of memory retention. Thyronamines promptly regulate body temperature. Lastly, there are numerous inactivation mechanisms for the hormones, including decarboxylation, deiodination, oxidative deamination, glucuronidation, sulfation and acetylation. Therefore, at the current state of the research field, thyroid hormones and their derivatives satisfy most, but not all, of the criteria for definition as neurotransmitters.
Collapse
Affiliation(s)
- Joseph V. Martin
- Biology Department, Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, United States
| | - Pradip K. Sarkar
- Department of Basic Sciences, Parker University, Dallas, TX, United States
| |
Collapse
|
2
|
Petito G, Cioffi F, Magnacca N, de Lange P, Senese R, Lanni A. Adipose Tissue Remodeling in Obesity: An Overview of the Actions of Thyroid Hormones and Their Derivatives. Pharmaceuticals (Basel) 2023; 16:ph16040572. [PMID: 37111329 PMCID: PMC10146771 DOI: 10.3390/ph16040572] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Metabolic syndrome and obesity have become important health issues of epidemic proportions and are often the cause of related pathologies such as type 2 diabetes (T2DM), hypertension, and cardiovascular disease. Adipose tissues (ATs) are dynamic tissues that play crucial physiological roles in maintaining health and homeostasis. An ample body of evidence indicates that in some pathophysiological conditions, the aberrant remodeling of adipose tissue may provoke dysregulation in the production of various adipocytokines and metabolites, thus leading to disorders in metabolic organs. Thyroid hormones (THs) and some of their derivatives, such as 3,5-diiodo-l-thyronine (T2), exert numerous functions in a variety of tissues, including adipose tissues. It is known that they can improve serum lipid profiles and reduce fat accumulation. The thyroid hormone acts on the brown and/or white adipose tissues to induce uncoupled respiration through the induction of the uncoupling protein 1 (UCP1) to generate heat. Multitudinous investigations suggest that 3,3',5-triiodothyronine (T3) induces the recruitment of brown adipocytes in white adipose depots, causing the activation of a process known as "browning". Moreover, in vivo studies on adipose tissues show that T2, in addition to activating brown adipose tissue (BAT) thermogenesis, may further promote the browning of white adipose tissue (WAT), and affect adipocyte morphology, tissue vascularization, and the adipose inflammatory state in rats receiving a high-fat diet (HFD). In this review, we summarize the mechanism by which THs and thyroid hormone derivatives mediate adipose tissue activity and remodeling, thus providing noteworthy perspectives on their efficacy as therapeutic agents to counteract such morbidities as obesity, hypercholesterolemia, hypertriglyceridemia, and insulin resistance.
Collapse
Affiliation(s)
- Giuseppe Petito
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", 81100 Caserta, Italy
| | - Federica Cioffi
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Nunzia Magnacca
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", 81100 Caserta, Italy
| | - Pieter de Lange
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", 81100 Caserta, Italy
| | - Rosalba Senese
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", 81100 Caserta, Italy
| | - Antonia Lanni
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", 81100 Caserta, Italy
| |
Collapse
|
3
|
Spencer CA. Laboratory Thyroid Tests: A Historical Perspective. Thyroid 2023; 33:407-419. [PMID: 37037032 DOI: 10.1089/thy.2022.0397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Background: This review presents a timeline showing how technical advances made over the last seven decades have impacted the development of laboratory thyroid tests. Summary: Thyroid tests have evolved from time-consuming manual procedures using isotopically labeled iodine as signals (131I and later 125I) performed in nuclear medicine laboratories, to automated nonisotopic tests performed on multianalyte instruments in routine clinical chemistry laboratories. The development of isotopic radioimmunoassay techniques around 1960, followed by the advent of monoclonal antibody technology in the mid-1970s, led to the development of a nonisotopic immunometric assay methodology that forms the backbone of present-day thyroid testing. This review discusses the development of methods for measuring total thyroxine and triiodothyronine, direct and indirect free thyroid hormone measurements and estimates (free thyroxine and free triiodothyronine), thyrotropin (TSH), thyroid autoantibodies (thyroperoxidase, thyroglobulin [Tg] and TSH receptor autoantibodies), and Tg protein. Despite progressive improvements made in sensitivity and specificity, current thyroid tests remain limited by between-method differences in the numeric values they report, as well as nonspecific interferences with test reagents and interferences from analyte autoantibodies. Conclusions: Thyroid disease affects ∼10% of the U.S. population and is mostly managed on an outpatient basis, generating 60% of endocrine laboratory tests. In future, it is hoped that interferences will be eliminated, and the standardization/harmonization of tests will facilitate the establishment of universal test reference ranges.
Collapse
Affiliation(s)
- Carole Ann Spencer
- Department of Endocrinology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
4
|
Exogenous 3-Iodothyronamine (T1AM) Can Affect Phosphorylation of Proteins Involved on Signal Transduction Pathways in In Vitro Models of Brain Cell Lines, but These Effects Are Not Strengthened by Its Catabolite, 3-Iodothyroacetic Acid (TA1). Life (Basel) 2022; 12:life12091352. [PMID: 36143389 PMCID: PMC9502970 DOI: 10.3390/life12091352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 12/02/2022] Open
Abstract
T1AM, a derivative of thyroid hormones, and its major catabolite, TA1, produce effects on memory acquisition in rodents. In the present study, we compared the effects of exogenous T1AM and TA1 on protein belonging to signal transduction pathways, assuming that TA1 may strengthen T1AM’s effects in brain tissue. A hybrid line of cancer cells of mouse neuroblastoma and rat glioma (NG 108-15), as well as a human glioblastoma cell line (U-87 MG) were used. We first characterized the in vitro model by analyzing gene expression of proteins involved in the glutamatergic cascade and cellular uptake of T1AM and TA1. Then, cell viability, glucose consumption, and protein expression were assessed. Both cell lines expressed receptors implicated in glutamatergic pathway, namely Nmdar1, Glur2, and EphB2, but only U-87 MG cells expressed TAAR1. At pharmacological concentrations, T1AM was taken up and catabolized to TA1 and resulted in more cytotoxicity compared to TA1. The major effect, highlighted in both cell lines, albeit on different proteins involved in the glutamatergic signaling, was an increase in phosphorylation, exerted by T1AM but not reproduced by TA1. These findings indicate that, in our in vitro models, T1AM can affect proteins involved in the glutamatergic and other signaling pathways, but these effects are not strengthened by TA1.
Collapse
|
5
|
Martínez Brito D, Botrè F, Romanelli F, de la Torre X. Thyroid metabolism and supplementation. A review framed in sports environment. Drug Test Anal 2022; 14:1176-1186. [PMID: 35315230 DOI: 10.1002/dta.3257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVES This paper aimed to consider those features that may suggest a link between thyroid hormones pharmacology and athletes' health based on current consumption trends in a population of athletes. METHODS Methods used were observation, description, and synthesis, mainly. Among the documents reviewed were: books, scientific articles, and review articles peer-reviewed. The review covered sources published in the period 1961 to 2021. Only references with a traceable origin were accepted (DOI numbering, ISSN and ISBN, as well as peer-reviewed journals). The data on the consumption of thyroid hormones derivatives were extracted from the Doping Control Forms of athlete samples received at Laboratorio Antidoping FMSI of Rome from 2017 to 2021. RESULTS An overview of the biosynthesis, pharmacology, and metabolism of thyroid hormones, including thyronamines and thyronacetic acids, was presented. Likewise, a summary is presented on the relationship between thyroid hormones and ethnic and gender differences, their physiology in sport, and the reasons why their use could be considered attractive for athletes. CONCLUSION Today, thyroid hormones are not listed as a prohibited substance by the World Anti-Doping Agency. However, several requests to include levothyroxine on the prohibited list are documented. The observation that the number of athletes taking thyroid hormones is growing, particularly in sports such as cycling, triathlons, and skating, should prompt an update on this topic.
Collapse
Affiliation(s)
| | - Francesco Botrè
- Laboratorio Antidoping FMSI, Federazione Medico Sportiva Italiana, Rome, Italy.,REDs - Research and Expertise on Anti-Doping Sciences, Institute of Sport Science, University of Lausanne, Lausanne, Switzerland
| | - Francesco Romanelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Roma, Italy
| | - Xavier de la Torre
- Laboratorio Antidoping FMSI, Federazione Medico Sportiva Italiana, Rome, Italy
| |
Collapse
|
6
|
Gellrich L, Heitel P, Heering J, Kilu W, Pollinger J, Goebel T, Kahnt A, Arifi S, Pogoda W, Paulke A, Steinhilber D, Proschak E, Wurglics M, Schubert-Zsilavecz M, Chaikuad A, Knapp S, Bischoff I, Fürst R, Merk D. l-Thyroxin and the Nonclassical Thyroid Hormone TETRAC Are Potent Activators of PPARγ. J Med Chem 2020; 63:6727-6740. [DOI: 10.1021/acs.jmedchem.9b02150] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Leonie Gellrich
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Pascal Heitel
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Jan Heering
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Whitney Kilu
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Julius Pollinger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Tamara Goebel
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Astrid Kahnt
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Silvia Arifi
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Werner Pogoda
- Department of Forensic Toxicology, Institute of Forensic Medicine, Goethe University Frankfurt, Kennedyallee 104, D-60596 Frankfurt, Germany
| | - Alexander Paulke
- Department of Forensic Toxicology, Institute of Forensic Medicine, Goethe University Frankfurt, Kennedyallee 104, D-60596 Frankfurt, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Mario Wurglics
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Manfred Schubert-Zsilavecz
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Apirat Chaikuad
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, D-60438 Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, D-60438 Frankfurt, Germany
| | - Iris Bischoff
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| |
Collapse
|
7
|
Senese R, Cioffi F, Petito G, Goglia F, Lanni A. Thyroid hormone metabolites and analogues. Endocrine 2019; 66:105-114. [PMID: 31359245 DOI: 10.1007/s12020-019-02025-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/18/2019] [Indexed: 12/17/2022]
Abstract
Several metabolic products that derive from L-thyroxine (T4) and 3,3'5-L-triiodothyronine (T3), the main thyroid hormones secreted by the thyroid gland, possess biologic activities. Among these metabolites or derivatives showing physiological actions some have received greater attention: diiodothyronines, iodothyronamines, acetic acid analogues. It is known that increased thyroid hormone (T3 and T4) levels can improve serum lipid profiles and reduce body fat. These positive effects are, however, counterbalanced by adverse effects on the heart, muscle and bone, limiting their use. In addition to the naturally occurring metabolites, thyroid hormone analogues have been developed that either have selective effects on specific tissues or bind selectively to thyroid hormone receptor (TR) isoform. Among these GC-1, KB141, KB2115, and DITPA were deeply investigated and displayed promising therapeutic results in the potential treatment of conditions such as dyslipidemias and obesity. In this review, we summarize the current knowledge of metabolites and analogues of T4 and T3 with reference to their possible clinical application in the treatment of human diseases.
