1
|
Sabatino L, Lapi D, Del Seppia C. Factors and Mechanisms of Thyroid Hormone Activity in the Brain: Possible Role in Recovery and Protection. Biomolecules 2024; 14:198. [PMID: 38397435 PMCID: PMC10886502 DOI: 10.3390/biom14020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Thyroid hormones (THs) are essential in normal brain development, and cognitive and emotional functions. THs act through a cascade of events including uptake by the target cells by specific cell membrane transporters, activation or inactivation by deiodinase enzymes, and interaction with nuclear thyroid hormone receptors. Several thyroid responsive genes have been described in the developing and in the adult brain and many studies have demonstrated a systemic or local reduction in TH availability in neurologic disease and after brain injury. In this review, the main factors and mechanisms associated with the THs in the normal and damaged brain will be evaluated in different regions and cellular contexts. Furthermore, the most common animal models used to study the role of THs in brain damage and cognitive impairment will be described and the use of THs as a potential recovery strategy from neuropathological conditions will be evaluated. Finally, particular attention will be given to the link observed between TH alterations and increased risk of Alzheimer's Disease (AD), the most prevalent neurodegenerative and dementing condition worldwide.
Collapse
Affiliation(s)
- Laura Sabatino
- Institute of Clinical Physiology, National Council of Research, Via Moruzzi 1, 56124 Pisa, Italy;
| | - Dominga Lapi
- Department of Biology, University of Pisa, 56127 Pisa, Italy;
| | - Cristina Del Seppia
- Institute of Clinical Physiology, National Council of Research, Via Moruzzi 1, 56124 Pisa, Italy;
| |
Collapse
|
2
|
Goto K, Kimura R, Masuda R, Karasaki T, Sase S. Demonstration of the Formation of a Selenocysteine Selenenic Acid through Hydrolysis of a Selenocysteine Selenenyl Iodide Utilizing a Protective Molecular Cradle. Molecules 2023; 28:7972. [PMID: 38138461 PMCID: PMC10746021 DOI: 10.3390/molecules28247972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Selenocysteine selenenic acids (Sec-SeOHs) and selenocysteine selenenyl iodides (Sec-SeIs) have long been recognized as crucial intermediates in the catalytic cycle of glutathione peroxidase (GPx) and iodothyronine deiodinase (Dio), respectively. However, the observation of these reactive species remained elusive until our recent study, where we successfully stabilized Sec-SeOHs and Sec-SeIs using a protective molecular cradle. Here, we report the first demonstration of the chemical transformation from a Sec-SeI to a Sec-SeOH through alkaline hydrolysis. A stable Sec-SeI derived from a selenocysteine methyl ester was synthesized using the protective cradle, and its structure was determined by crystallographic analysis. The alkaline hydrolysis of the Sec-SeI at -50 °C yielded the corresponding Sec-SeOH in an 89% NMR yield, the formation of which was further confirmed by its reaction with dimedone. The facile and nearly quantitative conversion of the Sec-SeI to the Sec-SeOH not only validates the potential involvement of this process in the catalytic mechanism of Dio, but also highlights its utility as a method for producing a Sec-SeOH.
Collapse
Affiliation(s)
- Kei Goto
- Department of Chemistry, School of Science, Tokyo Institute of Technology, Tokyo 152-8551, Japan
| | | | | | | | | |
Collapse
|
3
|
Marino L, Kim A, Ni B, Celi FS. Thyroid hormone action and liver disease, a complex interplay. Hepatology 2023:01515467-990000000-00521. [PMID: 37535802 DOI: 10.1097/hep.0000000000000551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/05/2023] [Indexed: 08/05/2023]
Abstract
Thyroid hormone action is involved in virtually all physiological processes. It is well known that the liver and thyroid are intimately linked, with thyroid hormone playing important roles in de novo lipogenesis, beta-oxidation (fatty acid oxidation), cholesterol metabolism, and carbohydrate metabolism. Clinical and mechanistic research studies have shown that thyroid hormone can be involved in chronic liver diseases, including alcohol-associated or NAFLD and HCC. Thyroid hormone action and synthetic thyroid hormone analogs can exert beneficial actions in terms of lowering lipids, preventing chronic liver disease and as liver anticancer agents. More recently, preclinical and clinical studies have indicated that some analogs of thyroid hormone could also play a role in the treatment of liver disease. These synthetic molecules, thyromimetics, can modulate lipid metabolism, particularly in NAFLD/NASH. In this review, we first summarize the thyroid hormone signaling axis in the context of liver biology, then we describe the changes in thyroid hormone signaling in liver disease and how liver diseases affect the thyroid hormone homeostasis, and finally we discuss the use of thyroid hormone-analog for the treatment of liver disease.
Collapse
Affiliation(s)
- Luigi Marino
- Department of Medicine, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - Adam Kim
- Division of Gastroenterology and Hepatology, Department of Medicine, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| | - Bin Ni
- Alliance Pharma, Philadelphia, Pennsylvania, USA
| | - Francesco S Celi
- Department of Medicine, UConn Health, University of Connecticut, Farmington, Connecticut, USA
| |
Collapse
|
4
|
Marsan ES, Dreab A, Bayse CA. In silico insights into the dimer structure and deiodinase activity of type III iodothyronine deiodinase from bioinformatics, molecular dynamics simulations, and QM/MM calculations. J Biomol Struct Dyn 2023; 41:4819-4829. [PMID: 35579922 PMCID: PMC9878935 DOI: 10.1080/07391102.2022.2073271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/27/2022] [Indexed: 01/28/2023]
Abstract
The homodimeric family of iodothyronine deiodinases (Dios) regioselectively remove iodine from thyroid hormones. Currently, structural data has only been reported for the monomer of the mus type III thioredoxin (Trx) fold catalytic domain (Dio3Trx), but the mode of dimerization has not yet been determined. Various groups have proposed dimer structures that are similar to the A-type and B-type dimerization modes of peroxiredoxins. Computational methods are used to compare the sequence of Dio3Trx to related proteins known to form A-type and B-type dimers. Sequence analysis and in silico protein-protein docking methods suggest that Dio3Trx is more consistent with proteins that adopt B-type dimerization. Molecular dynamics (MD) simulations of the refined Dio3Trx dimer constructed using the SymmDock and GalaxyRefineComplex databases indicate stable dimer formation along the β4α3 interface consistent with other Trx fold B-type dimers. Free energy calculations show that the dimer is stabilized by interdimer interactions between the β-sheets and α-helices. A comparison of MD simulations of the apo and thyroxine-bound dimers suggests that the active site binding pocket is not affected by dimerization. Determination of the transition state for deiodination of thyroxine from the monomer structure using QM/MM methods provides an activation barrier consistent with previous small model DFT studies.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Eric S Marsan
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA
| | - Ana Dreab
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA
| | - Craig A Bayse
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA
| |
Collapse
|
5
|
Liu M, Yu Z, Yang F, Zhao Z, Zhou M, Wang C, Zhang B, Liang G, Liu X, Shao J. BDE209-promoted Dio2 degradation in H4 glioma cells through the autophagy pathway, resulting in hypothyroidism and leading to neurotoxicity. Toxicology 2023:153581. [PMID: 37330034 DOI: 10.1016/j.tox.2023.153581] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/07/2023] [Accepted: 06/15/2023] [Indexed: 06/19/2023]
Abstract
Decabromodiphenyl ether (BDE209), the homologue with the highest number of brominates in polybrominated diphenyl ethers (PBDEs), is one of the most widespread environmental persistent organic pollutants (POPs) due to its mass production and extensive application in recent decades. BDE209 is neurotoxic, possibly related to its interference with the thyroid hormone (TH) system. However, the underlying molecular mechanisms of BDE209-induced TH interference and neurobehavioral disorders remains unknown. Here, we explored how BDE209 manipulated the major enzyme, human type II iodothyronine deiodinase (Dio2), that is most important in regulating local cerebral TH equilibrium by neuroglial cells, using an in vitro model of human glioma H4 cells. Clonogenic cell survival assay and LC/MS/MS analysis showed that BDE209 could induce chronic neurotoxicity by inducing TH interference. Co-IP assay, RT-qPCR and confocal assay identified that BDE209 destroyed the stability of Dio2 without affecting its expression, and promoted its binding to p62, thereby enhancing its autophagic degradation, thus causing TH metabolism disorder and neurotoxicity. Furthermore, molecular docking studies predicted that BDE209 could effectively suppress Dio2 activity by competing with tetraiodothyronine (T4). Collectively, our study demonstrates that BDE209-induced Dio2 degradation and loss of its enzymatic activity in neuroglial cells are the fundamental pathogenic basis for BDE209-mediated cerebral TH disequilibrium and neurotoxicity, providing a target of interest for further investigation using glial/neuronal cell co-culture system and in vivo models.
Collapse
Affiliation(s)
- Min Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China; Neurology Department, Dalian University Affiliated Xinhua Hospital, Dalian, 116021, China
| | - Zhenlong Yu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Fangyu Yang
- General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Department of Neurosurgery, Shenyang, China
| | - Zikuang Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 116000, China
| | - Meirong Zhou
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Chao Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Baojing Zhang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Guobiao Liang
- General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Department of Neurosurgery, Shenyang, China.
| | - Xiaohui Liu
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Jing Shao
- Department of Environmental Health and Toxicology, School of Public Health, Dalian Medical University, Dalian, 116044, China; Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine; Liaoning Medical Center for Hematopoietic Stem Cell Transplantation; Dalian Key Laboratory of Hematology; Diamond Bay Institute of Hematology; Second Hospital of Dalian Medical University, Dalian, 116027, China.
| |
Collapse
|
6
|
Chaudière J. Biological and Catalytic Properties of Selenoproteins. Int J Mol Sci 2023; 24:10109. [PMID: 37373256 DOI: 10.3390/ijms241210109] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Selenocysteine is a catalytic residue at the active site of all selenoenzymes in bacteria and mammals, and it is incorporated into the polypeptide backbone by a co-translational process that relies on the recoding of a UGA termination codon into a serine/selenocysteine codon. The best-characterized selenoproteins from mammalian species and bacteria are discussed with emphasis on their biological function and catalytic mechanisms. A total of 25 genes coding for selenoproteins have been identified in the genome of mammals. Unlike the selenoenzymes of anaerobic bacteria, most mammalian selenoenzymes work as antioxidants and as redox regulators of cell metabolism and functions. Selenoprotein P contains several selenocysteine residues and serves as a selenocysteine reservoir for other selenoproteins in mammals. Although extensively studied, glutathione peroxidases are incompletely understood in terms of local and time-dependent distribution, and regulatory functions. Selenoenzymes take advantage of the nucleophilic reactivity of the selenolate form of selenocysteine. It is used with peroxides and their by-products such as disulfides and sulfoxides, but also with iodine in iodinated phenolic substrates. This results in the formation of Se-X bonds (X = O, S, N, or I) from which a selenenylsulfide intermediate is invariably produced. The initial selenolate group is then recycled by thiol addition. In bacterial glycine reductase and D-proline reductase, an unusual catalytic rupture of selenium-carbon bonds is observed. The exchange of selenium for sulfur in selenoproteins, and information obtained from model reactions, suggest that a generic advantage of selenium compared with sulfur relies on faster kinetics and better reversibility of its oxidation reactions.