Collapse
Affiliation(s)
- Rosalba Senese
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Federica Cioffi
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Giuseppe Petito
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy
| | - Fernando Goglia
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Antonia Lanni
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "L. Vanvitelli", Caserta, Italy.
| |
Collapse
|
8
|
Köhrle J, Biebermann H. 3-Iodothyronamine-A Thyroid Hormone Metabolite With Distinct Target Profiles and Mode of Action. Endocr Rev 2019; 40:602-630. [PMID: 30649231 DOI: 10.1210/er.2018-00182] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022]
Abstract
The rediscovery of the group of thyronamines (TAMs), especially the first detailed description of their most prominent congener 3-iodothyronamine (3T1AM) 14 years ago, boosted research on this thyroid hormone metabolite tremendously. TAMs exert actions partly opposite to and distinct from known functions of thyroid hormones. These fascinating metabolic, anapyrexic, cytoprotective, and brain effects quickly evoked the hope to use hormone-derived TAMs as a therapeutic option. The G protein-coupled receptor (GPCR) TAAR1, a member of the trace amine-associated receptor (TAAR) family, was identified as the first target and effector of TAM action. The initial enthusiasm on pharmacological actions of exogenous TAMs elicited many questions, such as sites of biosynthesis, analytics, modes of action, inactivation, and role of TAMs in (patho)physiology. Meanwhile, it became clear that TAMs not only interact with TAAR1 or other TAAR family members but also with several aminergic receptors and non-GPCR targets such as transient receptor potential channels, mitochondrial proteins, and the serum TAM-binding protein apolipoprotein B100, thus classifying 3T1AM as a multitarget ligand. The physiological mode of action of TAMs is still controversial because regulation of endogenous TAM production and the sites of its biosynthesis are not fully elucidated. Methods for 3T1AM analytics need further validation, as they revealed different blood and tissue concentrations depending on detection principles used such as monoclonal antibody-based immunoassay vs liquid chromatography- matrix-assisted laser desorption/ionization mass spectrometry or time-of-flight mass spectrometry. In this review, we comprehensively summarize and critically evaluate current basic, translational, and clinical knowledge on 3T1AM and its main metabolite 3-iodothyroacetic acid, focusing on endocrine-relevant aspects and open but highly challenging issues.
Collapse
Affiliation(s)
- Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Heike Biebermann
- Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
9
|
Abstract
Trace amines are endogenous compounds classically regarded as comprising β-phenylethyalmine, p-tyramine, tryptamine, p-octopamine, and some of their metabolites. They are also abundant in common foodstuffs and can be produced and degraded by the constitutive microbiota. The ability to use trace amines has arisen at least twice during evolution, with distinct receptor families present in invertebrates and vertebrates. The term "trace amine" was coined to reflect the low tissue levels in mammals; however, invertebrates have relatively high levels where they function like mammalian adrenergic systems, involved in "fight-or-flight" responses. Vertebrates express a family of receptors termed trace amine-associated receptors (TAARs). Humans possess six functional isoforms (TAAR1, TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9), whereas some fish species express over 100. With the exception of TAAR1, TAARs are expressed in olfactory epithelium neurons, where they detect diverse ethological signals including predators, spoiled food, migratory cues, and pheromones. Outside the olfactory system, TAAR1 is the most thoroughly studied and has both central and peripheral roles. In the brain, TAAR1 acts as a rheostat of dopaminergic, glutamatergic, and serotonergic neurotransmission and has been identified as a novel therapeutic target for schizophrenia, depression, and addiction. In the periphery, TAAR1 regulates nutrient-induced hormone secretion, suggesting its potential as a novel therapeutic target for diabetes and obesity. TAAR1 may also regulate immune responses by regulating leukocyte differentiation and activation. This article provides a comprehensive review of the current state of knowledge of the evolution, physiologic functions, pharmacology, molecular mechanisms, and therapeutic potential of trace amines and their receptors in vertebrates and invertebrates.
Collapse
Affiliation(s)
- Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); Skolkovo Institute of Science and Technology (Skoltech), Moscow, Russia (R.R.G.); Neuroscience, Ophthalmology, and Rare Diseases Discovery and Translational Area, pRED, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (M.C.H.); and Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada (M.D.B.)
| | - Marius C Hoener
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); Skolkovo Institute of Science and Technology (Skoltech), Moscow, Russia (R.R.G.); Neuroscience, Ophthalmology, and Rare Diseases Discovery and Translational Area, pRED, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (M.C.H.); and Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada (M.D.B.)
| | - Mark D Berry
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); Skolkovo Institute of Science and Technology (Skoltech), Moscow, Russia (R.R.G.); Neuroscience, Ophthalmology, and Rare Diseases Discovery and Translational Area, pRED, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (M.C.H.); and Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada (M.D.B.)
| |
Collapse
|
10
|
Lehmphul I, Hoefig CS, Köhrle J. 3-Iodothyronamine reduces insulin secretion in vitro via a mitochondrial mechanism. Mol Cell Endocrinol 2018; 460:219-228. [PMID: 28754352 DOI: 10.1016/j.mce.2017.07.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/17/2017] [Accepted: 07/24/2017] [Indexed: 12/29/2022]
Abstract
PURPOSE 3-iodothyronamine (3-T1AM), a decarboxylated and deiodinated thyroid hormone metabolite, leads at pharmacological doses to hypoinsulinemia, hyperglucagonemia and hyperglycemia in vivo. As the pancreatic Langerhans islets express thyroid hormone transmembrane transporters (THTT), we tested the hypothesis that not only plasma membrane-mediated 3-T1AM binding to and activation of G-protein coupled receptors, but also 3-T1AM metabolite(s) generated by 3-T1AM uptake and metabolism might decrease glucose-stimulated insulin secretion (GSIS). METHODS Murine pancreatic β-cells MIN6 were characterized for gene expression of THTT, deiodinases and monoamine oxidases. 3-T1AM uptake and intracellular metabolism to the corresponding 3-iodothyroacetic acid were analysed by liquid-chromatography tandem mass spectrometry (LC-MS/MS) at different time points in cells as well as the conditioned medium. Mitochondrial activity, especially ATP-production, was monitored real-time after 3-T1AM application using Seahorse Bioanalyzer technique. Effect of 3-T1AM on GSIS into the culture medium was assayed by ELISA. RESULTS MIN6 cells express classical THTT, proposed to transport 3-T1AM, as well as 3-T1AM metabolizing enzymes comparable to murine primary pancreatic islets. 3-T1AM accumulates in MIN6 cells and is metabolized by intracellular MaoB to 3-iodothyroacetic, which in turn is rapidly exported. 3-T1AM decreases mitochondrial ATP-production concentration dependently. GSIS is diminished by 3-T1AM treatment. Using LC-MS/MS, no further 3-T1AM metabolites except 3-iodothyroacetic were detectable. CONCLUSIONS This data provides a first link between cellular 3-T1AM uptake and regulation of mitochondrial energy metabolism in ß-cells, resulting in reduced insulin secretion. We conclude that MIN6 is an appropriate cell model to study 3-T1AM-dependent (intra-)cellular biochemical mechanisms affecting insulin production in vitro.
Collapse
Affiliation(s)
- Ina Lehmphul
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institut für Experimentelle Endokrinologie, 13353 Berlin, Germany
| | - Carolin S Hoefig
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institut für Experimentelle Endokrinologie, 13353 Berlin, Germany
| | - Josef Köhrle
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Institut für Experimentelle Endokrinologie, 13353 Berlin, Germany.
| |
Collapse
|
11
|
Abstract
More than a century after the discovery of L-Thyroxine, the main thyroid hormone secreted solely by the thyroid gland, several metabolites of this iodinated, tyrosine-derived ancestral hormone have been identified. These are utilized as hormones during development, differentiation, metamorphosis, and regulation of most biochemical reactions in vertebrates and their precursor species. Among those metabolites are the thyromimetically active 3,3',5-Triiodo-L-thyronine (T3) and 3,5-Diiodo-L-thronine, reverse-T3 (3,3',5'-Triiodo-L-thyronine) with still unclear function, the recently re-discovered thyronamines (e.g., 3-Iodo-thyronamine), which exert in part T3-antagonistic functions, the thyroacetic acids (e.g., Tetrac and Triac), as well as various sulfated or glucuronidated metabolites of this panel of iodinated signaling compounds. In the blood most of these hydrophobic metabolites are tightly bound to the serum distributor proteins thyroxine binding globulin (TBG), transthyretin (TTR), albumin or apolipoprotein B100. Cellular import and export of these charged, highly hydrophobic amino acid derivatives requires a number of cell-membrane transporters or facilitators such as MCT8 or MCT10 and members of the OATP and LAT families of transporters. Depending on their structure, the thyroid hormone metabolites exert their cellular action by binding and thus modulating the function of various receptors systems (e.g., ανβ3 integrin receptor and transient receptor potential channels (TRPM8) of the cell membrane), in part linked to intracellular downstream kinase signaling cascades, and several isoforms of membrane-associated, mitochondrial or nuclear thyroid hormone receptors (TR), which are members of the c-erbA family of ligand-modulated transcription factors. Intracellular deiodinase selenoenzymes, which obligatory are membrane integrated enzymes, ornithine decarboxylase and monoamine oxidases control local availability of biologically active thyroid hormone metabolites. Inactivation of thyroid hormone metabolites occurs mainly by deiodination, sulfation or glucuronidation, reactions which favor their renal or fecal elimination.