Collapse
Affiliation(s)
- Jean Chaudière
- CBMN (CNRS, UMR 5248), University of Bordeaux, 33600 Pessac, France
| |
Collapse
|
7
|
Mayasich SA, Goldsmith MR, Mattingly KZ, LaLone CA. Combining In Vitro and In Silico New Approach Methods to Investigate Type 3 Iodothyronine Deiodinase Chemical Inhibition Across Species. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2023; 42:1032-1048. [PMID: 36825751 PMCID: PMC10895443 DOI: 10.1002/etc.5591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
New approach methodologies (NAMs) are being developed to reduce and replace vertebrate animal testing in support of ecotoxicology and risk assessment. The US Environmental Protection Agency's Sequence Alignment to Predict Across Species Susceptibility (SeqAPASS) bioinformatic tool was used to evaluate amino acid sequence conservation of the type 3 iodothyronine deiodinase (DIO3) enzyme across species to demonstrate NAM applications for understanding effects of chemical interactions with a specific protein target. Existing literature was used to identify critical amino acids for thyroid hormone binding and interaction with a reducing cofactor. The SeqAPASS tool identifies whether known critical amino acids involved in ligand binding are exact, partial, or not matches across species compared with a template species based on molecular weight and side chain classification. This evaluation guided the design of variant proteins representing critical amino acid substitutions found in various species. Site-directed mutagenesis of the wild-type (WT) human DIO3 gene sequence was used to create six variant proteins expressed in cell culture, which were then tested in vitro for chemical inhibition. Significant differences in in vitro median inhibitory concentration results were observed among variants for potential competitive inhibitors. A molecular model representing the WT human DIO3 was constructed using Molecular Operating Environment (MOE) software and mutated in silico to create the six variants. The MOE Site Finder tool identified the proposed catalytic and cofactor sites and potential alternative binding sites. Virtual docking did not provide affinity scores with sufficient resolution to rank the potency of the chemical inhibitors. Chemical characteristics, function and location of substituted amino acids, and complexities of the protein target are important considerations in developing NAMs to evaluate chemical susceptibility across species. Environ Toxicol Chem 2023;42:1032-1048. © 2023 University of Wisconsin-Madison. Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Sally A. Mayasich
- Aquatic Sciences Center, University of Wisconsin‐Madison, Madison, Wisconsin, USA
- Office of Research and Development, Center for Computational Toxicology and Ecology, Great Lakes Toxicology and Ecology Division, US Environmental Protection Agency, Duluth, Minnesota
| | - Michael R. Goldsmith
- Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina
- Congruence Therapeutics, Montreal, Quebec, Canada
| | | | - Carlie A. LaLone
- Office of Research and Development, Center for Computational Toxicology and Ecology, Great Lakes Toxicology and Ecology Division, US Environmental Protection Agency, Duluth, Minnesota
| |
Collapse
|
8
|
Giri D, Raja K, Mugesh G. A Simple Substitution on Thyroid Hormones Remarkably Alters the Regioselectivity of Deiodination by a Deiodinase Mimic. Chemistry 2023; 29:e202203111. [PMID: 36380701 DOI: 10.1002/chem.202203111] [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: 10/06/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022]
Abstract
The regioselective deiodinations of L-thyroxine (T4) play key roles in the thyroid hormone homeostasis. These reactions are catalyzed by three isoforms of the selenoenzymes, iodothyronine deiodinases (Dio1, Dio2 and Dio3), which are highly homologous in nature. Dio1 mediates 5'- or 5-deiodinations of T4 to produce T3 and rT3, respectively. In contrast, Dio2 and Dio3 are selective to 5'- or 5-deiodination to produce T3 and rT3, respectively. Understanding of the regioselectivity of deiodination at the molecular level is important as abnormal levels of thyroid hormone have been implicated in various clinical conditions, such as hypoxia, myocardial infarction, neuronal ischemia and cancer. In this paper, we report that the electronic properties of the iodine atoms in thyroxine (T4) can be modulated through a simple substitution in the 4'-phenolic moiety. This leads to the change in the regioselectivity of deiodination by different small molecule mimics of Dio enzymes. By using this chemical approach, we also show that the substitution of a strong electron withdrawing group facilitates the removal of all four iodine atoms in the T4 derivative. Theoretical investigations on the hydrogen bonded adducts of T4 with imidazole indicate that the charge on the iodine atoms depend on the nature of hydrogen bond between the -OH group of T4 and the imidazole moiety. While the imidazole can act as either hydrogen bond acceptor (HBA) or hydrogen bond donor (HBD), the protonated imidazole acts exclusively as HBD in T4-imidazole complex. These studies support the earlier observations that the histidine residue at the active sites of the deiodinases play an important role not only in the substrate binding, but also in altering the regioselectivity of the deiodination reactions.
Collapse
Affiliation(s)
- Debasish Giri
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Karuppusamy Raja
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
9
|
Arai K, Toba H, Yamamoto N, Ito M, Mikami R. Modeling Type-1 Iodothyronine Deiodinase with Peptide-Based Aliphatic Diselenides: Potential Role of Highly Conserved His and Cys Residues as a General Acid Catalyst. Chemistry 2023; 29:e202202387. [PMID: 36254793 DOI: 10.1002/chem.202202387] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Indexed: 11/07/2022]
Abstract
Type-1 iodothyronine deiodinase (ID-1) catalyzes the reductive elimination of 5'-I and 5-I on the phenolic and tyrosyl rings of thyroxine (T4), respectively. Chemically verifying whether I atoms with different chemical properties undergo deiodination through a common mechanism is challenging. Herein, we report the modeling of ID-1 using aliphatic diselenide (Se-Se) and selenenylsulfide (Se-S) compounds. Mechanistic investigations of deiodination using the ID-1-like reagents suggested that the 5'-I and 5-I deiodinations proceed via the same mechanism through an unstable intermediate containing a Se⋅⋅⋅I halogen bond between a selenolate anion, reductively produced from Se-Se (or Se-S) in the compound, and an I atom in T4. Moreover, imidazolium and thiol groups, which may act as general acid catalysts, promoted the heterolytic cleavage of the C-I bond in the Se⋅⋅⋅I intermediate, which is the rate-determining step, by donating a proton to the C atom.
Collapse
Affiliation(s)
- Kenta Arai
- Department of Chemistry, School of Science, Tokai University Kitakaname, Hiratsuka-shi, Kanagawa, 259-1292, Japan.,Institute of Advanced Biosciences, Tokai University Kitakaname, Hiratsuka-shi, Kanagawa, 259-1292, Japan
| | - Haruka Toba
- Department of Chemistry, School of Science, Tokai University Kitakaname, Hiratsuka-shi, Kanagawa, 259-1292, Japan
| | - Nozomi Yamamoto
- Department of Chemistry, School of Science, Tokai University Kitakaname, Hiratsuka-shi, Kanagawa, 259-1292, Japan
| | - Mao Ito
- Department of Chemistry, School of Science, Tokai University Kitakaname, Hiratsuka-shi, Kanagawa, 259-1292, Japan
| | - Rumi Mikami
- Department of Chemistry, School of Science, Tokai University Kitakaname, Hiratsuka-shi, Kanagawa, 259-1292, Japan
| |
Collapse
|
10
|
Zhang W, Liang H. Relationships Between Maternal Selected Metals (Cu, Mg, Zn and Fe), Thyroid Function and Blood Glucose Levels During Pregnancy. Biol Trace Elem Res 2022:10.1007/s12011-022-03455-5. [PMID: 36418636 DOI: 10.1007/s12011-022-03455-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/18/2022] [Indexed: 11/27/2022]
Abstract
The aim of this study were to understand the intake of selected metals (copper (Cu), zinc (Zn), iron (Fe) and magnesium (Mg)) during pregnancy; to detect serum Cu, Mg, Zn and Fe levels in pregnant women; to analyze the relationships among the selected metals, maternal thyroid function and fasting blood glucose (FBG) levels; to investigate the impact of the selected metals and maternal thyroid function on the risk of gestational diabetes mellitus (GDM); and to provide clinical value for the rational intake of the selected metals and iodine during pregnancy to ensure normal fetal development. The population was recruited from pregnant women presenting to the obstetrics outpatient clinic of Shanxi Provincial People's Hospital (February 2021 to April 2022). Selected metal, thyroid hormone (TH (free thyroxine (FT4), free tri-iodothyronine (FT3), and thyroid-stimulating hormone (TSH)) and FBG levels were measured in pregnant women during early, middle and late pregnancy. Covariance analysis was used to analyze the overall trends in selected metal, TH and FBG levels during pregnancy, and binary logistic regression models were used to assess the impacts of the selected metals and thyroid function on the risk of GDM. In addtion, the potential mediation effects of thyroid functions were explored in the mediation analyses. A total of 65 pregnant women were included in this study. Regression models showed that maternal Mg and Cu levels were positively associated with the risk of GDM, conversely, logFT4 was negatively associated with the risk of GDM. Mediation analyses suggested that the associations between the selected metals (Zn, Cu and Mg) and GDM might be mediated by FT3 levels, and that the Cu-GDM and Zn-GDM association could be explained by FT4 levels. Additionally, the Zn-GDM association could also potentially be mediated by the FT3/FT4 ratio. Our findings suggest that Mg, Cu and FT4 levels may act as influencing factors for the development of GDM, and maternal FT3, FT4 and the FT3/FT4 ratio might be the potential mediators of the associations between the selected metals and GDM risk during pregnancy.
Collapse
Affiliation(s)
- WeiYi Zhang
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, 030000, Shanxi, China
| | - HongPing Liang
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, 030000, Shanxi, China.
| |
Collapse
|
11
|
Köhrle J, Frädrich C. Deiodinases control local cellular and systemic thyroid hormone availability. Free Radic Biol Med 2022; 193:59-79. [PMID: 36206932 DOI: 10.1016/j.freeradbiomed.2022.09.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022]
Abstract
Iodothyronine deiodinases (DIO) are a family of selenoproteins controlling systemic and local availability of the major thyroid hormone l-thyroxine (T4), a prohormone secreted by the thyroid gland. T4 is activated to the active 3,3'-5-triiodothyronine (T3) by two 5'-deiodinases, DIO1 and DIO2. DIO3, a 5-deiodinase selenoenzyme inactivates both the prohormone T4 and its active form T3. DIOs show species-specific different patterns of temporo-spatial expression, regulation and function and exhibit different mechanisms of reaction and inhibitor sensitivities. The main regulators of DIO expression and function are the thyroid hormone status, several growth factors, cytokines and altered pathophysiological conditions. Selenium (Se) status has a modest impact on DIO expression and translation. DIOs rank high in the priority of selenium supply to various selenoproteins; thus, their function is impaired only during severe selenium deficiency. DIO variants, polymorphisms, SNPs and rare mutations have been identified. Development of DIO isozyme selective drugs is ongoing. A first X-ray structure has been reported for DIO3. This review focusses on the biochemical characteristics and reaction mechanisms, the relationships between DIO selenoproteins and their importance for local and systemic provision of the active hormone T3. Nutritional, pharmacological, and environmental factors and inhibitors, such as endocrine disruptors, impact DIO functions.