Collapse
Affiliation(s)
- Josef Köhrle
- Charité-Universitätsmedizin Berlin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zuBerlin, and Berlin Institute of Health, Institut für Experimentelle Endokrinologie, Berlin, Germany.
| |
Collapse
|
12
|
Groeneweg S, Peeters RP, Visser TJ, Visser WE. Therapeutic applications of thyroid hormone analogues in resistance to thyroid hormone (RTH) syndromes. Mol Cell Endocrinol 2017; 458:82-90. [PMID: 28235578 DOI: 10.1016/j.mce.2017.02.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/17/2017] [Accepted: 02/18/2017] [Indexed: 10/20/2022]
Abstract
Thyroid hormone (TH) is crucial for normal development and metabolism of virtually all tissues. TH signaling is predominantly mediated through binding of the bioactive hormone 3,3',5-triiodothyronine (T3) to the nuclear T3-receptors (TRs). The intracellular TH levels are importantly regulated by transporter proteins that facilitate the transport of TH across the cell membrane and by the three deiodinating enzymes. Defects at the level of the TRs, deiodinases and transporter proteins result in resistance to thyroid hormone (RTH) syndromes. Compounds with thyromimetic potency but with different (bio)chemical properties compared to T3 may hold therapeutic potential in these syndromes by bypassing defective transporters or binding to mutant TRs. Such TH analogues have the potential to rescue TH signaling. This review describes the role of TH analogues in the treatment of RTH syndromes. In particular, the application of 3,3',5-triiodothyroacetic acid (Triac) in RTH due to defective TRβ and the role of 3,5-diiodothyropropionic acid (DITPA), 3,3',5,5'-tetraiodothyroacetic acid (Tetrac) and Triac in MCT8 deficiency will be highlighted.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Theo J Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - W Edward Visser
- Department of Internal Medicine, Academic Center for Thyroid Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
13
|
Bellusci L, Laurino A, Sabatini M, Sestito S, Lenzi P, Raimondi L, Rapposelli S, Biagioni F, Fornai F, Salvetti A, Rossi L, Zucchi R, Chiellini G. New Insights into the Potential Roles of 3-Iodothyronamine (T1AM) and Newly Developed Thyronamine-Like TAAR1 Agonists in Neuroprotection. Front Pharmacol 2017; 8:905. [PMID: 29311919 PMCID: PMC5732922 DOI: 10.3389/fphar.2017.00905] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/28/2017] [Indexed: 12/30/2022] Open
Abstract
3-Iodothyronamine (T1AM) is an endogenous high-affinity ligand of the trace amine-associated receptor 1 (TAAR1), detected in mammals in many organs, including the brain. Recent evidence indicates that pharmacological TAAR1 activation may offer a novel therapeutic option for the treatment of a wide range of neuropsychiatric and metabolic disorders. To assess potential neuroprotection by TAAR1 agonists, in the present work, we initially investigated whether T1AM and its corresponding 3-methylbiaryl-methane analog SG-2 can improve learning and memory when systemically administered to mice at submicromolar doses, and whether these effects are modified under conditions of MAO inhibition by clorgyline. Our results revealed that when i.p. injected to mice, both T1AM and SG-2 produced memory-enhancing and hyperalgesic effects, while increasing ERK1/2 phosphorylation and expression of transcription factor c-fos. Notably, both compounds appeared to rely on the action of ubiquitous enzymes MAO to produce the corresponding oxidative metabolites that were then able to activate the histaminergic system. Since autophagy is key for neuronal plasticity, in a second line of experiments we explored whether T1AM and synthetic TAAR1 agonists SG1 and SG2 were able to induce autophagy in human glioblastoma cell lines (U-87MG). After treatment of U-87MG cells with 1 μM T1AM, SG-1, SG-2 transmission electron microscopy (TEM) and immunofluorescence (IF) showed a significant time-dependent increase of autophagy vacuoles and microtubule-associated protein 1 light chain 3 (LC3). Consistently, Western blot analysis revealed a significant increase of the LC3II/LC3I ratio, with T1AM and SG-1 being the most effective agents. A decreased level of the p62 protein was also observed after treatment with T1AM and SG-1, which confirms the efficacy of these compounds as autophagy inducers in U-87MG cells. In the process to dissect which pathway induces ATG, the effects of these compounds were evaluated on the PI3K-AKT-mTOR pathway. We found that 1 μM T1AM, SG-1 and SG-2 decreased pAKT/AKT ratio at 0.5 and 4 h after treatment, suggesting that autophagy is induced by inhibiting mTOR phosphorylation by PI3K-AKT-mTOR pathway. In conclusion, our study shows that T1AM and thyronamine-like derivatives SG-1 and SG-2 might represent valuable tools to therapeutically intervene with neurological disorders.
Collapse
Affiliation(s)
- Lorenza Bellusci
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Pisa, Italy
| | - Annunziatina Laurino
- Section of Pharmacology and Toxicology, Department of Psychology, Neurology, Drug Sciences, Health of the Child, Pharmacology, University of Florence, Florence, Italy
| | - Martina Sabatini
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Pisa, Italy
| | - Simona Sestito
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Paola Lenzi
- Unit of Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Laura Raimondi
- Section of Pharmacology and Toxicology, Department of Psychology, Neurology, Drug Sciences, Health of the Child, Pharmacology, University of Florence, Florence, Italy
| | - Simona Rapposelli
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Francesco Fornai
- Unit of Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Alessandra Salvetti
- Unit of Experimental Biology and Genetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Leonardo Rossi
- Unit of Experimental Biology and Genetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Riccardo Zucchi
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Pisa, Italy
| | - Grazia Chiellini
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Pisa, Italy
| |
Collapse
|
14
|
Lorenzini L, Ghelardoni S, Saba A, Sacripanti G, Chiellini G, Zucchi R. Recovery of 3-Iodothyronamine and Derivatives in Biological Matrixes: Problems and Pitfalls. Thyroid 2017; 27:1323-1331. [PMID: 28859548 DOI: 10.1089/thy.2017.0111] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Difficulties have been reported in quantitating 3-iodothyronamine (T1AM) in blood or serum, and tentatively attributed to problems in extraction or other pre-analytical steps. For this reason, even cell culture experiments have often be performed with unphysiological protein-free media. The aim of this study was to evaluate the recovery of exogenous T1AM added to a standard cell culture medium, namely Dulbecco's minimum essential medium (DMEM) supplemented with fetal bovine serum (FBS), and to other biological matrixes. METHODS Cell culture media (Krebs-Ringer buffer, DMEM, FBS, DMEM + FBS, used either in the absence or in the presence of NG108-15 cells) and other biological matrixes (rat brain and liver homogenates, human plasma, and blood) were spiked with T1AM and/or deuterated T1AM (d4-T1AM) and incubated for times ranging from 0 to 240 minutes. Samples were then extracted using a liquid/liquid method and analyzed using liquid chromatography coupled to mass spectrometry in order to assay T1AM and its metabolites, namely 3-iodothyroacetic acid (TA1), thyronamine, thyroacetic acid, N-acetyl-T1AM, and T1AM esters. RESULTS In FBS-containing buffers, T1AM decreased exponentially over time, with a half-life of 6-17 minutes, depending on FBS content, and after 60 minutes, it averaged 0-10% of the baseline. T1AM metabolites were not detected, except for minimum amounts of TA1. Notably, d4-T1AM decreased over time at a much lower rate, reaching 50-70% of the baseline at 60 minutes. These effects were completely abolished by protein denaturation and partly reduced by semicarbazide. In the presence of cells, T1AM concentration decreased virtually to 0 within 60 minutes, but TA1 accumulated in the incubation medium, with quantitative recovery. Spontaneous decrease in T1AM concentration with isotopic difference was confirmed in rat organ homogenates and human blood. CONCLUSIONS These results suggest binding and sequestration of T1AM and/or its aldehyde derivative by blood and tissue proteins, with significant isotope effects. These issues might account for the technical problems complicating the analytical assays of endogenous T1AM.
Collapse
|
15
|
Groeneweg S, Peeters RP, Visser TJ, Visser WE. Triiodothyroacetic acid in health and disease. J Endocrinol 2017; 234:R99-R121. [PMID: 28576869 DOI: 10.1530/joe-17-0113] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 12/16/2022]
Abstract
Thyroid hormone (TH) is crucial for development and metabolism of many tissues. The physiological relevance and therapeutic potential of TH analogs have gained attention in the field for many years. In particular, the relevance and use of 3,3',5-triiodothyroacetic acid (Triac, TA3) has been explored over the last decades. Although TA3 closely resembles the bioactive hormone T3, differences in transmembrane transport and receptor isoform-specific transcriptional activation potency exist. For these reasons, the application of TA3 as a treatment for resistance to TH (RTH) syndromes, especially MCT8 deficiency, is topic of ongoing research. This review is a summary of all currently available literature about the formation, metabolism, action and therapeutic applications of TA3.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine and Academic Center for Thyroid DiseasesErasmus University Medical Center, Rotterdam, The Netherlands
| | - Robin P Peeters
- Department of Internal Medicine and Academic Center for Thyroid DiseasesErasmus University Medical Center, Rotterdam, The Netherlands
| | - Theo J Visser
- Department of Internal Medicine and Academic Center for Thyroid DiseasesErasmus University Medical Center, Rotterdam, The Netherlands
| | - W Edward Visser
- Department of Internal Medicine and Academic Center for Thyroid DiseasesErasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
16
|
Fernando R, Placzek E, Reese EA, Placzek AT, Schwartz S, Trierweiler A, Niziol LM, Raychaudhuri N, Atkins S, Scanlan TS, Smith TJ. Elevated Serum Tetrac in Graves Disease: Potential Pathogenic Role in Thyroid-Associated Ophthalmopathy. J Clin Endocrinol Metab 2017; 102:776-785. [PMID: 27768856 PMCID: PMC5460682 DOI: 10.1210/jc.2016-2762] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/18/2016] [Indexed: 11/19/2022]
Abstract
CONTEXT The sources and biological impact of 3,3',5,5' tetraiodothyroacetic acid (TA4) are uncertain. CD34+ fibrocytes express several proteins involved in the production of thyroid hormones. They infiltrate the orbit in Graves disease (GD), an autoimmune process known as thyroid-associated ophthalmopathy. It appears that the thyrotropin receptor plays an important role in the pathogenesis of thyroid-associated ophthalmopathy. OBJECTIVE To quantify levels of TA4 in healthy participants and those with GD, determine whether fibrocytes generate this thyroid hormone analogue, and determine whether TA4 influences the actions of thyroid-stimulating hormone and thyroid-stimulating immunoglobulins in orbital fibroblasts. DESIGN/SETTING/PARTICIPANTS Patients with GD and healthy donors in an academic medical center clinical practice were recruited. MAIN OUTCOME MEASURES Liquid chromatography-tandem mass spectrometry, autoradiography, real-time polymerase chain reaction, hyaluronan immunoassay. RESULTS Serum levels of TA4 are elevated in GD. TA4 levels are positively correlated with those of thyroxine and negatively correlated with serum levels of triiodothyronine. Several cell types in culture generate TA4 from ambient thyroxine, including fibrocytes, HELA cells, human Müller stem cells, and retinal pigmented epithelial cells. Propylthiouracil inhibits TA4 generation. TA4 enhances the induction by thyrotropin and thyroid-stimulating immunoglobulins of several participants in the pathogenesis of thyroid-associated ophthalmopathy, including interleukin 6, hyaluronan synthase 1, prostaglandin endoperoxide H synthase 2, and haluronan production. CONCLUSION TA4 may be ubiquitously generated in many tissues and enhances the biological impact of thyrotropin and thyroid-stimulating immunoglobulins in orbital connective tissue. These findings may identify a physiologically important determinant of extrathyroidal thyroid-stimulating hormone action.