Collapse
Affiliation(s)
- Josef Köhrle
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Max Rubner Center (MRC) für Kardiovaskuläre-metabolische-renale Forschung in Berlin, Institut für Experimentelle Endokrinologie, 10115, Berlin, Germany.
| | - Caroline Frädrich
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Max Rubner Center (MRC) für Kardiovaskuläre-metabolische-renale Forschung in Berlin, Institut für Experimentelle Endokrinologie, 10115, Berlin, Germany
| |
Collapse
|
12
|
Emamnejad R, Dass M, Mahlis M, Bozkurt S, Ye S, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. Thyroid hormone-dependent oligodendroglial cell lineage genomic and non-genomic signaling through integrin receptors. Front Pharmacol 2022; 13:934971. [PMID: 36133808 PMCID: PMC9483185 DOI: 10.3389/fphar.2022.934971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous autoimmune disease whereby the pathological sequelae evolve from oligodendrocytes (OLs) within the central nervous system and are targeted by the immune system, which causes widespread white matter pathology and results in neuronal dysfunction and neurological impairment. The progression of this disease is facilitated by a failure in remyelination following chronic demyelination. One mediator of remyelination is thyroid hormone (TH), whose reliance on monocarboxylate transporter 8 (MCT8) was recently defined. MCT8 facilitates the entry of THs into oligodendrocyte progenitor cell (OPC) and pre-myelinating oligodendrocytes (pre-OLs). Patients with MS may exhibit downregulated MCT8 near inflammatory lesions, which emphasizes an inhibition of TH signaling and subsequent downstream targeted pathways such as phosphoinositide 3-kinase (PI3K)-Akt. However, the role of the closely related mammalian target of rapamycin (mTOR) in pre-OLs during neuroinflammation may also be central to the remyelination process and is governed by various growth promoting signals. Recent research indicates that this may be reliant on TH-dependent signaling through β1-integrins. This review identifies genomic and non-genomic signaling that is regulated through mTOR in TH-responsive pre-OLs and mature OLs in mouse models of MS. This review critiques data that implicates non-genomic Akt and mTOR signaling in response to TH-dependent integrin receptor activation in pre-OLs. We have also examined whether this can drive remyelination in the context of neuroinflammation and associated sequelae. Importantly, we outline how novel therapeutic small molecules are being designed to target integrin receptors on oligodendroglial lineage cells and whether these are viable therapeutic options for future use in clinical trials for MS.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Michael Mahlis
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Paschalis Theotokis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
- *Correspondence: Steven Petratos,
| |
Collapse
|
13
|
Rodriguez-Ruiz A, Braun D, Pflug S, Brol A, Sylvester M, Steegborn C, Schweizer U. Insights into the Mechanism of Human Deiodinase 1. Int J Mol Sci 2022; 23:5361. [PMID: 35628173 PMCID: PMC9141512 DOI: 10.3390/ijms23105361] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 02/07/2023] Open
Abstract
The three isoenzymes of iodothyronine deiodinases (DIO1-3) are membrane-anchored homo-dimeric selenoproteins which share the thioredoxin-fold structure. Several questions regarding their catalytic mechanisms still remain open. Here, we addressed the roles of several cysteines which are conserved among deiodinase isoenzymes and asked whether they may contribute to dimerization and reduction of the oxidized enzyme with physiological reductants. We also asked whether amino acids previously identified in DIO3 play the same role in DIO1. Human DIO1 and 2 were recombinantly expressed in insect cells with selenocysteine replaced with cysteine (DIO1U126C) or in COS7 cells as selenoprotein. Enzyme activities were studied by radioactive deiodination assays with physiological reducing agents and recombinant proteins were characterized by mass spectrometry. Mutation of Cys124 in DIO1 prevented reduction by glutathione, while 20 mM dithiothreitol still regenerated the enzyme. Protein thiol reductants, thioredoxin and glutaredoxin, did not reduce DIO1U126C. Mass spectrometry demonstrated the formation of an intracellular disulfide between the side-chains of Cys124 and Cys(Sec)126. We conclude that the proximal Cys124 forms a selenenyl-sulfide with the catalytic Sec126 during catalysis, which is the substrate of the physiological reductant glutathione. Mutagenesis studies support the idea of a proton-relay pathway from solvent to substrate that is shared between DIO1 and DIO3.
Collapse
Affiliation(s)
- Alfonso Rodriguez-Ruiz
- Institut für Biochemie und Molekularbiologie, Universitätsklinikum Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany; (A.R.-R.); (D.B.); (S.P.); (A.B.)
| | - Doreen Braun
- Institut für Biochemie und Molekularbiologie, Universitätsklinikum Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany; (A.R.-R.); (D.B.); (S.P.); (A.B.)
| | - Simon Pflug
- Institut für Biochemie und Molekularbiologie, Universitätsklinikum Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany; (A.R.-R.); (D.B.); (S.P.); (A.B.)
| | - Alexander Brol
- Institut für Biochemie und Molekularbiologie, Universitätsklinikum Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany; (A.R.-R.); (D.B.); (S.P.); (A.B.)
| | - Marc Sylvester
- Core Facility Mass Spectrometry, Universitätsklinikum Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany;
| | | | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Universitätsklinikum Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany; (A.R.-R.); (D.B.); (S.P.); (A.B.)
| |
Collapse
|
14
|
Wu W, Lu J, Ruan X, Ma C, Lu W, Luo Y, Luo D, Mu X. Maternal essential metals, thyroid hormones, and fetal growth: Association and mediation analyses in Chinese pregnant women. J Trace Elem Med Biol 2021; 68:126809. [PMID: 34153934 DOI: 10.1016/j.jtemb.2021.126809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/24/2021] [Accepted: 06/10/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Essential metals play critical roles in fetal growth and development, but results from human studies are inconsistent. Additionally, whether maternal thyroid hormone (TH) levels mediate the associations between essential metals and fetal growth remains unknown. METHODS Data for analysis were extracted from the Information System of Guangdong Women and Children Hospital between January 2017 and December 2019. Maternal levels of essential metals [copper (Cu), zinc (Zn), magnesium (Mg), and iron (Fe)] and THs were measured at the second trimester. Multivariate linear models were introduced to evaluate the potential associations between maternal essential metals, thyroid functions, and fetal growth, and the possible mediation effects of thyroid functions were explored in the median analyses. RESULTS A total of 4186 mother-infant pairs were included in the present study. Maternal Fe levels were found to significantly increase birth weight in 272.91 g (95 % CI: 15.59, 530.22) among anemia group. Maternal Cu levels were positively associated with increased free triiodothyronine/free thyroxine ratio (FT3/FT4). Negative associations of Fe and Mg levels with thyroid-stimulating hormone (TSH) concentrations were observed, accompanied with the positive associations in relation to FT3, FT4 and FT3/FT4 ratio. Mediation analyses suggested that 72.01 % of the associations between Fe levels and birth length might be mediated by FT3 levels. Additionally, 25.85 % of the Cu-birth length association and 44.53 % of the Fe-birth length association could be explained by FT3/FT4 ratio. CONCLUSION Our findings suggest that maternal Cu, Mg, and Fe levels can alter TH concentrations, and maternal FT3 and FT3/FT4 ratio might be potential mediators on the developmental effects of Cu and Fe levels.
Collapse
Affiliation(s)
- Weixiang Wu
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, 511443, China
| | - Jianhua Lu
- Department of Information, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xiaolin Ruan
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, 511443, China
| | - Congli Ma
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, 511443, China
| | - Weiming Lu
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, 511443, China
| | - Yasha Luo
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, 511443, China
| | - Dan Luo
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, China
| | - Xiaoping Mu
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, 511443, China.
| |
Collapse
|
15
|
Targeting the DIO3 enzyme using first-in-class inhibitors effectively suppresses tumor growth: a new paradigm in ovarian cancer treatment. Oncogene 2021; 40:6248-6257. [PMID: 34556811 DOI: 10.1038/s41388-021-02020-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/26/2021] [Accepted: 09/10/2021] [Indexed: 02/08/2023]
Abstract
The enzyme iodothyronine deiodinase type 3 (DIO3) contributes to cancer proliferation by inactivating the tumor-suppressive actions of thyroid hormone (T3). We recently established DIO3 involvement in the progression of high-grade serous ovarian cancer (HGSOC). Here we provide a link between high DIO3 expression and lower survival in patients, similar to common disease markers such as Ki67, PAX8, CA-125, and CCNE1. These observations suggest that DIO3 is a logical target for inhibition. Using a DIO3 mimic, we developed original DIO3 inhibitors that contain a core of dibromomaleic anhydride (DBRMD) as scaffold. Two compounds, PBENZ-DBRMD and ITYR-DBRMD, demonstrated attenuated cell counts, induction in apoptosis, and a reduction in cell proliferation in DIO3-positive HGSOC cells (OVCAR3 and KURAMOCHI), but not in DIO3-negative normal ovary cells (CHOK1) and OVCAR3 depleted for DIO3 or its substrate, T3. Potent tumor inhibition with a high safety profile was further established in HGSOC xenograft model, with no effect in DIO3-depleted tumors. The antitumor effects are mediated by downregulation in an array of pro-cancerous proteins, the majority of which known to be repressed by T3. To conclude, using small molecules that specifically target the DIO3 enzyme we present a new treatment paradigm for ovarian cancer and potentially other DIO3-dependent malignancies.
Collapse
|
16
|
Abstract
Deiodinases modify the biological activity of thyroid hormone (TH) molecules, ie, they may activate thyroxine (T4) to 3,5,3'-triiodothyronine (T3), or they may inactivate T3 to 3,3'-diiodo-L-thyronine (T2) or T4 to reverse triiodothyronine (rT3). Although evidence of deiodination of T4 to T3 has been available since the 1950s, objective evidence of TH metabolism was not established until the 1970s. The modern paradigm considers that the deiodinases not only play a role in the homeostasis of circulating T3, but they also provide dynamic control of TH signaling: cells that express the activating type 2 deiodinase (D2) have enhanced TH signaling due to intracellular build-up of T3; the opposite is seen in cells that express type 3 deiodinase (D3), the inactivating deiodinase. D2 and D3 are expressed in metabolically relevant tissues such as brown adipose tissue, skeletal muscle and liver, and their roles have been investigated using cell, animal, and human models. During development, D2 and D3 expression customize for each tissue/organ the timing and intensity of TH signaling. In adult cells, D2 is induced by cyclic adenosine monophosphate (cAMP), and its expression is invariably associated with enhanced T3 signaling, expression of PGC1 and accelerated energy expenditure. In contrast, D3 expression is induced by hypoxia-inducible factor 1α (HIF-1a), dampening T3 signaling and the metabolic rate. The coordinated expression of these enzymes adjusts TH signaling in a time- and tissue-specific fashion, affecting metabolic pathways in health and disease states.
Collapse
Affiliation(s)
- Samuel C Russo
- Section of Endocrinology, Diabetes & Metabolism, University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Federico Salas-Lucia
- Section of Endocrinology, Diabetes & Metabolism, University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Antonio C Bianco
- Section of Endocrinology, Diabetes & Metabolism, University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
17
|
Coscia F, Taler-Verčič A. Cryo-EM: A new dawn in thyroid biology. Mol Cell Endocrinol 2021; 531:111309. [PMID: 33964321 PMCID: PMC8316605 DOI: 10.1016/j.mce.2021.111309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/25/2021] [Accepted: 04/27/2021] [Indexed: 11/09/2022]
Abstract
The thyroid gland accumulates the rare dietary element iodine and incorporates it into iodinated thyroid hormones, utilising several tightly regulated reactions and molecular mechanisms. Thyroid hormones are essential in vertebrates and play a central role in many biological processes, such as development, thermogenesis and growth. The control of these functions is exerted through the binding of hormones to nuclear thyroid hormone receptors that rule the transcription of numerous metabolic genes. Over the last 50 years, thyroid biology has been studied extensively at the cellular and organismal levels, revealing its multiple clinical implications, yet, a complete molecular understanding is still lacking. This includes the atomic structures of crucial pathway components that would be needed to elucidate molecular mechanisms. Here we review the currently known protein structures involved in thyroid hormone synthesis, regulation, transport, and actions. We also highlight targets for future investigations that will significantly benefit from recent advances in macromolecular structure determination by electron cryo-microscopy (cryo-EM). As an example, we demonstrate how cryo-EM was crucial to obtain the structure of the large thyroid hormone precursor protein, thyroglobulin. We discuss modern cryo-EM compared to other structure determination methods and how an integrated structural and cell biological approach will help filling the molecular knowledge gap in our understanding of thyroid hormone metabolism. Together with clinical, cellular and high-throughput 'omics' studies, atomic structures of thyroid components will provide an important framework to map disease mutations and to interpret and predict thyroid phenotypes.
Collapse
Affiliation(s)
- Francesca Coscia
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK; Human Technopole, Via Cristina Belgioioso 171, 20157, Milano, Italy.
| | - Ajda Taler-Verčič
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, Vrazov Trg 2, 1000, Ljubljana, Slovenia
| |
Collapse
|
18
|
Pagnin M, Kondos-Devcic D, Chincarini G, Cumberland A, Richardson SJ, Tolcos M. Role of thyroid hormones in normal and abnormal central nervous system myelination in humans and rodents. Front Neuroendocrinol 2021; 61:100901. [PMID: 33493504 DOI: 10.1016/j.yfrne.2021.100901] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/07/2021] [Accepted: 01/16/2021] [Indexed: 12/13/2022]
Abstract
Thyroid hormones (THs) are instrumental in promoting the molecular mechanisms which underlie the complex nature of neural development and function within the central nervous system (CNS) in vertebrates. The key neurodevelopmental process of myelination is conserved between humans and rodents, of which both experience peak fetal TH concentrations concomitant with onset of myelination. The importance of supplying adequate levels of THs to the myelin producing cells, the oligodendrocytes, for promoting their maturation is crucial for proper neural function. In this review we examine the key TH distributor and transport proteins, including transthyretin (TTR) and monocarboxylate transporter 8 (MCT8), essential for supporting proper oligodendrocyte and myelin health; and discuss disorders with impaired TH signalling in relation to abnormal CNS myelination in humans and rodents. Furthermore, we explore the importance of using novel TH analogues in the treatment of myelination disorders associated with abnormal TH signalling.
Collapse
Affiliation(s)
- Maurice Pagnin
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Delphi Kondos-Devcic
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Ginevra Chincarini
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Angela Cumberland
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | | | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia.
| |
Collapse
|
19
|
Mayasich SA, Korte JJ, Denny JS, Hartig PC, Olker JH, DeGoey P, O'Flanagan J, Degitz SJ, Hornung MW. Xenopus laevis and human type 3 iodothyronine deiodinase enzyme cross-species sensitivity to inhibition by ToxCast chemicals. Toxicol In Vitro 2021; 73:105141. [PMID: 33713820 DOI: 10.1016/j.tiv.2021.105141] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/12/2021] [Accepted: 03/08/2021] [Indexed: 11/15/2022]
Abstract
Deiodinase enzymes are critical for tissue-specific and temporal control of activation or inactivation of thyroid hormones during vertebrate development, including amphibian metamorphosis. We previously screened ToxCast chemicals for inhibitory activity toward human recombinant Type 3 iodothyronine deiodinase enzyme (hDIO3) and subsequently produced Xenopus laevis recombinant dio3 enzyme (Xldio3) with the goals to identify specific chemical inhibitors of Xldio3, to evaluate cross-species sensitivity and explore whether the human assay results are predictive of the amphibian. We identified a subset of 356 chemicals screened against hDIO3 to test against Xldio3, initially at a single concentration (200 μM), and further tested 79 in concentration-response mode. Most chemicals had IC50 values lower for hDIO3 than for Xldio3 and many had steep Hill slopes (a potential indication of non-specific inhibition). However, eight of the most potent chemicals are likely specific inhibitors, with IC50 values of 14 μM or less, Hill slopes near -1 and curves not significantly different between species likely due to conservation of catalytically active amino acids. Controlling for assay conditions, human in vitro screening results can be predictive of activity in the amphibian assay. This study lays the groundwork for future studies using recombinant non-mammalian proteins to test cross-species sensitivity to chemicals. DISCLAIMER: The views expressed in this paper are those of the authors and do not necessarily reflect the views or policies of the U.S. Environmental Protection Agency. Mention of trade names or commercial products does not constitute endorsement or recommendation for use.
Collapse
Affiliation(s)
- Sally A Mayasich
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA; Great Lakes Toxicology and Ecology Division, Center for Computational Toxicology and Ecology, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, USA
| | - Joseph J Korte
- Great Lakes Toxicology and Ecology Division, Center for Computational Toxicology and Ecology, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, USA
| | - Jeffrey S Denny
- Great Lakes Toxicology and Ecology Division, Center for Computational Toxicology and Ecology, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, USA
| | - Phillip C Hartig
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Jennifer H Olker
- Great Lakes Toxicology and Ecology Division, Center for Computational Toxicology and Ecology, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, USA
| | - Philip DeGoey
- Great Lakes Toxicology and Ecology Division, Center for Computational Toxicology and Ecology, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, USA
| | - Joseph O'Flanagan
- Great Lakes Toxicology and Ecology Division, Center for Computational Toxicology and Ecology, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, USA; Oak Ridge Associated Universities, Oak Ridge, TN, USA
| | - Sigmund J Degitz
- Great Lakes Toxicology and Ecology Division, Center for Computational Toxicology and Ecology, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, USA
| | - Michael W Hornung
- Great Lakes Toxicology and Ecology Division, Center for Computational Toxicology and Ecology, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, USA.
| |
Collapse
|
20
|
França MM, German A, Fernandes GW, Liao XH, Bianco AC, Refetoff S, Dumitrescu AM. Human Type 1 Iodothyronine Deiodinase ( DIO1) Mutations Cause Abnormal Thyroid Hormone Metabolism. Thyroid 2021; 31:202-207. [PMID: 32718224 PMCID: PMC7891200 DOI: 10.1089/thy.2020.0253] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Iodothyronine deiodinase-1 (D1) selenoenzyme regulates the systemic supply of active thyroid hormone (TH). Transient decrease in D1 enzymatic activity is clinically relevant and adaptive in nonthyroidal illness such as fasting or acute illness. However, DIO1 gene defects have not been reported in humans. Methods: Genetic analysis was performed using whole-exome sequencing in members of two unrelated families presenting with abnormal serum thyroid function tests. Plasmid constructs containing the two pathogenic DIO1 variants were used for in vitro studies assessing the kinetics of their enzymatic activity. Thyroid function tests were measured in Dio1 heterozygous-null mice. Results: We report the novel identification and characterization of two missense DIO1 pathogenic variants (resulting in p.Asn94Lys and p.Met201Ile) in two unrelated families presenting with abnormal TH metabolism with elevated serum reverse triiodothyronine (rT3) levels and rT3/T3 ratios. These characteristic in vivo parameters are also present in Dio1 heterozygous-null mice. Kinetic studies of the resulting mutant D1 proteins demonstrate two- to threefold higher Km indicating lower substrate affinity and slower enzyme velocity. Conclusions: We report the identification and characterization of two missense DIO1 pathogenic variants identified in families with abnormal TH metabolism. This is the first demonstration of inherited D1 deficiency in humans.
Collapse
Affiliation(s)
- Monica M. França
- Section of Endocrinology Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Alina German
- Department of Pediatric Endocrine Unit, Bnei Zion Medical Center, Clalit Health Service and Technion-the Israel Institute of Technology, Haifa, Israel
| | - Gustavo W. Fernandes
- Section of Endocrinology Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Xiao-Hui Liao
- Section of Endocrinology Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Antonio C. Bianco
- Section of Endocrinology Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Samuel Refetoff
- Section of Endocrinology Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
- Department of Pediatrics and Committees, The University of Chicago, Chicago, Illinois, USA
- Genetics, The University of Chicago, Chicago, Illinois, USA
| | - Alexandra M. Dumitrescu
- Section of Endocrinology Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
- Molecular Metabolism and Nutrition, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
21
|
Common modifications of selenocysteine in selenoproteins. Essays Biochem 2020; 64:45-53. [PMID: 31867620 DOI: 10.1042/ebc20190051] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/09/2019] [Accepted: 12/12/2019] [Indexed: 01/03/2023]
Abstract
Selenocysteine (Sec), the sulfur-to-selenium substituted variant of cysteine (Cys), is the defining entity of selenoproteins. These are naturally expressed in many diverse organisms and constitute a unique class of proteins. As a result of the physicochemical characteristics of selenium when compared with sulfur, Sec is typically more reactive than Cys while participating in similar reactions, and there are also some qualitative differences in the reactivities between the two amino acids. This minireview discusses the types of modifications of Sec in selenoproteins that have thus far been experimentally validated. These modifications include direct covalent binding through the Se atom of Sec to other chalcogen atoms (S, O and Se) as present in redox active molecular motifs, derivatization of Sec via the direct covalent binding to non-chalcogen elements (Ni, Mb, N, Au and C), and the loss of Se from Sec resulting in formation of dehydroalanine. To understand the nature of these Sec modifications is crucial for an understanding of selenoprotein reactivities in biological, physiological and pathophysiological contexts.
Collapse
|
22
|
Moskovich D, Alfandari A, Finkelshtein Y, Weisz A, Katzav A, Kidron D, Edelstein E, Veroslavski D, Perets R, Arbib N, Kadan Y, Fishman A, Lerer B, Ellis M, Ashur-Fabian O. DIO3, the thyroid hormone inactivating enzyme, promotes tumorigenesis and metabolic reprogramming in high grade serous ovarian cancer. Cancer Lett 2020; 501:224-233. [PMID: 33221455 DOI: 10.1016/j.canlet.2020.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/18/2020] [Accepted: 11/11/2020] [Indexed: 12/27/2022]
Abstract
High grade serous ovarian cancer (HGSOC) is the most lethal gynecologic malignancy with a need for better understanding the disease pathogenesis. The biologically active thyroid hormone, T3, is considered a tumor suppressor by promoting cell differentiation and mitochondrial respiration. Tumors evolved a strategy to avoid these anticancer actions by expressing the T3 catabolizing enzyme, Deiodinase type 3 (DIO3). This stimulates cancer proliferation and aerobic glycolysis (Warburg effect). We identified DIO3 expression in HGSOC cell lines, tumor tissues from mice and human patients, fallopian tube (FT) premalignant lesion and secretory cells of normal FT, considered the disease site-of-origin. Stable DIO3 knockdown (DIO3-KD) in HGSOC cells led to increased T3 bioavailability and demonstrated induced apoptosis and attenuated proliferation, migration, colony formation, oncogenic signaling, Warburg effect and tumor growth in mice. Proteomics analysis further indicated alterations in an array of cancer-relevant proteins, the majority of which are involved in tumor suppression and metabolism. Collectively this study establishes the functional role of DIO3 in facilitating tumorigenesis and metabolic reprogramming, and proposes this enzyme as a promising target for inhibition in HGSOC.
Collapse
Affiliation(s)
- Dotan Moskovich
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Alfandari
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Finkelshtein
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avivit Weisz
- Department of Pathology, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Israel
| | - Aviva Katzav
- Department of Pathology, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Israel
| | - Debora Kidron
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Pathology, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Israel
| | - Evgeny Edelstein
- Department of Pathology, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Israel
| | - Daniel Veroslavski
- Clinical Research Institute at Rambam, Division of Oncology, Rambam Health Care Campus, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ruth Perets
- Clinical Research Institute at Rambam, Division of Oncology, Rambam Health Care Campus, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nissim Arbib
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Gynecological Oncology Unit, The Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel
| | - Yfat Kadan
- Gynecological Oncology Unit, The Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel
| | - Ami Fishman
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Gynecological Oncology Unit, The Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel
| | - Bernard Lerer
- Biological Psychiatry Laboratory Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Martin Ellis
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Ashur-Fabian
- Translational Oncology Laboratory, Hematology Institute and Blood Bank, Meir Medical Center, Kfar-Saba, Israel; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
23
|
Extension of the taxonomic coverage of the family GH126 outside Firmicutes and in silico characterization of its non-catalytic terminal domains. 3 Biotech 2020; 10:420. [PMID: 32953382 PMCID: PMC7479077 DOI: 10.1007/s13205-020-02415-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 08/27/2020] [Indexed: 01/01/2023] Open
Abstract
The family GH126 is a family of glycoside hydrolases established in 2011. Officially, in the CAZy database, it counts ~ 1000 sequences originating solely from bacterial phylum Firmicutes. Two members, the proteins CPF_2247 from Clostridium perfringens and PssZ from Listeria monocytogenes have been characterized as a probable α-amylase and an exopolysaccharide-specific glycosidase, respectively; their three-dimensional structures being also solved as possessing catalytic (α/α)6-barrel fold. Previously, based on a detailed in silico analysis, the seven conserved sequence regions (CSRs) were identified for the family along with elucidating basic evolutionary relationships within the family members. The present study represents a continuation study focusing on two particular aims: (1) to find out whether the taxonomic coverage of the family GH126 might be extended outside the Firmicutes and, if positive, to deliver those out-of-Firmicutes proteins with putting them into the context of the family; and (2) to identify the family members containing the N- and/or C-terminal extensions of their polypeptide chain, additional to the catalytic (α/α)6-barrel domain, and perform the bioinformatics characterization of the extra domains. The main results could be summarized as follows: (1) 17 bacterial proteins caught by BLAST searches outside Firmicutes (especially from phyla Proteobacteria, Actinobacteria and Bacteroidetes) have been found and convincingly suggested as new family GH126 members; and (2) a thioredoxin-like fold and various leucine-rich repeat motifs identified by Phyre2 structure homology modelling have been recognized as extra domains occurring most frequently in the N-terminal extensions of family GH126 members possessing a modular organization.