Collapse
Affiliation(s)
| | - Ekaterina Placzek
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon 97239
| | - Edmund A Reese
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon 97239
| | - Andrew T Placzek
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon 97239
| | | | | | | | | | | | - Thomas S Scanlan
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon 97239
| | - Terry J Smith
- Department of Ophthalmology and Visual Sciences and
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105; and
| |
Collapse
|
17
|
Schanze N, Jacobi SF, Rijntjes E, Mergler S, Del Olmo M, Hoefig CS, Khajavi N, Lehmphul I, Biebermann H, Mittag J, Köhrle J. 3-Iodothyronamine Decreases Expression of Genes Involved in Iodide Metabolism in Mouse Thyroids and Inhibits Iodide Uptake in PCCL3 Thyrocytes. Thyroid 2017; 27:11-22. [PMID: 27788620 DOI: 10.1089/thy.2016.0182] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND 3-Iodothyronamine (3-T1AM) is an endogenous decarboxylated thyroid hormone (TH) metabolite. Pharmacological doses of 3-T1AM decrease heart rate, body temperature, and metabolic rate in rodents-effects that are contrary to classic TH excess. Furthermore, a single dose of 3-T1AM was shown to suppress the hypothalamic-pituitary-thyroid (HPT) axis in rats. It was hypothesized that 3-T1AM might play a role in the fine-tuning of TH action and might have a direct regulatory effect on the thyroid gland. METHODS This study tested whether repeated 3-T1AM treatment interfered with thyroid function and the HPT axis in mice. Therefore, male C57BL/6 mice were intraperitoneally injected with 5 mg/kg of 3-T1AM or vehicle daily for seven days. Additionally, the effects of 3-T1AM on the differentiated rat thyrocyte cell line PCCL3 were analyzed. RESULTS Repeated administration of 3-T1AM decreased thyroidal mRNA content of the sodium iodide symporter (Nis), thyroglobulin, and pendrin in mice. No interference with the HPT axis was observed, as determined by unaltered pituitary mRNA levels of triiodothyronine-responsive genes, including thyrotropin subunit β. Furthermore, 3-T1AM treatment did not change transcript levels of hepatic triiodothyronine-responsive genes, such as deiodinase 1. In line with this, serum TH concentrations were not changed after the treatment period of seven days. In concordance with the in vivo findings, 3-T1AM decreased the thyrotropin-dependent expression of Nis and functional iodide uptake in PCCL3 cells in vitro. Additionally, uptake and metabolism of 3-T1AM by PCCL3 cells was observed, as well as 3-T1AM-dependent changes in intracellular Ca2+ concentration that might be involved in mediating the reported effects. CONCLUSIONS In conclusion, 3-T1AM application decreased expression of selected TH synthesis genes by acting directly on the thyroid gland, and it might therefore affect TH synthesis without involvement of the HPT axis.
Collapse
Affiliation(s)
- Nancy Schanze
- 1 Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
- 2 Department of Cell and Molecular Biology, Karolinska Institutet , Stockholm, Sweden
| | - Simon Friedrich Jacobi
- 2 Department of Cell and Molecular Biology, Karolinska Institutet , Stockholm, Sweden
- 3 Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Eddy Rijntjes
- 1 Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Stefan Mergler
- 4 Experimentelle Ophthalmologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Marta Del Olmo
- 1 Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Carolin Stephanie Hoefig
- 1 Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
- 2 Department of Cell and Molecular Biology, Karolinska Institutet , Stockholm, Sweden
| | - Noushafarin Khajavi
- 3 Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Ina Lehmphul
- 1 Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Heike Biebermann
- 3 Institut für Experimentelle Pädiatrische Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Jens Mittag
- 2 Department of Cell and Molecular Biology, Karolinska Institutet , Stockholm, Sweden
- 5 Molecular Endocrinology, Universitätsklinikum Schleswig-Holstein , Medizinische Klinik I/CBBM, Lübeck, Germany
| | - Josef Köhrle
- 1 Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| |
Collapse
|
18
|
Glossmann HH, Lutz OMD. Torpor: The Rise and Fall of 3-Monoiodothyronamine from Brain to Gut-From Gut to Brain? Front Endocrinol (Lausanne) 2017; 8:118. [PMID: 28620354 PMCID: PMC5450037 DOI: 10.3389/fendo.2017.00118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022] Open
Abstract
3-Monoiodothyronamine (T1AM), first isolated from rat brain, is reported to be an endogenous, rapidly acting metabolite of thyroxine. One of its numerous effects is the induction of a "torpor-like" state in experimental animals. A critical analysis of T1AM, to serve as an endogenous cryogen, is given. The proposed biosynthetic pathway for formation of T1AM, which includes deiodinases and ornithine decarboxylase in the upper intestinum, is an unusual one. To reach the brain via systemic circulation, enterohepatic recycling and passage through the liver may occur. The possible role of gut microbiota is discussed. T1AM concentrations in human serum, measured by a specific monoclonal assay are up to three orders of magnitude higher compared to values obtained by MS/MS technology. The difference is explained by the presence of a high-affinity binder for T1AM (Apolipoprotein B-100) in serum, which permits the immunoassay to measure the total concentration of the analyte but limits MS/MS technology to detect only the unbound (free) analyte, a view, which is contested here.
Collapse
Affiliation(s)
- Hartmut H. Glossmann
- Institut für Biochemische Pharmakologie, Innsbruck, Austria
- *Correspondence: Hartmut H. Glossmann,
| | | |
Collapse
|
19
|
Hoefig CS, Zucchi R, Köhrle J. Thyronamines and Derivatives: Physiological Relevance, Pharmacological Actions, and Future Research Directions. Thyroid 2016; 26:1656-1673. [PMID: 27650974 DOI: 10.1089/thy.2016.0178] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Thyronamines (3-T1AM, T0AM) are endogenous compounds probably derived from L-thyroxine or its intermediate metabolites. Combined activities of intestinal deiodinases and ornithine decarboxylase generate 3-T1AM in vitro. Alternatively, 3-T1AM might be formed by the thyroid gland and secreted into the blood. 3-T1AM and T0AM concentrations have been determined by liquid chromatography-tandem mass spectrometry analysis (LC-MS/MS) in tissues, serum, and cell lines. However, large variations of 3-T1AM concentrations in human serum were reported by LC-MS/MS compared with a monoclonal antibody-based immunoassay. These differences might be caused by strong binding of the highly hydrophobic 3-T1AM to apolipoprotein B100. Pharmacological administration of 3-T1AM results in dose-dependent reversible effects on body temperature, cardiac function, energy metabolism, and neurological functions. The physiological relevance of these actions is unclear, but may occur at tissue concentrations close to the estimated endogenous concentrations of 3-T1AM or its metabolites T0AM or thyroacetic acid (TA1). A number of putative receptors, binding sites, and cellular target molecules mediating actions of the multi-target ligand 3-T1AM have been proposed. Among those are members of the trace amine associated receptor family, the adrenergic receptor ADRα2a, and the thermosensitive transient receptor potential melastatin 8 channel. Preclinical studies employing various animal experimental models are in progress, and more stable receptor-selective agonistic and antagonistic analogues of 3-T1AM are now available for testing. The potent endogenous thyroid hormone-derived biogenic amine 3-T1AM exerts marked cryogenic, metabolic, cardiac and central actions and represents a valuable lead compound linking endocrine, metabolic, and neuroscience research to advance development of new drugs.
Collapse
Affiliation(s)
- Carolin Stephanie Hoefig
- 1 Institut für Experimentelle Endokrinologie Charité, Universitätsmedizin Berlin , Berlin, Germany
| | - Riccardo Zucchi
- 2 Laboratory of Biochemistry, Department of Pathology, University of Pisa , Pisa, Italy
| | - Josef Köhrle
- 1 Institut für Experimentelle Endokrinologie Charité, Universitätsmedizin Berlin , Berlin, Germany
| |
Collapse
|
20
|
Langouche L, Lehmphul I, Perre SV, Köhrle J, Van den Berghe G. Circulating 3-T1AM and 3,5-T2 in Critically Ill Patients: A Cross-Sectional Observational Study. Thyroid 2016; 26:1674-1680. [PMID: 27676423 DOI: 10.1089/thy.2016.0214] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Critical illness is hallmarked by low circulating thyroxine (T4) and triiodothyronine (T3) concentrations, in the presence of elevated reverse T3 (rT3) and low-normal thyrotropin (TSH), referred to as nonthyroidal illness (NTI). Thyroid hormone (TH) metabolism is substantially increased during NTI, in part explained by enhanced deiodinase 3 (D3) activity. T4- and T3-sulfate concentrations are elevated, due to suppressed D1 activity in the presence of unaltered sulfotransferase activity, and 3,3'-diiodothyronine (3,3'-T2) concentrations are normal. To elucidate further the driving forces behind increased TH metabolism during NTI, two other potential T4 metabolites-3,5-diiodothyronine (3,5-T2) and 3-iodothyronamine (3-T1AM)-were measured and related to their potential TH precursors. METHODS Morning blood samples were collected cross-sectionally from 83 critically ill patients on a University Hospital intensive care unit and from 38 demographically matched healthy volunteers. Serum TH and binding proteins were quantified with commercial assays, and 3,5-T2 and 3-T1AM with in-house developed immunoassays. RESULTS Critically ill patients revealed, besides the NTI, a median 44% lower serum 3-T1AM concentration (p < 0.0001) and a 30% higher serum 3,5-T2 concentration (p = 0.01) than healthy volunteers did. Non-survivors and patients diagnosed with sepsis upon admission to the intensive-care unit had significantly higher 3,5-T2 (p ≤ 0.01) but comparable 3-T1AM (p > 0.2) concentrations than other patients did. Multivariable linear regression analysis adjusted for potential precursors revealed that the reduced serum 3-T1AM was positively correlated with the low serum T3 (p < 0.001) but unrelated to serum T4 or rT3. The elevated 3,5-T2 concentration did not independently correlate with TH. CONCLUSIONS Increased TH metabolism during NTI could not be explained by increased conversion to 3-T1AM, as circulating 3-T1AM was suppressed in proportion to the concomitantly low T3 concentrations. Increased conversion of T4 and/or T3 to 3,5-T2 could be possible, as serum 3,5-T2 concentrations were elevated. Whether 3-T1AM or 3,5-T2 plays a functional role during critical illness needs further investigation.
Collapse
Affiliation(s)
- Lies Langouche
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven , Leuven, Belgium
| | - Ina Lehmphul
- 2 Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Sarah Vander Perre
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven , Leuven, Belgium
| | - Josef Köhrle
- 2 Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin , Berlin, Germany
| | - Greet Van den Berghe
- 1 Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven , Leuven, Belgium
| |
Collapse
|
21
|
Gnocchi D, Steffensen KR, Bruscalupi G, Parini P. Emerging role of thyroid hormone metabolites. Acta Physiol (Oxf) 2016; 217:184-216. [PMID: 26748938 DOI: 10.1111/apha.12648] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 07/28/2015] [Accepted: 01/03/2016] [Indexed: 12/15/2022]
Abstract
Thyroid hormones (THs) are essential for the regulation of development and metabolism in key organs. THs produce biological effects both by directly affecting gene expression through the interaction with nuclear receptors (genomic effects) and by activating protein kinases and/or ion channels (short-term effects). Such activations can be either direct, in the case of ion channels, or mediated by membrane or cytoplasmic receptors. Short-term-activated signalling pathways often play a role in the regulation of genomic effects. Several TH intermediate metabolites, which were previously considered without biological activity, have now been associated with a broad range of actions, mostly attributable to short-term effects. Here, we give an overview of the physiological roles and mechanisms of action of THs, focusing on the emerging position that TH metabolites are acquiring as important regulators of physiology and metabolism.
Collapse
Affiliation(s)
- D. Gnocchi
- Division of Clinical Chemistry; Department of Laboratory Medicine; Karolinska Institutet at Karolinska University Hospital Huddinge; Stockholm Sweden
| | - K. R. Steffensen
- Division of Clinical Chemistry; Department of Laboratory Medicine; Karolinska Institutet at Karolinska University Hospital Huddinge; Stockholm Sweden
| | - G. Bruscalupi
- Department of Biology and Biotechnology ‘Charles Darwin’; Sapienza University of Rome; Rome Italy
| | - P. Parini
- Division of Clinical Chemistry; Department of Laboratory Medicine; Karolinska Institutet at Karolinska University Hospital Huddinge; Stockholm Sweden
- Metabolism Unit; Department of Medicine; Karolinska Institutet at Karolinska University Hospital Huddinge; Stockholm Sweden
| |
Collapse
|
22
|
Mondal S, Raja K, Schweizer U, Mugesh G. Chemie und Biologie der Schilddrüsenhormon-Biosynthese und -Wirkung. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601116] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Santanu Mondal
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore Indien
| | - Karuppusamy Raja
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore Indien
| | - Ulrich Schweizer
- Rheinische Friedrich-Wilhelms-Universität Bonn; Institut für Biochemie und Molekularbiologie; Nussallee 11 53115 Bonn Deutschland
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore Indien
| |
Collapse
|
23
|
Mondal S, Raja K, Schweizer U, Mugesh G. Chemistry and Biology in the Biosynthesis and Action of Thyroid Hormones. Angew Chem Int Ed Engl 2016; 55:7606-30. [DOI: 10.1002/anie.201601116] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Santanu Mondal
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore India
| | - Karuppusamy Raja
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore India
| | - Ulrich Schweizer
- Rheinische Friedrich-Wilhelms-Universität Bonn; Institut für Biochemie und Molekularbiologie; Nussallee 11 53115 Bonn Germany
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry; Indian Institute of Science; Bangalore India
| |
Collapse
|
24
|
Hoefig CS, Jacobi SF, Warner A, Harder L, Schanze N, Vennström B, Mittag J. 3-Iodothyroacetic acid lacks thermoregulatory and cardiovascular effects in vivo. Br J Pharmacol 2015; 172:3426-33. [PMID: 25765843 DOI: 10.1111/bph.13131] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 01/20/2015] [Accepted: 03/10/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE 3-Iodothyronamine (3-T1 AM) is an endogenous thyroid hormone derivative reported to induce strong hypothermia and bradycardia within minutes upon injection in rodents. Although 3-T1 AM is rapidly converted to several other metabolites in vivo, these strong pharmacological responses were solely attributed to 3-T1 AM, leaving potential contributions of downstream products untested. We therefore examined the cardiometabolic effects of 3-iodothyroacetic acid (TA1 ), the main degradation product of 3-T1 AM. EXPERIMENTAL APPROACH We used a sensitive implantable radiotelemetry system in C57/Bl6J mice to study the effects of TA1 on body temperature and heart rate, as well as other metabolic parameters. KEY RESULTS Interestingly, despite using pharmacological TA1 doses, we observed no effects on heart rate or body temperature after a single TA1 injection (50 mg·kg(-1) , i.p.) compared to sham-injected controls. Repeated administration of TA1 (5 mg·kg(-1) , i.p. for 7 days) likewise did not alter body weight, food and water intake, heart rate, blood pressure, brown adipose tissue (BAT) thermogenesis or body temperature. Moreover, mRNA expression of tissue specific genes in heart, kidney, liver, BAT and lung was also not altered by TA1 compared to sham-injected controls. CONCLUSIONS AND IMPLICATIONS Our data therefore conclusively demonstrate that TA1 does not contribute to the cardiovascular or thermoregulatory effects observed after 3-T1 AM administration in mice, suggesting that the oxidative deamination constitutes an important deactivation mechanism for 3-T1 AM with possible implications for cardiovascular and thermoregulatory functions.
Collapse
Affiliation(s)
- Carolin S Hoefig
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Simon F Jacobi
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Amy Warner
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Lisbeth Harder
- Center of Brain Behavior and Metabolism CBBM/Medizinische Klinik I, University of Lübeck, Lübeck, Germany
| | - Nancy Schanze
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Björn Vennström
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jens Mittag
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.,Center of Brain Behavior and Metabolism CBBM/Medizinische Klinik I, University of Lübeck, Lübeck, Germany
| |
Collapse
|
25
|
Chiellini G, Nesi G, Digiacomo M, Malvasi R, Espinoza S, Sabatini M, Frascarelli S, Laurino A, Cichero E, Macchia M, Gainetdinov RR, Fossa P, Raimondi L, Zucchi R, Rapposelli S. Design, Synthesis, and Evaluation of Thyronamine Analogues as Novel Potent Mouse Trace Amine Associated Receptor 1 (mTAAR1) Agonists. J Med Chem 2015; 58:5096-107. [DOI: 10.1021/acs.jmedchem.5b00526] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Giulia Nesi
- Deptartment
of Pharmacy, University of Pisa, via Bonanno 6, 56100 Pisa, Italy
| | - Maria Digiacomo
- Deptartment
of Pharmacy, University of Pisa, via Bonanno 6, 56100 Pisa, Italy
| | - Rossella Malvasi
- Deptartment
of Pharmacy, University of Pisa, via Bonanno 6, 56100 Pisa, Italy
| | - Stefano Espinoza
- Department
of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | | | | | - Annunziatina Laurino
- Department
of NEUROFARBA, Section of Pharmacology, University of Florence, 50121 Firenze, Italy
| | - Elena Cichero
- Department
of Pharmacy, University of Genoa, 16126 Genoa, Italy
| | - Marco Macchia
- Deptartment
of Pharmacy, University of Pisa, via Bonanno 6, 56100 Pisa, Italy
| | - Raul R. Gainetdinov
- Institute
of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia
- Skolkovo Institute of Science and Technology (Skoltech), Skolkovo, Moscow region, 143025, Russia
| | - Paola Fossa
- Department
of Pharmacy, University of Genoa, 16126 Genoa, Italy
| | - Laura Raimondi
- Department
of NEUROFARBA, Section of Pharmacology, University of Florence, 50121 Firenze, Italy
| | - Riccardo Zucchi
- Department
of Pathology, University of Pisa, 56100 Pisa, Italy
| | - Simona Rapposelli
- Deptartment
of Pharmacy, University of Pisa, via Bonanno 6, 56100 Pisa, Italy
| |
Collapse
|
26
|
Musilli C, De Siena G, Manni ME, Logli A, Landucci E, Zucchi R, Saba A, Donzelli R, Passani MB, Provensi G, Raimondi L. Histamine mediates behavioural and metabolic effects of 3-iodothyroacetic acid, an endogenous end product of thyroid hormone metabolism. Br J Pharmacol 2015; 171:3476-84. [PMID: 24641572 DOI: 10.1111/bph.12697] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/23/2014] [Accepted: 03/13/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE 3-Iodothyroacetic acid (TA1) is an end product of thyroid hormone metabolism. So far, it is not known if TA1 is present in mouse brain and if it has any pharmacological effects. EXPERIMENTAL APPROACH TA1 levels in mouse brain were measured by HPLC coupled to mass spectrometry. After i.c.v. administration of exogenous TA1 (0.4, 1.32 and 4 μg·kg(-1) ) to mice, memory acquisition-retention (passive avoidance paradigm with a light-dark box), pain threshold to thermal stimulus (51.5°C; hot plate test) and plasma glucose (glucorefractometer) were evaluated. Similar assays were performed in mice pretreated with s.c. injections of the histamine H1 receptor antagonist pyrilamine (10 mg·kg(-1) ) or the H2 receptor antagonist zolantidine (5 mg·kg(-1) ). TA1 (1.32 and 4 μg·kg(-1) ) was also given i.c.v. to mice lacking histidine decarboxylase (HDC(-/-) ) and the corresponding WT strain. KEY RESULTS TA1 was found in the brain of CD1 but not of HDC mice. Exogenous TA1 induced amnesia (at 0.4 μg·kg(-1) ), stimulation of learning (1.32 and 4 μg·kg(-1) ), hyperalgesia (0.4, 1.32 and 4 μg·kg(-1) ) and hyperglycaemia (1.32 and 4 μg·kg(-1) ). All these effects were modulated by pyrilamine and zolantidine. In HDC(-/-) mice, TA1 (1.32 and 4 μg·kg(-1) ) did not increase plasma glucose or induce hyperalgesia. CONCLUSIONS AND IMPLICATIONS Behavioural and metabolic effects of TA1 disclosed interactions between the thyroid and histaminergic systems.