Collapse
|
24
|
Abstract
Iodothyronine deiodinases (Dios) are important selenoproteins that control the concentration of the active thyroid hormone (TH) triiodothyronine through regioselective deiodination. The X-ray structure of a truncated monomer of Type III Dio (Dio3), which deiodinates TH inner rings through a selenocysteine (Sec) residue, revealed a thioredoxin-fold catalytic domain supplemented with an unstructured Ω-loop. Loop dynamics are driven by interactions of the conserved Trp207 with solvent in multi-microsecond molecular dynamics simulations of the Dio3 thioredoxin(Trx)-fold domain. Hydrogen bonding interactions of Glu200 with residues conserved across the Dio family anchor the loop’s N-terminus to the active site Ser-Cys-Thr-Sec sequence. A key long-lived loop conformation coincides with the opening of a cryptic pocket that accommodates thyroxine (T4) through an I⋯Se halogen bond to Sec170 and the amino acid group with a polar cleft. The Dio3-T4 complex is stabilized by an I⋯O halogen bond between an outer ring iodine and Asp211, consistent with Dio3 selectivity for inner ring deiodination. Non-conservation of residues, such as Asp211, in other Dio types in the flexible portion of the loop sequence suggests a mechanism for regioselectivity through Dio type-specific loop conformations. Cys168 is proposed to attack the selenenyl iodide intermediate to regenerate Dio3 based upon structural comparison with related Trx-fold proteins.
Collapse
|
25
|
A detailed in silico analysis of the amylolytic family GH126 and its possible relatedness to family GH76. Carbohydr Res 2020; 494:108082. [PMID: 32634753 DOI: 10.1016/j.carres.2020.108082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/13/2020] [Accepted: 06/14/2020] [Indexed: 11/21/2022]
Abstract
The glycoside hydrolase (GH) family 126 was established based on the X-ray structure determination of the amylolytic enzyme CPF_2247 from Clostridium perfringens genome. Its original identification as a putative carbohydrate-active enzyme was based on its low, yet significant sequence identity to members of the family GH8, which are inverting endo-β-1,4-glucanases. As the family GH8 forms the clan GH-M with GH48, the CPF_2247 protein also exhibits similarities with members of the family GH48. The original screening of the CPF_2247 on carbohydrate substrates demonstrated its activity on glycogen and amylose, thus classifying this protein as an "α-amylase". It should be pointed out, however, there are apparent inconsistencies concerning the exact enzyme specificity of the "amylase" CPF_2247, since it exhibits both the endo- and exo-fashion of action. The family GH126 currently counts ~1000 amino acid sequences solely from Bacteria; all belonging to the phylum Firmicutes. The present study delivers the first detailed bioinformatics study of 117 selected amino acid sequences from the family GH126, featuring the insightful sequence-structure comparison with the aim to define seven conserved sequence regions and elucidate the evolutionary relationships within the family. In addition, a comparative structural analysis of the GH126 members with representatives of other GH families adopting the same (α/α)6-barrel catalytic domain fold indicates the possible sharing a catalytic residue between the families GH126 and GH76.
Collapse
|
26
|
Stanishevska NV. Selenoproteins and their emerging roles in signaling pathways. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The functional activity of selenoproteins has a wide range of effects on complex pathogenetic processes, including teratogenesis, immuno-inflammatory, neurodegenerative. Being active participants and promoters of many signaling pathways, selenoproteins support the lively interest of a wide scientific community. This review is devoted to the analysis of recent data describing the participation of selenoproteins in various molecular interactions mediating important signaling pathways. Data processing was carried out by the method of complex analysis. For convenience, all selenoproteins were divided into groups depending on their location and function. Among the group of selenoproteins of the ER membrane, selenoprotein N affects the absorption of Ca2+ by the endoplasmic reticulum mediated by oxidoreductin (ERO1), a key player in the CHOP/ERO1 branch, a pathogenic mechanism that causes myopathy. Another selenoprotein of the ER membrane selenoprotein K binding to the DHHC6 protein affects the IP3R receptor that regulates Ca2+ flux. Selenoprotein K is able to affect another protein of the endoplasmic reticulum CHERP, also appearing in Ca2+ transport. Selenoprotein S, associated with the lumen of ER, is able to influence the VCP protein, which ensures the incorporation of selenoprotein K into the ER membrane. Selenoprotein M, as an ER lumen protein, affects the phosphorylation of STAT3 by leptin, which confirms that Sel M is a positive regulator of leptin signaling. Selenoprotein S also related to luminal selenoproteins ER is a modulator of the IRE1α-sXBP1 signaling pathway. Nuclear selenoprotein H will directly affect the suppressor of malignant tumours, p53 protein, the activation of which increases with Sel H deficiency. The same selenoprotein is involved in redox regulation. Among the cytoplasmic selenoproteins, abundant investigations are devoted to SelP, which affects the PI3K/Akt/Erk signaling pathway during ischemia/reperfusion, is transported into the myoblasts through the plasmalemma after binding to the apoER2 receptor, and into the neurons to the megaline receptor and in general, selenoprotein P plays the role of a pool that stores the necessary trace element and releases it, if necessary, for vital selenoproteins. The thioredoxin reductase family plays a key role in the invasion and metastasis of salivary adenoid cystic carcinoma through the influence on the TGF-β-Akt/GSK-3β pathway during epithelial-mesenchymal transition. The deletion of thioredoxin reductase 1 affects the levels of messengers of the Wnt/β-catenin signaling pathway. No less studied is the glutathione peroxidase group, of which GPX3 is able to inhibit signaling in the Wnt/β-catenin pathway and thereby inhibit thyroid metastasis, as well as suppress protein levels in the PI3K/Akt/c-fos pathway. A key observation is that in cases of carcinogenesis, a decrease in GPX3 and its hypermethylation are almost always found. Among deiodinases, deiodinase 3 acts as a promoter of the oncogenes BRAF, MEK or p38, while stimulating a decrease in the expression of cyclin D1. The dependence of the level of deiodinase 3 on the Hedgehog (SHH) signaling pathway is also noted. Methionine sulfoxide reductase A can compete for the uptake of ubiquitin, reduce p38, JNK and ERK promoters of the MAPK signaling pathway; methionine sulfoxide reductase B1 suppresses MAPK signaling messengers, and also increases PARP and caspase 3.
Collapse
|
27
|
Marsan ES, Bayse CA. Halogen Bonding Interactions of Polychlorinated Biphenyls and the Potential for Thyroid Disruption. Chemistry 2020; 26:5200-5207. [PMID: 31849117 PMCID: PMC8812442 DOI: 10.1002/chem.201903904] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/13/2019] [Indexed: 11/21/2023]
Abstract
Polychlorinated biphenyl (PCB) flame retardants are persistent pollutants and inhibit neurodevelopment, particularly in the early stages of life. Halogen bonding (XB) to the iodothyronine deiodinases (Dio) that modulate thyroid hormones (THs) is a potential mechanism for endocrine disruption. Cl⋅⋅⋅Se XB interactions of PCBs with SeMe- , a small model of the Dio active site selenocysteine, are compared with previous results on polybrominated diphenylethers (PBDEs) and THs using density functional theory. PCBs generally display weaker XB interactions compared to PBDEs and THs, consistent with the dependence of XB strength on the size of the halogen (I>Br>Cl). PCBs also do not meet a proposed energy threshold for substrates to undergo dehalogenation, suggesting they may behave as competitive inhibitors of Dio in addition to other mechanisms of endocrine disruption. XB interactions in PCBs are position-dependent, with ortho interactions slightly more favorable than meta and para interactions, suggesting that PCBs may have a greater effect on certain classes of Dio. Flexibility of PCBs around the biphenyl C-C bond is limited by ortho substitutions relative to the biphenyl linkage, which may contribute to the ability to inhibit Dio and other TH-related proteins.
Collapse
Affiliation(s)
- Eric S Marsan
- Department of Chemistry and Biochemistry, Old Dominion University, 1 Old Dominion University, Norfolk, VA, 23529, USA
| | - Craig A Bayse
- Department of Chemistry and Biochemistry, Old Dominion University, 1 Old Dominion University, Norfolk, VA, 23529, USA
| |
Collapse
|
28
|
Groeneweg S, van Geest FS, Peeters RP, Heuer H, Visser WE. Thyroid Hormone Transporters. Endocr Rev 2020; 41:5637505. [PMID: 31754699 DOI: 10.1210/endrev/bnz008] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ferdy S van Geest
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - W Edward Visser
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
29
|
A Halogen Bonding Perspective on Iodothyronine Deiodinase Activity. Molecules 2020; 25:molecules25061328. [PMID: 32183289 PMCID: PMC7144113 DOI: 10.3390/molecules25061328] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 12/25/2022] Open
Abstract
Iodothyronine deiodinases (Dios) are involved in the regioselective removal of iodine from thyroid hormones (THs). Deiodination is essential to maintain TH homeostasis, and disruption can have detrimental effects. Halogen bonding (XB) to the selenium of the selenocysteine (Sec) residue in the Dio active site has been proposed to contribute to the mechanism for iodine removal. Polybrominated diphenyl ethers (PBDEs) and polychlorinated biphenyls (PCBs) are known disruptors of various pathways of the endocrine system. Experimental evidence shows PBDEs and their hydroxylated metabolites (OH-BDEs) can inhibit Dio, while data regarding PCB inhibition are limited. These xenobiotics could inhibit Dio activity by competitively binding to the active site Sec through XB to prevent deiodination. XB interactions calculated using density functional theory (DFT) of THs, PBDEs, and PCBs to a methyl selenolate (MeSe−) arrange XB strengths in the order THs > PBDEs > PCBs in agreement with known XB trends. THs have the lowest energy C–X*-type unoccupied orbitals and overlap with the Se lp donor leads to high donor-acceptor energies and the greatest activation of the C–X bond. The higher energy C–Br* and C–Cl* orbitals similarly result in weaker donor-acceptor complexes and less activation of the C–X bond. Comparison of the I···Se interactions for the TH group suggest that a threshold XB strength may be required for dehalogenation. Only highly brominated PBDEs have binding energies in the same range as THs, suggesting that these compounds may inhibit Dio and undergo debromination. While these small models provide insight on the I···Se XB interaction itself, interactions with other active site residues are governed by regioselective preferences observed in Dios.