Collapse
Affiliation(s)
- Claudia Musilli
- Department of Pharmacology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zucchi R, Accorroni A, Chiellini G. Update on 3-iodothyronamine and its neurological and metabolic actions. Front Physiol 2014; 5:402. [PMID: 25360120 PMCID: PMC4199266 DOI: 10.3389/fphys.2014.00402] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/28/2014] [Indexed: 11/23/2022] Open
Abstract
3-iodothyronamine (T1AM) is an endogenous amine, that has been detected in many rodent tissues, and in human blood. It has been hypothesized to derive from thyroid hormone metabolism, but this hypothesis still requires validation. T1AM is not a ligand for nuclear thyroid hormone receptors, but stimulates with nanomolar affinity trace amine-associated receptor 1 (TAAR1), a G protein-coupled membrane receptor. With a lower affinity it interacts with alpha2A adrenergic receptors. Additional targets are represented by apolipoprotein B100, mitochondrial ATP synthase, and membrane monoamine transporters, but the functional relevance of these interactions is still uncertain. Among the effects reported after administration of exogenous T1AM to experimental animals, metabolic and neurological responses deserve special attention, because they were obtained at low dosages, which increased endogenous tissue concentration by about one order of magnitude. Systemic T1AM administration favored fatty acid over glucose catabolism, increased ketogenesis and increased blood glucose. Similar responses were elicited by intracerebral infusion, which inhibited insulin secretion and stimulated glucagon secretion. However, T1AM administration increased ketogenesis and gluconeogenesis also in hepatic cell lines and in perfused liver preparations, providing evidence for a peripheral action, as well. In the central nervous system, T1AM behaved as a neuromodulator, affecting adrenergic and/or histaminergic neurons. Intracerebral T1AM administration favored learning and memory, modulated sleep and feeding, and decreased the pain threshold. In conclusion T1AM should be considered as a component of thyroid hormone signaling and might play a significant physiological and/or pathophysiological role. T1AM analogs have already been synthetized and their therapeutical potential is currently under investigation. 3-iodothyronamine (T1AM) is a biogenic amine whose structure is closely related to that of thyroid hormone (3,5,3′-triiodothyronine, or T3). The differences with T3 are the absence of the carboxylate group and the substitution of iodine with hydrogen in 5 and 3′ positions (Figure 1). In this paper we will review the evidence supporting the hypothesis that T1AM is a chemical messenger, namely that it is an endogenous substance able to interact with specific receptors producing significant functional effects. Special emphasis will be placed on neurological and metabolic effects, which are likely to have physiological and pathophysiological importance.
Collapse
Affiliation(s)
- Riccardo Zucchi
- Laboratory of Biochemistry, Department of Pathology, University of Pisa Pisa, Italy
| | - Alice Accorroni
- Laboratory of Biochemistry, Department of Pathology, University of Pisa Pisa, Italy
| | - Grazia Chiellini
- Laboratory of Biochemistry, Department of Pathology, University of Pisa Pisa, Italy
| |
Collapse
|
28
|
Ghelardoni S, Chiellini G, Frascarelli S, Saba A, Zucchi R. Uptake and metabolic effects of 3-iodothyronamine in hepatocytes. J Endocrinol 2014; 221:101-10. [PMID: 24627446 DOI: 10.1530/joe-13-0311] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
3-Iodothyronamine (T1AM) is an endogenous relative of thyroid hormone with profound metabolic effects. In different experimental models, T1AM increased blood glucose, and it is not clear whether this effect is entirely accounted by changes in insulin and/or glucagone secretion. Thus, in the present work, we investigated the uptake of T1AM by hepatocytes, which was compared with the uptake of thyroid hormones, and the effects of T1AM on hepatic glucose and ketone body production. Two different experimental models were used: HepG2 cells and perfused rat liver. Thyronines and thyronamines (T0AMs) were significantly taken up by hepatocytes. In HepG2 cells exposed to 1 μM T1AM, at the steady state, the cellular concentration of T1AM exceeded the medium concentration by six- to eightfold. Similar accumulation occurred with 3,5,3'-triiodothyronine and thyroxine. Liver experiments confirmed significant T1AM uptake. T1AM was partly catabolized and the major catabolites were 3-iodothyroacetic acid (TA1) (in HepG2 cells) and T0AM (in liver). In both preparations, infusion with 1 μM T1AM produced a significant increase in glucose production, if adequate gluconeogenetic substrates were provided. This effect was dampened at higher concentration (10 μM) or in the presence of the amine oxidase inhibitor iproniazid, while TA1 was ineffective, suggesting that T1AM may have a direct gluconeogenetic effect. Ketone body release was significantly increased in liver, while variable results were obtained in HepG2 cells incubated with gluconeogenetic substrates. These findings are consistent with the stimulation of fatty acid catabolism, and a shift of pyruvate toward gluconeogenesis. Notably, these effects are independent from hormonal changes and might have physiological and pathophysiological importance.
Collapse
Affiliation(s)
- Sandra Ghelardoni
- Dipartimento di Patologia Chirurgica Medica Molecolare e dell'Area Critica, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | | | | | | | | |
Collapse
|
29
|
Manni ME, De Siena G, Saba A, Marchini M, Landucci E, Gerace E, Zazzeri M, Musilli C, Pellegrini-Giampietro D, Matucci R, Zucchi R, Raimondi L. Pharmacological effects of 3-iodothyronamine (T1AM) in mice include facilitation of memory acquisition and retention and reduction of pain threshold. Br J Pharmacol 2013; 168:354-62. [PMID: 22889145 DOI: 10.1111/j.1476-5381.2012.02137.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 06/18/2012] [Accepted: 07/28/2012] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE 3-Iodothyronamine (T1AM), an endogenous derivative of thyroid hormones, is regarded as a rapid modulator of behaviour and metabolism. To determine whether brain thyroid hormone levels contribute to these effects, we investigated the effect of central administration of T1AM on learning and pain threshold of mice either untreated or pretreated with clorgyline (2.5 mg·kg(-1) , i.p.), an inhibitor of amine oxidative metabolism. EXPERIMENTAL APPROACH T1AM (0.13, 0.4, 1.32 and 4 μg·kg(-1) ) or vehicle was injected i.c.v. into male mice, and after 30 min their effects on memory acquisition capacity, pain threshold and curiosity were evaluated by the following tests: passive avoidance, licking latency on the hot plate and movements on the hole-board platform. Plasma glycaemia was measured using a glucorefractometer. Brain levels of triiodothyroxine (T3), thyroxine (T4) and T1AM were measured by HPLC coupled to tandem MS. ERK1/2 activation and c-fos expression in different brain regions were evaluated by Western blot analysis. RESULTS T1AM improved learning capacity, decreased pain threshold to hot stimuli, enhanced curiosity and raised plasma glycaemia in a dose-dependent way, without modifying T3 and T4 brain concentrations. T1AM effects on learning and pain were abolished or significantly affected by clorgyline, suggesting a role for some metabolite(s), or that T1AM interacts at the rapid desensitizing target(s). T1AM activated ERK in different brain areas at lower doses than those effective on behaviour. CONCLUSIONS AND IMPLICATIONS T1AM is a novel memory enhancer. This feature might have important implications for the treatment of endocrine and neurodegenerative-induced memory disorders.