Collapse
|
30
|
Mondal S, Manna D, Raja K, Mugesh G. Halogen Bonding in Biomimetic Deiodination of Thyroid Hormones and their Metabolites and Dehalogenation of Halogenated Nucleosides. Chembiochem 2020; 21:911-923. [PMID: 31773854 DOI: 10.1002/cbic.201900619] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Indexed: 12/14/2022]
Abstract
Thyroid hormones (THs) are key players in the endocrine system and play pivotal roles in carbohydrate and fat metabolism, protein synthesis, overall growth, and brain development. The thyroid gland predominantly produces thyroxine or 3,5,3',5'-tetraiodothyronine (T4) as a prohormone; three isoforms of a mammalian selenoenzyme-iodothyronine deiodinase (DIO1, DIO2 and DIO3)-catalyze the regioselective deiodination of T4 to produce biologically active and inactive metabolites. Whereas DIO1 catalyzes both 5- and 5'-deiodination of T4, DIO2 and DIO3 selectively mediate 5- and 5'-deiodination, respectively. In this review we discuss the regioselective deiodination of THs in the presence of organochalcogen compounds. Naphthalene-based compounds containing sulfur and/or selenium at the peri positions mediate regioselective 5-deiodination of THs, detailed mechanistic studies having revealed that the heterolytic cleavage of the C-I bond is facilitated by the formation of cooperative Se/S⋅⋅⋅I halogen bonds and Se/S⋅⋅⋅Se chalcogen bonds. We also discuss the biomimetic deiodination of several TH metabolites, including sulfated THs, iodothyronamines, and iodotyrosines. A brief discussion on the dehalogenation of halogenated nucleosides and nucleobases in the presence of organochalcogen compounds is also included.
Collapse
Affiliation(s)
- Santanu Mondal
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Debasish Manna
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Karuppusamy Raja
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
31
|
Kolanu BR, Vadakedath S, Boddula V, Kandi V. Activities of Serum Magnesium and Thyroid Hormones in Pre-, Peri-, and Post-menopausal Women. Cureus 2020; 12:e6554. [PMID: 32042527 PMCID: PMC6996468 DOI: 10.7759/cureus.6554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/03/2020] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION Females go through a complex hormonal variation once they reach menarche. The menstrual cycle repeats every month regularly and is dependent on the normal functioning of the hypothalamus, pituitary, and ovarian hormones. The overall wellness of the females during the menstrual cycle depends greatly on nutritional status. It is common that women develop menstrual cycle-related symptoms and are routinely prone to thyroid dysfunction. The present study is carried out to assess the activities of Mg and thyroid hormones in pre-, peri-, and post-menopausal women. METHODS A total of 165 women were recruited in the study after satisfying the inclusion criteria. An equal number of age-matched subjects were included as controls. All the subjects included in the study were selected from the patients attending various out-patient departments of the Prathima Institute of Medical Sciences, Karimnagar, Telangana, India. Blood samples from each subject were collected and analyzed by a semi-automated analyzer for the activities of Mg, and thyroid hormones tetra-iodothyronine (T4), tri-iodothyronine (T3), and thyroid-stimulating hormone (TSH). RESULTS There was a statistically significant relationship between the serum Mg activities and the thyroid hormones between the study subjects and the control group. The activities of the serum Mg (1.72±0.33) in relation to the TSH (5.09±7.54) in the cases were found statistically significant (p <0.001) when compared to the serum Mg (1.8±0.20) in relation to the TSH in the control group (2.41±2.05). The activities of Mg were noted to fall in women through the peri (1.70±0.43), and postmenopausal age (1.60±0.34). There was a significant increase in the activities of TSH in women of premenopause (4.27±5.76), perimenopause (5.65±8.53), and postmenopausal age (7.19±11.07). Conclusion: From the results of the present study, it can be concluded that the women reaching menopause could suffer from hypomagnesemia and inturn may develop thyroid and other hormonal disorders.
Collapse
Affiliation(s)
| | | | - Venugopal Boddula
- Biochemistry, Prathima Institute of Medical Sciences, Karimnagar, IND
| | - Venkataramana Kandi
- Clinical Microbiology, Prathima Institute of Medical Sciences, Karimnagar, IND
| |
Collapse
|
32
|
Uterine Inertia due to Severe Selenium Deficiency in a Parturient Mare. J Equine Vet Sci 2019; 85:102845. [PMID: 31952635 DOI: 10.1016/j.jevs.2019.102845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/09/2019] [Accepted: 11/08/2019] [Indexed: 11/22/2022]
Abstract
A 12-year-old, multiparous, parturient show jumper embryo-recipient mare presented at a veterinary hospital, seven days past her due date and with a dilated cervix, for evaluation of mild colic. Gastrointestinal or metabolic abnormalities and fetal maldispositions were excluded as causes of dystocia, and a diagnosis of uterine inertia was made. There was no uterine response to oxytocin treatment. A live filly was delivered via C-section, and severe selenium deficiency was eventually confirmed in the mare, her offspring, and in the herd of origin. The filly was born with severe white muscle disease and required intensive treatment. This report suggests that selenium deficiency is an underlying cause of equine uterine inertia in the absence of other causes of dystocia.
Collapse
|
33
|
Steegborn C, Schweizer U. Structure and Mechanism of Iodothyronine Deiodinases - What We Know, What We Don't Know, and What Would Be Nice to Know. Exp Clin Endocrinol Diabetes 2019; 128:375-378. [PMID: 31698481 DOI: 10.1055/a-1022-9916] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Deiodinases catalyze the specific removal of iodine atoms from one of the two iodinated phenyl rings in iodothyronines. They thereby fine-regulate local thyroid hormone concentrations in organs or cells. The chemical reaction is unique in the sense that in metazoans the reductive elimination of iodide depends on the rare amino acid selenocysteine in the enzymes' active centers. While there is no prokaryotic homologue of such deiodinases, the solution of the crystal structure of a catalytic domain of mouse deiodinase 3 has revealed that the ancient peroxiredoxin structure has been repurposed, and improved using selenocysteine, as a deiodinase during metazoan evolution. Likewise, many biochemical findings obtained over decades can now be interpreted in light of the molecular structure. Despite this leap in our understanding of deiodinase structure, there are still several open questions that need to be addressed in order to fully understand substrate binding, catalytic mechanism, and regulation of deiodinases. We surmise that these issues as well as differences between the three highly homologous isoenzymes must be understood in order to develop modulators of deiodinases that could be valuable in clinical use.
Collapse
Affiliation(s)
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
34
|
Bianco AC, Dumitrescu A, Gereben B, Ribeiro MO, Fonseca TL, Fernandes GW, Bocco BMLC. Paradigms of Dynamic Control of Thyroid Hormone Signaling. Endocr Rev 2019; 40:1000-1047. [PMID: 31033998 PMCID: PMC6596318 DOI: 10.1210/er.2018-00275] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/15/2019] [Indexed: 12/17/2022]
Abstract
Thyroid hormone (TH) molecules enter cells via membrane transporters and, depending on the cell type, can be activated (i.e., T4 to T3 conversion) or inactivated (i.e., T3 to 3,3'-diiodo-l-thyronine or T4 to reverse T3 conversion). These reactions are catalyzed by the deiodinases. The biologically active hormone, T3, eventually binds to intracellular TH receptors (TRs), TRα and TRβ, and initiate TH signaling, that is, regulation of target genes and other metabolic pathways. At least three families of transmembrane transporters, MCT, OATP, and LAT, facilitate the entry of TH into cells, which follow the gradient of free hormone between the extracellular fluid and the cytoplasm. Inactivation or marked downregulation of TH transporters can dampen TH signaling. At the same time, dynamic modifications in the expression or activity of TRs and transcriptional coregulators can affect positively or negatively the intensity of TH signaling. However, the deiodinases are the element that provides greatest amplitude in dynamic control of TH signaling. Cells that express the activating deiodinase DIO2 can rapidly enhance TH signaling due to intracellular buildup of T3. In contrast, TH signaling is dampened in cells that express the inactivating deiodinase DIO3. This explains how THs can regulate pathways in development, metabolism, and growth, despite rather stable levels in the circulation. As a consequence, TH signaling is unique for each cell (tissue or organ), depending on circulating TH levels and on the exclusive blend of transporters, deiodinases, and TRs present in each cell. In this review we explore the key mechanisms underlying customization of TH signaling during development, in health and in disease states.
Collapse
Affiliation(s)
- Antonio C Bianco
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| | - Alexandra Dumitrescu
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Miriam O Ribeiro
- Developmental Disorders Program, Center of Biologic Sciences and Health, Mackenzie Presbyterian University, São Paulo, São Paulo, Brazil
| | - Tatiana L Fonseca
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| | - Gustavo W Fernandes
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| | - Barbara M L C Bocco
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago Medical Center, Chicago, Illinois
| |
Collapse
|
35
|
Köhrle J. The Colorful Diversity of Thyroid Hormone Metabolites. Eur Thyroid J 2019; 8:115-129. [PMID: 31259154 PMCID: PMC6587369 DOI: 10.1159/000497141] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/22/2019] [Indexed: 12/17/2022] Open
Abstract
Since the discovery of L-thyroxine, the main secretory product of the thyroid gland, and its major metabolite T3, which exerts the majority of thyroid hormone action via ligand-dependent modulation of the function of T3 receptors in nuclei, mitochondria, and other subcellular compartments, various other T4-derived endogenous metabolites have been identified in blood and tissues of humans, animals, and early protochordates. This review addresses major historical milestones and experimental findings resulting in the discovery of the key enzymes of thyroid hormone metabolism, the three selenoprotein deiodinases, as well as the decarboxylases and amine oxidases involved in formation and degradation of recently identified endogenous thyroid hormone metabolites, i.e. 3-iodothyronamine and 3-thyroacetic acid. The concerted action of deiodinases 2 and 3 in regulation of local T3 availability is discussed. Special attention is given to the role of the thyromimetic "hot" metabolite 3,5-T2 and the "cool" 3-iodothyronamine, especially after administration of pharmacological doses of these endogenous thyroid hormone metabolites in various animal experimental models. In addition, available information on the biological roles of the two major acetic acid derivatives of thyroid hormones, i.e. Tetrac and Triac, as well as sulfated metabolites of thyroid hormones is reviewed. This review addresses the consequences of the existence of this broad spectrum of endogenous thyroid hormone metabolites, the "thyronome," beyond the classical thyroid hormone profile comprising T4, T3, and rT3 for appropriate analytical coverage and clinical diagnostics using mass spectrometry versus immunoassays for determination of total and free concentrations of thyroid hormone metabolites in blood and tissues.
Collapse
Affiliation(s)
- Josef Köhrle
- Institut für Experimentelle Endokrinologie, Charité Campus Virchow-Klinikum (CVK), Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
36
|
Chekan JR, Lee GY, El Gamal A, Purdy TN, Houk KN, Moore BS. Bacterial Tetrabromopyrrole Debrominase Shares a Reductive Dehalogenation Strategy with Human Thyroid Deiodinase. Biochemistry 2019; 58:5329-5338. [PMID: 31117392 DOI: 10.1021/acs.biochem.9b00318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Enzymatic dehalogenation is an important and well-studied biological process in both the detoxification and catabolism of small molecules, many of which are anthropogenic in origin. However, dedicated dehalogenation reactions that replace a halogen atom with a hydrogen are rare in the biosynthesis of natural products. In fact, the debrominase Bmp8 is the only known example. It catalyzes the reductive debromination of the coral settlement cue and the potential human toxin 2,3,4,5-tetrabromopyrrole as part of the biosynthesis of the antibiotic pentabromopseudilin. Using a combination of protein crystallography, mutagenesis, and computational modeling, we propose a catalytic mechanism for Bmp8 that is reminiscent of that catalyzed by human deiodinases in the maintenance of thyroid hormones. The identification of the key catalytic residues enabled us to recognize divergent functional homologues of Bmp8. Characterization of one of these homologues demonstrated its debromination activity even though it is found in a completely distinct genomic context. This observation suggests that additional enzymes outside those associated with the tetrabromopyrrole biosynthetic pathway may be able to alter the lifetime of this compound in the environment.