Collapse
|
30
|
Murk AJ, Rijntjes E, Blaauboer BJ, Clewell R, Crofton KM, Dingemans MML, Furlow JD, Kavlock R, Köhrle J, Opitz R, Traas T, Visser TJ, Xia M, Gutleb AC. Mechanism-based testing strategy using in vitro approaches for identification of thyroid hormone disrupting chemicals. Toxicol In Vitro 2013; 27:1320-46. [PMID: 23453986 DOI: 10.1016/j.tiv.2013.02.012] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 02/07/2013] [Accepted: 02/18/2013] [Indexed: 11/16/2022]
Abstract
The thyroid hormone (TH) system is involved in several important physiological processes, including regulation of energy metabolism, growth and differentiation, development and maintenance of brain function, thermo-regulation, osmo-regulation, and axis of regulation of other endocrine systems, sexual behaviour and fertility and cardiovascular function. Therefore, concern about TH disruption (THD) has resulted in strategies being developed to identify THD chemicals (THDCs). Information on potential of chemicals causing THD is typically derived from animal studies. For the majority of chemicals, however, this information is either limited or unavailable. It is also unlikely that animal experiments will be performed for all THD relevant chemicals in the near future for ethical, financial and practical reasons. In addition, typical animal experiments often do not provide information on the mechanism of action of THDC, making it harder to extrapolate results across species. Relevant effects may not be identified in animal studies when the effects are delayed, life stage specific, not assessed by the experimental paradigm (e.g., behaviour) or only occur when an organism has to adapt to environmental factors by modulating TH levels. Therefore, in vitro and in silico alternatives to identify THDC and quantify their potency are needed. THDC have many potential mechanisms of action, including altered hormone production, transport, metabolism, receptor activation and disruption of several feed-back mechanisms. In vitro assays are available for many of these endpoints, and the application of modern '-omics' technologies, applicable for in vivo studies can help to reveal relevant and possibly new endpoints for inclusion in a targeted THDC in vitro test battery. Within the framework of the ASAT initiative (Assuring Safety without Animal Testing), an international group consisting of experts in the areas of thyroid endocrinology, toxicology of endocrine disruption, neurotoxicology, high-throughput screening, computational biology, and regulatory affairs has reviewed the state of science for (1) known mechanisms for THD plus examples of THDC; (2) in vitro THD tests currently available or under development related to these mechanisms; and (3) in silico methods for estimating the blood levels of THDC. Based on this scientific review, the panel has recommended a battery of test methods to be able to classify chemicals as of less or high concern for further hazard and risk assessment for THD. In addition, research gaps and needs are identified to be able to optimize and validate the targeted THD in vitro test battery for a mechanism-based strategy for a decision to opt out or to proceed with further testing for THD.
Collapse
Affiliation(s)
- AlberTinka J Murk
- Wageningen University, Sub-department of Toxicology, Tuinlaan 5, 6703 HE Wageningen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Dietrich JW, Landgrafe G, Fotiadou EH. TSH and Thyrotropic Agonists: Key Actors in Thyroid Homeostasis. J Thyroid Res 2012; 2012:351864. [PMID: 23365787 PMCID: PMC3544290 DOI: 10.1155/2012/351864] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 11/21/2012] [Indexed: 12/11/2022] Open
Abstract
This paper provides the reader with an overview of our current knowledge of hypothalamic-pituitary-thyroid feedback from a cybernetic standpoint. Over the past decades we have gained a plethora of information from biochemical, clinical, and epidemiological investigation, especially on the role of TSH and other thyrotropic agonists as critical components of this complex relationship. Integrating these data into a systems perspective delivers new insights into static and dynamic behaviour of thyroid homeostasis. Explicit usage of this information with mathematical methods promises to deliver a better understanding of thyrotropic feedback control and new options for personalised diagnosis of thyroid dysfunction and targeted therapy, also by permitting a new perspective on the conundrum of the TSH reference range.
Collapse
Affiliation(s)
- Johannes W. Dietrich
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
| | - Gabi Landgrafe
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
- Klinik für Allgemein- und Visceralchirurgie, Agaplesion Bethesda Krankenhaus Wuppertal gGmbH, Hainstraße 35, 42109 Wuppertal, NRW, Germany
| | - Elisavet H. Fotiadou
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
| |
Collapse
|
32
|
Hackenmueller SA, Marchini M, Saba A, Zucchi R, Scanlan TS. Biosynthesis of 3-iodothyronamine (T1AM) is dependent on the sodium-iodide symporter and thyroperoxidase but does not involve extrathyroidal metabolism of T4. Endocrinology 2012; 153:5659-67. [PMID: 22948220 PMCID: PMC3473208 DOI: 10.1210/en.2012-1254] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
3-Iodothyronamine (T(1)AM) is an endogenous thyroid hormone derivative with unknown biosynthetic origins. Structural similarities have led to the hypothesis that T(1)AM is an extrathyroidal metabolite of T(4). This study uses an isotope-labeled T(4) [heavy-T(4) (H-T(4))] that can be distinguished from endogenous T(4) by mass spectrometry, which allows metabolites to be identified based on the presence of this unique isotope signature. Endogenous T(1)AM levels depend upon thyroid status and decrease upon induction of hypothyroidism. However, in hypothyroid mice replaced with H-T(4), the isotope-labeled H-T(3) metabolite is detected, but no isotope-labeled T(1)AM is detected. These data suggest that T(1)AM is not an extrathyroidal metabolite of T(4), yet is produced by a process that requires the same biosynthetic factors necessary for T(4) synthesis.
Collapse
Affiliation(s)
- Sarah A Hackenmueller
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, 97239, USA
| | | | | | | | | |
Collapse
|
33
|
Identification and quantification of 3-iodothyronamine metabolites in mouse serum using liquid chromatography-tandem mass spectrometry. J Chromatogr A 2012; 1256:89-97. [PMID: 22885046 DOI: 10.1016/j.chroma.2012.07.052] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 07/10/2012] [Accepted: 07/12/2012] [Indexed: 11/22/2022]
Abstract
3-Iodothyronamine (T(1)AM) is an endogenous derivative of thyroxine. Recently there have been numerous reports of analytical methods to quantify endogenous T(1)AM levels, but substantial discrepancies in concentration depending on the method of analysis (LC-MS/MS or immunoassay) suggest endogenous T(1)AM may be covalently modified in vivo. Using information dependent acquisition methods to perform unbiased scans for T(1)AM metabolites following a single IP injection in mice, we have identified O-sulfonate-T(1)AM, N-acetyl-T(1)AM and T(1)AM-glucuronide as conjugates occurring in vivo, as well as the oxidatively deaminated 3-iodothyroacetic acid and non-iodinated thyroacetic acid. 3-iodothyroacetic acid, O-sulfonate-T(1)AM and T(1)AM-glucuronide are present in serum at greater concentrations that unmodified T(1)AM and all metabolites are extensively distributed to tissues. These results suggest covalent modifications of T(1)AM may play a critical role in regulating distribution and biological activity of T(1)AM, and analytical methods to quantify endogenous T(1)AM should be able to account for these metabolites as well.
Collapse
|
34
|
Hoefig CS, Renko K, Piehl S, Scanlan TS, Bertoldi M, Opladen T, Hoffmann GF, Klein J, Blankenstein O, Schweizer U, Köhrle J. Does the aromatic L-amino acid decarboxylase contribute to thyronamine biosynthesis? Mol Cell Endocrinol 2012; 349:195-201. [PMID: 22061622 DOI: 10.1016/j.mce.2011.10.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 10/19/2011] [Accepted: 10/21/2011] [Indexed: 01/26/2023]
Abstract
Thyronamines (TAM), recently described endogenous signaling molecules, exert metabolic and pharmacological actions partly opposing those of the thyromimetic hormone T(3). TAM biosynthesis from thyroid hormone (TH) precursors requires decarboxylation of the L-alanine side chain and several deiodination steps to convert e.g. L-thyroxine (T(4)) into the most potent 3-T(1)AM. Aromatic L-amino acid decarboxylase (AADC) was proposed to mediate TAM biosynthesis via decarboxylation of TH. This hypothesis was tested by incubating recombinant human AADC, which actively catalyzes dopamine production from DOPA, with several TH. Under all reaction conditions tested, AADC failed to catalyze TH decarboxylation, thus challenging the initial hypothesis. These in vitro observations are supported by detection of 3-T(1)AM in plasma of patients with AADC-deficiency at levels (46 ± 18 nM, n=4) similar to those of healthy controls. Therefore, we propose that the enzymatic decarboxylation needed to form TAM from TH is catalyzed by another unique, perhaps TH-specific, decarboxylase.
Collapse
Affiliation(s)
- Carolin S Hoefig
- Institut für Experimentelle Endokrinologie, Charité - Universitätsmedizin Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Orsi G, Frascarelli S, Zucchi R, Vozzi G. LTI Models for 3-Iodothyronamine Time Dynamics: A Multiscale View. IEEE Trans Biomed Eng 2011; 58:3513-7. [DOI: 10.1109/tbme.2011.2163716] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
36
|
Roy G, Placzek E, Scanlan TS. ApoB-100-containing lipoproteins are major carriers of 3-iodothyronamine in circulation. J Biol Chem 2011; 287:1790-800. [PMID: 22128163 DOI: 10.1074/jbc.m111.275552] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
3-Iodothyronamine (T(1)AM) is a biogenic amine derivative of thyroid hormone present in tissue and blood of vertebrates. Approximately 99% of the circulating thyroid hormones are bound to plasma proteins, including three major thyroid hormone-binding proteins, and the question arises as to whether circulating T(1)AM is also bound to serum factors. We report here that T(1)AM is largely bound to a single protein component of human serum. Using T(1)AM-affinity chromatography, we isolated this protein, and sequence analysis identified it as apolipoprotein B-100 (apoB-100), the protein component of several low density lipoprotein particles. Consistent with this finding, we demonstrate that >90% of specifically bound T(1)AM in human serum resides in the apoB-100-containing low density lipoprotein fraction. T(1)AM reversibly binds to apoB-100-containing lipoprotein particles with an equilibrium dissociation constant (K(D)) of 17 nm and a T(1)AM/apoB-100 stoichiometry of 1:1. Competition binding assays demonstrate that this binding site is highly selective for T(1)AM. Intracellular T(1)AM uptake is significantly enhanced by apoB-100-containing lipoprotein particles. Modest enhancements to apoB-100 cellular uptake and secretion by T(1)AM were observed; however, multidose T(1)AM treatment did not affect lipid or lipoprotein inventory in vivo. Thus, it appears that apoB-100 serves as a carrier of circulating T(1)AM and affords a novel mechanism by which T(1)AM gains entry to cells.