Collapse
Affiliation(s)
- Jonathan R Chekan
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography , University of California San Diego , La Jolla , California 92093 , United States
| | - Ga Young Lee
- Department of Chemistry and Biochemistry , University of California , Los Angeles , California 90095 , United States
| | - Abrahim El Gamal
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography , University of California San Diego , La Jolla , California 92093 , United States
| | - Trevor N Purdy
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography , University of California San Diego , La Jolla , California 92093 , United States
| | - K N Houk
- Department of Chemistry and Biochemistry , University of California , Los Angeles , California 90095 , United States
| | - Bradley S Moore
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography , University of California San Diego , La Jolla , California 92093 , United States.,Skaggs School of Pharmacy and Pharmaceutical Sciences , University of California San Diego , La Jolla , California 92093 , United States
| |
Collapse
|
37
|
Jo S, Fonseca TL, Bocco BMLC, Fernandes GW, McAninch EA, Bolin AP, Da Conceição RR, Werneck-de-Castro JP, Ignacio DL, Egri P, Németh D, Fekete C, Bernardi MM, Leitch VD, Mannan NS, Curry KF, Butterfield NC, Bassett JD, Williams GR, Gereben B, Ribeiro MO, Bianco AC. Type 2 deiodinase polymorphism causes ER stress and hypothyroidism in the brain. J Clin Invest 2019; 129:230-245. [PMID: 30352046 PMCID: PMC6307951 DOI: 10.1172/jci123176] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/11/2018] [Indexed: 12/31/2022] Open
Abstract
Levothyroxine (LT4) is a form of thyroid hormone used to treat hypothyroidism. In the brain, T4 is converted to the active form T3 by type 2 deiodinase (D2). Thus, it is intriguing that carriers of the Thr92Ala polymorphism in the D2 gene (DIO2) exhibit clinical improvement when liothyronine (LT3) is added to LT4 therapy. Here, we report that D2 is a cargo protein in ER Golgi intermediary compartment (ERGIC) vesicles, recycling between ER and Golgi. The Thr92-to-Ala substitution (Ala92-D2) caused ER stress and activated the unfolded protein response (UPR). Ala92-D2 accumulated in the trans-Golgi and generated less T3, which was restored by eliminating ER stress with the chemical chaperone 4-phenyl butyric acid (4-PBA). An Ala92-Dio2 polymorphism-carrying mouse exhibited UPR and hypothyroidism in distinct brain areas. The mouse refrained from physical activity, slept more, and required additional time to memorize objects. Enhancing T3 signaling in the brain with LT3 improved cognition, whereas restoring proteostasis with 4-PBA eliminated the Ala92-Dio2 phenotype. In contrast, primary hypothyroidism intensified the Ala92-Dio2 phenotype, with only partial response to LT4 therapy. Disruption of cellular proteostasis and reduced Ala92-D2 activity may explain the failure of LT4 therapy in carriers of Thr92Ala-DIO2.
Collapse
Affiliation(s)
- Sungro Jo
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
| | - Tatiana L. Fonseca
- Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Barbara M. L. C. Bocco
- Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Gustavo W. Fernandes
- Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Elizabeth A. McAninch
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
| | - Anaysa P. Bolin
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
- Department of Pharmacology, Biomedical Science Institute, University of São Paulo, and
| | - Rodrigo R. Da Conceição
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
- Laboratory of Molecular and Translational Endocrinology, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | | | - Daniele L. Ignacio
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
| | - Péter Egri
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Dorottya Németh
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Csaba Fekete
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Maria Martha Bernardi
- Graduate Program of Environmental and Experimental Pathology, Graduate Program of Dentistry, Universidade Paulista, São Paulo, SP, Brazil
| | - Victoria D. Leitch
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Naila S. Mannan
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Katharine F. Curry
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Natalie C. Butterfield
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - J.H. Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, United Kingdom
| | - Balázs Gereben
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Miriam O. Ribeiro
- Developmental Disorders Program, Center of Biological Science and Health, Mackenzie Presbyterian University, São Paulo, SP, Brazil
| | - Antonio C. Bianco
- Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
38
|
Goemann IM, Marczyk VR, Romitti M, Wajner SM, Maia AL. Current concepts and challenges to unravel the role of iodothyronine deiodinases in human neoplasias. Endocr Relat Cancer 2018; 25:R625-R645. [PMID: 30400023 DOI: 10.1530/erc-18-0097] [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: 02/26/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Thyroid hormones (THs) are essential for the regulation of several metabolic processes and the energy consumption of the organism. Their action is exerted primarily through interaction with nuclear receptors controlling the transcription of thyroid hormone-responsive genes. Proper regulation of TH levels in different tissues is extremely important for the equilibrium between normal cellular proliferation and differentiation. The iodothyronine deiodinases types 1, 2 and 3 are key enzymes that perform activation and inactivation of THs, thus controlling TH homeostasis in a cell-specific manner. As THs seem to exert their effects in all hallmarks of the neoplastic process, dysregulation of deiodinases in the tumoral context can be critical to the neoplastic development. Here, we aim at reviewing the deiodinases expression in different neoplasias and exploit the mechanisms by which they play an essential role in human carcinogenesis. TH modulation by deiodinases and other classical pathways may represent important targets with the potential to oppose the neoplastic process.
Collapse
Affiliation(s)
- Iuri Martin Goemann
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Vicente Rodrigues Marczyk
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Mirian Romitti
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Brussels, Belgium
| | - Simone Magagnin Wajner
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Luiza Maia
- Thyroid Unit, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
39
|
Bayse CA. Halogen Bonding from the Bonding Perspective with Considerations for Mechanisms of Thyroid Hormone Activation and Inhibition. NEW J CHEM 2018; 42:10623-10632. [PMID: 30778278 PMCID: PMC6376990 DOI: 10.1039/c8nj00557e] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Halogen bonding interactions are often discussed in terms of an area of positive electrostatic potential on the halogen center along the bond axis called the σ-hole, yet various authors have noted a lack of completeness in this model. The nature of the XB interaction is explored from the perspective of bonding theories beginning from models that explain the electrostatic σ-hole and continuing to orbital-based donor-acceptor descriptions in which the donor lone pair MO mixes with the acceptor R-X and R-X* MOs to form a set of XB MOs related to three-center-four-electron bonding in hypervalent molecules. The strength of the XB interaction for a large series of RX···Cl- and RX···SeMe2 complexes correlate well with the energy of the acceptor R-X* MO and the contribution of the halide to the R-X and R-X* MOs, factors relevant to favourable overlap with the donor lone pair. An orbital-based focus accounts for the partial covalency of the XB interaction and can be extended to descriptions of enzymatic dehalogenation mechanisms. Applications of this MO perspective to the deiodination of thyroid hormones by the iodothyronine deiodinases and a possibly related mechanism of inhibition are discussed.
Collapse
Affiliation(s)
- Craig A Bayse
- Department of Chemistry and Biochemistry, Old Dominion University, Hampton Boulevard, Norfolk, Virginia, United States. ; Tel: 01 757 683 4097;
| |
Collapse
|
40
|
Valea A, Georgescu CE. Selenoproteins in human body: focus on thyroid pathophysiology. Hormones (Athens) 2018; 17:183-196. [PMID: 29873029 DOI: 10.1007/s42000-018-0033-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 04/18/2018] [Indexed: 02/07/2023]
Abstract
Selenium (Se) has a multilevel, complex and dynamic effect on the human body as a major component of selenocysteine, incorporated into selenoproteins, which include the selenocysteine-containing enzymes iodothyronine deiodinases. At the thyroid level, these proteins play an essential role in antioxidant protection and hormone metabolism. This is a narrative review based on PubMed/Medline database research regarding thyroid physiology and conditions with Se and Se-protein interferences. In humans, Se-dependent enzyme functions are best expressed through optimal Se intake, although there is gap in our knowledge concerning the precise mechanisms underlying the interrelation. There is a good level of evidence linking low serum Se to autoimmune thyroid diseases and, to a lesser extent, differentiated thyroid cancer. However, when it comes to routine supplementation, the results are heterogeneous, except in the case of mild Graves' orbitopathy. Autoimmune hypothyroidism is associated with a state of higher oxidative stress, but not all studies found an improvement of thyroid function after Se was introduced as antioxidant support. Meanwhile, no routine supplementation is recommended. Low Se intake is correlated with an increased risk of developing antithyroid antibodies, its supplementation decreasing their titres; there is also a potential reduction in levothyroxine replacement dose required for hypothyroidism and/or the possibility that it prevents progression of subclinical hypothyroidism, although not all studies agree. In thyroid-associated orbitopathy, euthyroidism is more rapidly achieved if the micronutrient is added to traditional drugs, while controls appear to benefit from the microelement only if they are deficient; thus, a basal assay of Se appears advisable to better select patients who need substitution. Clearly, further Se status biomarkers are required. Future introduction of individual supplementation algorithms based on baseline micronutrient levels, underlying or at-risk clinical conditions, and perhaps selenoprotein gene polymorphisms is envisaged.
Collapse
Affiliation(s)
- Ana Valea
- Department of Endocrinology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Endocrinology Clinic, Clinical County Hospital, Cluj-Napoca, Romania.
- , Cluj-Napoca, Romania.
| | - Carmen Emanuela Georgescu
- Department of Endocrinology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Endocrinology Clinic, Clinical County Hospital, Cluj-Napoca, Romania
| |
Collapse
|
41
|
Chekan JR, Moore BS. Preparation and Characterization of Tetrabromopyrrole Debrominase From Marine Proteobacteria. Methods Enzymol 2018; 605:253-265. [PMID: 29909826 PMCID: PMC6211843 DOI: 10.1016/bs.mie.2018.01.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
While halogenases have been studied for decades, the first natural product dehalogenase was only recently described. This bacterial enzyme, Bmp8, catalyzes the reductive debromination of 2,3,4,5-tetrabromopyrrole to form 2,3,4-tribromopyrrole as part of the biosynthesis of pentabromopseudilin, a marine natural product. Bmp8 is hypothesized to utilize a catalytic mechanism analogous to the important human thyroid hormone deiodinase enzyme family, potentially enabling Bmp8 to serve as model system to study this conserved mechanism. Herein, we describe a method for the soluble expression and purification of Bmp8. Furthermore, we detail activity assay protocols to quantify both consumption of the tetrabromopyrrole substrate and formation of the tribromopyrrole product. These methods will enable further study of this unusual enzyme and its catalytic mechanism.
Collapse
Affiliation(s)
- Jonathan R Chekan
- Scripps Institution of Oceanography, University of California at San Diego, La Jolla, CA, United States
| | - Bradley S Moore
- Scripps Institution of Oceanography, University of California at San Diego, La Jolla, CA, United States; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA, United States.
| |
Collapse
|
42
|
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
|
43
|
Abstract
Thyroid hormone signaling is customized in a time and cell-specific manner by the deiodinases, homodimeric thioredoxin fold containing selenoproteins. This ensures adequate T3 action in developing tissues, healthy adults and many disease states. D2 activates thyroid hormone by converting the pro-hormone T4 to T3, the biologically active thyroid hormone. D2 expression is tightly regulated by transcriptional mechanisms triggered by endogenous as well as environmental cues. There is also an on/off switch mechanism that controls D2 activity that is triggered by catalysis and functions via D2 ubiquitination/deubiquitination. D3 terminates thyroid hormone action by inactivation of both T4 and T3 molecules. Deiodinases play a role in thyroid hormone homeostasis, development, growth and metabolic control by affecting the intracellular levels of T3 and thus gene expression on a cell-specific basis. In many cases, tight control of these pathways by T3 is achieved with coordinated reciprocal changes in D2-mediated thyroid hormone activation D3-mediated thyroid hormone inactivation.