Collapse
Affiliation(s)
- Gouriprassana Roy
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon 97239-3098, USA
| | | | | |
Collapse
|
37
|
Klootwijk W, Friesema ECH, Visser TJ. A nonselenoprotein from amphioxus deiodinates triac but not T3: is triac the primordial bioactive thyroid hormone? Endocrinology 2011; 152:3259-67. [PMID: 21652724 DOI: 10.1210/en.2010-1408] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thyroid hormone (TH) is important for metamorphosis in many species, including the cephalochordate Branchiostoma floridae, a marine invertebrate (amphioxus) living in warmer coastal areas. Branchiostoma expresses a TH receptor, which is activated by 3,3',5-triiodothyroacetic acid (TA(3)) but not by T(3). The Branchiostoma genome also contains multiple genes coding for proteins homologous to iodothyronine deiodinases in vertebrates, selenoproteins catalyzing the activation or inactivation of TH. Three Branchiostoma deiodinases have been cloned: two have a catalytic Sec, and one, bfDy, has a Cys residue. We have studied the catalytic properties of bfDy in transfected COS1 cells by HPLC analysis of reactions with (125)I-labeled substrates and dithiothreitol as cofactor. We could not detect deiodination of T(4), T(3), or rT(3) by bfDy but observed rapid and selective inner ring deiodination (inactivation) of TA(3) and 3,3',5,5'-tetraiodothyroacetic acid (TA(4)). Deiodination of TA(3) by bfDy was optimal at 25 C and 10 mm dithiothreitol. bfDy was extremely labile at 37 C, showing a half-life of less than 2 min, in contrast with a half-life of more than 60 min at 25 C. Deiodination of labeled TA(3) was inhibited dose dependently by unlabeled TA(3)≈TA(4)>T(4)≈T(3). Michaelis-Menten analysis yielded Michaelis-Menten constant values of 6.8 and 68 nm and maximum velocity values of 1.4 and 5.4 pmol/min·mg protein for TA(3) and TA(4), respectively. bfDy was not inhibited by propylthiouracil and iodoacetate and only weakly by goldthioglucose and iopanoic acid. In conclusion, we demonstrate rapid inactivation of TA(3) and TA(4) but not of T(3) and T(4) by the first reported natural nonselenodeiodinase. Our findings support the hypothesis that TA(3) is a primordial bioactive TH.
Collapse
Affiliation(s)
- Wim Klootwijk
- Department of Internal Medicine, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | | |
Collapse
|
38
|
Abstract
Thyronamines (TAMs) are a newly identified class of endogenous signaling compounds. Their structure is identical to that of thyroid hormone and deiodinated thyroid hormone derivatives, except that TAMs do not possess a carboxylate group. Despite some initial publications dating back to the 1950s, TAMs did not develop into an independent area of research until 2004, when they were rediscovered as potential ligands to a class of G protein-coupled receptors called trace-amine associated receptors. Since this discovery, two representatives of TAMs, namely 3-iodothyronamine (3-T(1)AM) and thyronamine (T(0)AM), have been detected in vivo. Intraperitoneal or central injection of 3-T(1)AM or T(0)AM into mice, rats, or Djungarian hamsters caused various prompt effects, such as metabolic depression, hypothermia, negative chronotropy, negative inotropy, hyperglycemia, reduction of the respiratory quotient, ketonuria, and reduction of fat mass. Although their physiological function remains elusive, 3-T(1)AM and T(0)AM have already revealed promising therapeutic potential because they represent the only endogenous compounds inducing hypothermia as a prophylactic or acute treatment of stroke and might thus be expected to cause fewer side effects than synthetic compounds. This review article summarizes the still somewhat scattered data on TAMs obtained both recently and more than 20 yr ago to yield a complete and updated picture of the current state of TAM research.
Collapse
Affiliation(s)
- S Piehl
- Institut für Experimentelle Endokrinologie, Charité-Universitätsmedizin Berlin, Charité Campus Virchow-Klinikum (Südring 10), Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | | | |
Collapse
|
39
|
Saba A, Chiellini G, Frascarelli S, Marchini M, Ghelardoni S, Raffaelli A, Tonacchera M, Vitti P, Scanlan TS, Zucchi R. Tissue distribution and cardiac metabolism of 3-iodothyronamine. Endocrinology 2010; 151:5063-73. [PMID: 20739399 DOI: 10.1210/en.2010-0491] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
3-iodothyronamine (T1AM) is a novel relative of thyroid hormone, able to interact with specific G protein-coupled receptors, known as trace amine-associated receptors. Significant functional effects are produced by exogenous T1AM, including a negative inotropic and chronotropic effect in cardiac preparations. This work was aimed at estimating endogenous T1AM concentration in different tissues and determining its cardiac metabolism. A novel HPLC tandem mass spectrometry assay was developed, allowing detection of T1AM, thyronamine, 3-iodothyroacetic acid, and thyroacetic acid. T1AM was detected in rat serum, at the concentration of 0.3±0.03 pmol/ml, and in all tested organs (heart, liver, kidney, skeletal muscle, stomach, lung, and brain), at concentrations significantly higher than the serum concentration, ranging from 5.6±1.5 pmol/g in lung to 92.9±28.5 pmol/g in liver. T1AM was also identified for the first time in human blood. In H9c2 cardiomyocytes and isolated perfused rat hearts, significant Na+-dependent uptake of exogenous T1AM was observed, and at the steady state total cellular or tissue T1AM concentration exceeded extracellular concentration by more than 20-fold. In both preparations T1AM underwent oxidative deamination to 3-iodothyroacetic acid. T1AM deamination was inhibited by iproniazid but not pargyline or semicarbazide, suggesting the involvement of both monoamine oxidase and semicarbazide-sensitive amine oxidase. Thyronamine and thyroacetic acid were not detected in heart. Finally, evidence of T1AM production was observed in cardiomyocytes exposed to exogenous thyroid hormone, although the activity of this pathway was very low.
Collapse
Affiliation(s)
- Alessandro Saba
- Dipartimento di Chimica e Chimica Industriale, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Saba A, Chiellini G, Frascarelli S, Marchini M, Ghelardoni S, Raffaelli A, Tonacchera M, Vitti P, Scanlan TS, Zucchi R. Tissue Distribution and Cardiac Metabolism of 3-Iodothyronamine. Endocrinology 2010. [DOI: 10.1210/en.2010-0491 pmid: 20739399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
3-Iodothyronamine (T1AM) is a novel relative of thyroid hormone, able to interact with specific G protein-coupled receptors, known as trace amine-associated receptors. Significant functional effects are produced by exogenous T1AM, including a negative inotropic and chronotropic effect in cardiac preparations. This work was aimed at estimating endogenous T1AM concentration in different tissues and determining its cardiac metabolism. A novel HPLC tandem mass spectrometry assay was developed, allowing detection of T1AM, thyronamine, 3-iodothyroacetic acid, and thyroacetic acid. T1AM was detected in rat serum, at the concentration of 0.3 ± 0.03 pmol/ml, and in all tested organs (heart, liver, kidney, skeletal muscle, stomach, lung, and brain), at concentrations significantly higher than the serum concentration, ranging from 5.6 ± 1.5 pmol/g in lung to 92.9 ± 28.5 pmol/g in liver. T1AM was also identified for the first time in human blood. In H9c2 cardiomyocytes and isolated perfused rat hearts, significant Na+-dependent uptake of exogenous T1AM was observed, and at the steady state total cellular or tissue T1AM concentration exceeded extracellular concentration by more than 20-fold. In both preparations T1AM underwent oxidative deamination to 3-iodothyroacetic acid. T1AM deamination was inhibited by iproniazid but not pargyline or semicarbazide, suggesting the involvement of both monoamine oxidase and semicarbazide-sensitive amine oxidase. Thyronamine and thyroacetic acid were not detected in heart. Finally, evidence of T1AM production was observed in cardiomyocytes exposed to exogenous thyroid hormone, although the activity of this pathway was very low.
Collapse
Affiliation(s)
- Alessandro Saba
- Dipartimento di Chimica e Chimica Industriale (A.S.), University of Pisa, 56126 Pisa, Italy
| | - Grazia Chiellini
- Dipartimento di Scienze dell’Uomo e dell’Ambiente (G.C., S.F., M.M., S.G., R.Z.), University of Pisa, 56126 Pisa, Italy
| | - Sabina Frascarelli
- Dipartimento di Scienze dell’Uomo e dell’Ambiente (G.C., S.F., M.M., S.G., R.Z.), University of Pisa, 56126 Pisa, Italy
| | - Maja Marchini
- Dipartimento di Scienze dell’Uomo e dell’Ambiente (G.C., S.F., M.M., S.G., R.Z.), University of Pisa, 56126 Pisa, Italy
| | - Sandra Ghelardoni
- Dipartimento di Scienze dell’Uomo e dell’Ambiente (G.C., S.F., M.M., S.G., R.Z.), University of Pisa, 56126 Pisa, Italy
| | - Andrea Raffaelli
- Consiglio Nazionale delle Ricerche (A.R.), Istituto di Chimica dei Composti Organo Metallici, Pisa, Italy
| | - Massimo Tonacchera
- Dipartimento di Endocrinologia (M.T., P.V.), University of Pisa, 56126 Pisa, Italy
| | - Paolo Vitti
- Dipartimento di Endocrinologia (M.T., P.V.), University of Pisa, 56126 Pisa, Italy
| | - Thomas S. Scanlan
- Departments of Physiology and Pharmacology and Cell and Developmental Biology (T.S.S.), Oregon Health and Science University, Portland, Oregon 97239
| | - Riccardo Zucchi
- Dipartimento di Scienze dell’Uomo e dell’Ambiente (G.C., S.F., M.M., S.G., R.Z.), University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
41
|
Abstract
3-Iodothyronamine (T(1)AM) is an endogenous compound with chemical features that are similar to thyroid hormone. T(1)AM has a carbon skeleton identical to that of T(4) and contains a single carbon-iodine bond. Theoretically, T(1)AM could be produced from T(4) by enzymatic decarboxylation and deiodination. Recent studies show that T(1)AM and higher iodinated thyronamines are subject to similar metabolic processing as iodothyronines such as T(4), suggesting a biological linkage between iodothyronines and iodothyronamines. In addition, single doses of T(1)AM administered to rodents induce a hypometabolic state that in certain ways resembles hibernation and is opposite to the effects of excess T(4). This review will discuss the latest developments on this recently discovered thyroid hormone derivative.
Collapse
Affiliation(s)
- Thomas S Scanlan
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon 97239-3098, USA.
| |
Collapse
|