Collapse
|
44
|
Mondal S, Mugesh G. Novel thyroid hormone analogues, enzyme inhibitors and mimetics, and their action. Mol Cell Endocrinol 2017; 458:91-104. [PMID: 28408161 DOI: 10.1016/j.mce.2017.04.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/07/2017] [Accepted: 04/07/2017] [Indexed: 12/31/2022]
Abstract
Thyroid hormones (THs) play key roles in modulating the overall metabolism of the body, protein synthesis, fat metabolism, neuronal and bone growth, and cardiovascular as well as renal functions. In this review, we discuss on the thyroid hormone synthesis and activation, thyroid hormone receptors (TRs) and mechanism of action, applications of thyroid hormone analogues, particularly the compounds that are selective ligands for TRβ receptors, or enzyme inhibitors for the treatment of thyroidal disorders with a specific focus on thyroid peroxidase and iodothyronine deiodinases. We also discuss on the development of small-molecule deiodinase mimetics and their mechanism of deiodination, as these compounds have the potential to regulate the thyroid hormone levels.
Collapse
Affiliation(s)
- Santanu Mondal
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
45
|
Schweizer U, Towell H, Vit A, Rodriguez-Ruiz A, Steegborn C. Structural aspects of thyroid hormone binding to proteins and competitive interactions with natural and synthetic compounds. Mol Cell Endocrinol 2017; 458:57-67. [PMID: 28131741 DOI: 10.1016/j.mce.2017.01.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 12/25/2022]
Abstract
Thyroid hormones and their metabolites constitute a vast class of related iodothyronine compounds that contribute to the regulation of metabolic activity and cell differentiation. They are in turn transported, transformed and recognized as signaling molecules through binding to a variety of proteins from a wide range of evolutionary unrelated protein families, which renders these proteins and their iodothyronine binding sites an example for extensive convergent evolution. In this review, we will briefly summarize what is known about iodothyronine binding sites in proteins, the modes of protein/iodothyronine interaction, and the ligand conformations. We will then discuss physiological and synthetic compounds, including popular drugs and food components, that can interfere with iodothyronine binding and recognition by these proteins. The discussion also includes compounds persisting in the environment and acting as endocrine disrupting chemicals.
Collapse
Affiliation(s)
- Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany.
| | - Holly Towell
- Lehrstuhl für Biochemie, Universität Bayreuth, Bayreuth, Germany
| | - Allegra Vit
- Lehrstuhl für Biochemie, Universität Bayreuth, Bayreuth, Germany
| | - Alfonso Rodriguez-Ruiz
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | |
Collapse
|
46
|
Barbosa NV, Nogueira CW, Nogara PA, de Bem AF, Aschner M, Rocha JBT. Organoselenium compounds as mimics of selenoproteins and thiol modifier agents. Metallomics 2017; 9:1703-1734. [PMID: 29168872 DOI: 10.1039/c7mt00083a] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Selenium is an essential trace element for animals and its role in the chemistry of life relies on a unique functional group: the selenol (-SeH) group. The selenol group participates in critical redox reactions. The antioxidant enzymes glutathione peroxidase (GPx) and thioredoxin reductase (TrxR) exemplify important selenoproteins. The selenol group shares several chemical properties with the thiol group (-SH), but it is much more reactive than the sulfur analogue. The substitution of S by Se has been exploited in organic synthesis for a long time, but in the last 4 decades the re-discovery of ebselen (2-phenyl-1,2-benzisoselenazol-3(2H)-one) and the demonstration that it has antioxidant and therapeutic properties has renovated interest in the field. The ability of ebselen to mimic the reaction catalyzed by GPx has been viewed as the most important molecular mechanism of action of this class of compound. The term GPx-like or thiol peroxidase-like reaction was previously coined in the field and it is now accepted as the most important chemical attribute of organoselenium compounds. Here, we will critically review the literature on the capacity of organoselenium compounds to mimic selenoproteins (particularly GPx) and discuss some of the bottlenecks in the field. Although the GPx-like activity of organoselenium compounds contributes to their pharmacological effects, the superestimation of the GPx-like activity has to be questioned. The ability of these compounds to oxidize the thiol groups of proteins (the thiol modifier effects of organoselenium compounds) and to spare selenoproteins from inactivation by soft-electrophiles (MeHg+, Hg2+, Cd2+, etc.) might be more relevant for the explanation of their pharmacological effects than their GPx-like activity. In our view, the exploitation of the thiol modifier properties of organoselenium compounds can be harnessed more rationally than the use of low mass molecular structures to mimic the activity of high mass macromolecules that have been shaped by millions to billions of years of evolution.
Collapse
Affiliation(s)
- Nilda V Barbosa
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| | - Cristina W Nogueira
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| | - Pablo A Nogara
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| | - Andreza F de Bem
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - João B T Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
47
|
Sun Z, Su Q, Rokita SE. The distribution and mechanism of iodotyrosine deiodinase defied expectations. Arch Biochem Biophys 2017; 632:77-87. [PMID: 28774660 DOI: 10.1016/j.abb.2017.07.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/24/2017] [Accepted: 07/30/2017] [Indexed: 12/19/2022]
Abstract
Iodotyrosine deiodinase (IYD) is unusual for its reliance on flavin to promote reductive dehalogenation under aerobic conditions. As implied by the name, this enzyme was first discovered to catalyze iodide elimination from iodotyrosine for recycling iodide during synthesis of tetra- and triiodothyronine collectively known as thyroid hormone. However, IYD likely supports many more functions and has been shown to debrominate and dechlorinate bromo- and chlorotyrosines. A specificity for halotyrosines versus halophenols is well preserved from humans to bacteria. In all examples to date, the substrate zwitterion establishes polar contacts with both the protein and the isoalloxazine ring of flavin. Mechanistic data suggest dehalogenation is catalyzed by sequential one electron transfer steps from reduced flavin to substrate despite the initial expectations for a single two electron transfer mechanism. A purported flavin semiquinone intermediate is stabilized by hydrogen bonding between its N5 position and the side chain of a Thr. Mutation of this residue to Ala suppresses dehalogenation and enhances a nitroreductase activity that is reminiscent of other enzymes within the same structural superfamily.
Collapse
Affiliation(s)
- Zuodong Sun
- Department of Chemistry, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | - Qi Su
- Department of Chemistry, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | - Steven E Rokita
- Department of Chemistry, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA.
| |
Collapse
|
48
|
Cavina L, van der Born D, Klaren PHM, Feiters MC, Boerman OC, Rutjes FPJT. Design of Radioiodinated Pharmaceuticals: Structural Features Affecting Metabolic Stability towards in Vivo Deiodination. European J Org Chem 2017; 2017:3387-3414. [PMID: 28736501 PMCID: PMC5499721 DOI: 10.1002/ejoc.201601638] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Indexed: 11/09/2022]
Abstract
Radioiodinated pharmaceuticals are convenient tracers for clinical and research investigations because of the relatively long half-lives of radioactive iodine isotopes (i.e., 123I, 124I, and 131I) and the ease of their chemical insertion. Their application in radionuclide imaging and therapy may, however, be hampered by poor in vivo stability of the C-I bond. After an overview of the use of iodine in biology and nuclear medicine, we present here a survey of the catabolic pathways for iodinated xenobiotics, including their biodistribution, accumulation, and biostability. We summarize successful rational improvements in the biostability and conclude with general guidelines for the design of stable radioiodinated pharmaceuticals. It appears to be necessary to consider the whole molecule, rather than the radioiodinated fragment alone. Iodine radionuclides are generally retained in vivo on sp2 carbon atoms in iodoarenes and iodovinyl moieties, but not in iodinated heterocycles or on sp3 carbon atoms. Iodoarene substituents also have an influence, with increased in vivo deiodination in the cases of iodophenols and iodoanilines, whereas methoxylation and difluorination improve biostability.
Collapse
Affiliation(s)
- Lorenzo Cavina
- Institute of Molecules and MaterialsFaculty of ScienceRadboud UniversityHeyendaalseweg 1356525 AJ NijmegenNetherlands
- FutureChemistry Holding BV6525 ECNijmegenNetherlands
- Department of Animal Ecology & PhysiologyInstitute of Water & Wetland ResearchFaculty of ScienceRadboud UniversityPOB 90106500 GLNijmegenNetherlands
| | | | - Peter H. M. Klaren
- Department of Animal Ecology & PhysiologyInstitute of Water & Wetland ResearchFaculty of ScienceRadboud UniversityPOB 90106500 GLNijmegenNetherlands
| | - Martin C. Feiters
- Institute of Molecules and MaterialsFaculty of ScienceRadboud UniversityHeyendaalseweg 1356525 AJ NijmegenNetherlands
| | - Otto C. Boerman
- Department of Radiology & Nuclear MedicineRadboud University Medical Center6500 HBNijmegenthe Netherlands
| | - Floris P. J. T. Rutjes
- Institute of Molecules and MaterialsFaculty of ScienceRadboud UniversityHeyendaalseweg 1356525 AJ NijmegenNetherlands
| |
Collapse
|
49
|
Agarwal V, Miles ZD, Winter JM, Eustáquio AS, El Gamal AA, Moore BS. Enzymatic Halogenation and Dehalogenation Reactions: Pervasive and Mechanistically Diverse. Chem Rev 2017; 117:5619-5674. [PMID: 28106994 PMCID: PMC5575885 DOI: 10.1021/acs.chemrev.6b00571] [Citation(s) in RCA: 253] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Naturally produced halogenated compounds are ubiquitous across all domains of life where they perform a multitude of biological functions and adopt a diversity of chemical structures. Accordingly, a diverse collection of enzyme catalysts to install and remove halogens from organic scaffolds has evolved in nature. Accounting for the different chemical properties of the four halogen atoms (fluorine, chlorine, bromine, and iodine) and the diversity and chemical reactivity of their organic substrates, enzymes performing biosynthetic and degradative halogenation chemistry utilize numerous mechanistic strategies involving oxidation, reduction, and substitution. Biosynthetic halogenation reactions range from simple aromatic substitutions to stereoselective C-H functionalizations on remote carbon centers and can initiate the formation of simple to complex ring structures. Dehalogenating enzymes, on the other hand, are best known for removing halogen atoms from man-made organohalogens, yet also function naturally, albeit rarely, in metabolic pathways. This review details the scope and mechanism of nature's halogenation and dehalogenation enzymatic strategies, highlights gaps in our understanding, and posits where new advances in the field might arise in the near future.
Collapse
Affiliation(s)
- Vinayak Agarwal
- Center for Oceans and Human Health, Scripps Institution of Oceanography, University of California, San Diego
| | - Zachary D. Miles
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego
| | | | - Alessandra S. Eustáquio
- College of Pharmacy, Department of Medicinal Chemistry & Pharmacognosy and Center for Biomolecular Sciences, University of Illinois at Chicago
| | - Abrahim A. El Gamal
- Center for Oceans and Human Health, Scripps Institution of Oceanography, University of California, San Diego
| | - Bradley S. Moore
- Center for Oceans and Human Health, Scripps Institution of Oceanography, University of California, San Diego
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego
| |
Collapse
|
50
|
Marsan ES, Bayse CA. Halogen-Bonding Interactions of Polybrominated Diphenyl Ethers and Thyroid Hormone Derivatives: A Potential Mechanism for the Inhibition of Iodothyronine Deiodinase. Chemistry 2017; 23:6625-6633. [DOI: 10.1002/chem.201700407] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Eric S. Marsan
- Department of Chemistry and Biochemistry; Old Dominion University; 5115 Hampton Blvd Norfolk VA 23529 USA
| | - Craig A. Bayse
- Department of Chemistry and Biochemistry; Old Dominion University; 5115 Hampton Blvd Norfolk VA 23529 USA
| |
Collapse
|