1
|
Rivero EM, Castaño MP, Romero IMD, Martínez MJM, Losa FPDL. Anaphylaxis due to green beans ( Phaseolus vulgaris): a new phenotype? Allergol Immunopathol (Madr) 2025; 53:8-11. [PMID: 39786870 DOI: 10.15586/aei.v53i1.1176] [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: 07/11/2024] [Accepted: 09/25/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Anaphylaxis is a severe allergic reaction with increasing incidence in Europe. It is often caused by food, insect venom, and drugs. White, red, and green beans (Phaseolus vulgaris) are legumes of the Fabaceae family consumed worldwide. In Spain, beans are the third most consumed legume species after chickpeas and lentils. Allergy to different legumes is very frequent in children and represents the fifth cause of food allergy. OBJECTIVE Demonstrate a new phenotype in patients with green bean anaphylaxis. MATERIAL AND METHODS We report a 20-year-old woman who experienced anaphylaxis after consuming cooked green beans and required emergency treatment. There were no associated cofactors such as medications, alcohol, or physical exercise. After the episode, she tolerated well nuts, garlic, chickpeas, peas, and soy. Prick-prick with raw and cooked green bean, ISAC, SDS-PAGE and IgE-Western blot were performed. RESULTS The prick+prick test with raw and cooked green beans was positive. The sodium dodecyl sulfate-polyacrylamide gel electrophoresis-immunoglobulin E (SDS-PAGE/IgE)-western blot analysis with white and red beans as well as cooked and raw green beans showed identification of several proteins with a molecular weight of 25-75 kDa in the extract of raw beans. Only one protein with a molecular weight of approximately 10 kDa was recognized in cooked beans. CONCLUSION We present a case of anaphylaxis induced by green beans (Phaseolus vulgaris) because of a 10-kDa protein and tolerance to other legumes, which suggests a new phenotype.
Collapse
|
2
|
Nappi A, Moriello C, Morgante M, Fusco F, Crocetto F, Miro C. Effects of thyroid hormones in skeletal muscle protein turnover. J Basic Clin Physiol Pharmacol 2024; 35:253-264. [PMID: 39297559 DOI: 10.1515/jbcpp-2024-0139] [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: 07/31/2024] [Accepted: 08/29/2024] [Indexed: 11/01/2024]
Abstract
Thyroid hormones (THs) are critical regulators of muscle metabolism in both healthy and unhealthy conditions. Acting concurrently as powerful anabolic and catabolic factors, THs are endowed with a vital role in muscle mass maintenance. As a result, thyroid dysfunctions are the leading cause of a wide range of muscle pathologies, globally identified as myopathies. Whether muscle wasting is a common feature in patients with hyperthyroidism and is mainly caused by THs-dependent stimulation of muscle proteolysis, also muscle growth is often associated with hyperthyroid conditions, linked to THs-dependent stimulation of muscle protein synthesis. Noteworthy, also hypothyroid status negatively impacts on muscle physiology, causing muscle weakness and fatigue. Most of these symptoms are due to altered balance between muscle protein synthesis and breakdown. Thus, a comprehensive understanding of THs-dependent skeletal muscle protein turnover might facilitate the management of physical discomfort or weakness in conditions of thyroid disease. Herein, we describe the molecular mechanisms underlying the THs-dependent alteration of skeletal muscle structure and function associated with muscle atrophy and hypertrophy, thus providing new insights for targeted modulation of skeletal muscle dynamics.
Collapse
Affiliation(s)
- Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Caterina Moriello
- Department of Advanced Medical and Surgical Sciences, University of Naples "Luigi Vanvitelli", Naples, Italy
| | | | - Ferdinando Fusco
- Department of Women, Children and General and Specialist Surgery, University of Naples "Luigi Vanvitelli", Naples, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Caterina Miro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
3
|
Mudri D, Bilić Ćurčić I, Meštrović L, Mihaljević I, Kizivat T. Hyperthyroidism and Wnt Signaling Pathway: Influence on Bone Remodeling. Metabolites 2023; 13:metabo13020241. [PMID: 36837860 PMCID: PMC9968154 DOI: 10.3390/metabo13020241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Graves' disease is an autoimmune disease of the thyroid gland, characterized by increased production of thyroid hormones, which can affect many different organ systems in the body. Among other problems, it can cause disorders of the skeletal system, shortening the bone remodeling cycle and causing a decrease in bone density. The Wnt cascade signaling pathway and the β-catenin, as a part of the canonical Wnt pathway, also play roles in maintaining bone mass. Inhibition of the Wnt pathway can cause bone loss, and its stimulation can increase it. The Wnt signaling pathway influences the effectiveness of thyroid hormones by affecting receptors for thyroid hormones and deiodinase, while thyroid hormones can change levels of β-catenin within the cell cytoplasm. This indicates that the Wnt pathway and thyroid hormone levels, including hyperthyroidism, are linked and may act together to change bone density. In this review article, we attempt to explain the interplay between thyroid hormones and the Wnt pathway on bone density, with a focus on directions for further research and treatment options.
Collapse
Affiliation(s)
- Dunja Mudri
- Department of Nuclear Medicine and Oncology, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Osijek, 31000 Osijek, Croatia
| | - Ines Bilić Ćurčić
- Department of Pharmacology, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
- Department of Endocrinology and Metabolism Disorders, University Hospital Osijek, 31000 Osijek, Croatia
- Correspondence: (I.B.Ć.); (T.K.)
| | - Lucija Meštrović
- Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
| | - Ivica Mihaljević
- Department of Nuclear Medicine and Oncology, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Osijek, 31000 Osijek, Croatia
- Academy of Medical Sciences of Croatia, 31000 Osijek, Croatia
| | - Tomislav Kizivat
- Department of Nuclear Medicine and Oncology, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Osijek, 31000 Osijek, Croatia
- Correspondence: (I.B.Ć.); (T.K.)
| |
Collapse
|
4
|
Yang Y, Lu M, Qian J, Xu Y, Li B, Le G, Xie Y. Dietary Methionine Restriction Promotes Fat Browning and Attenuates Hepatic Lipid Accumulation in High-Choline-Fed Mice Associated with the Improvement of Thyroid Function. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1447-1463. [PMID: 36632677 DOI: 10.1021/acs.jafc.2c05535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
This study aims to explore the influences of a methionine-restricted diet (MRD) on fat browning and hepatic lipid accumulation in mice fed with a high-choline diet (HCD) and their possible mechanisms. ICR mice were randomly divided into three groups and fed with a normal diet (0.86% methionine + 0.20% choline, ND), HCD (0.86% methionine + 1.20% choline), or MRD (0.17% methionine + 1.20% choline) for 90 consecutive days. We found that MRD reduced body weight and fat mass; increased heat production and ambulatory locomotor activity; reduced hepatic and plasma lipid levels, hepatic fatty infiltration area, and adipocyte volume in white and brown adipose tissue; promoted fat browning, especially upregulated gene and protein expression levels of uncoupling protein 1 (UCP1); and promoted fat catabolism and inhibited fat anabolism in the liver and adipose tissue. Moreover, MRD increased antioxidant defenses and reduced inflammatory cytokine levels in the thyroid, blood, and liver. Furthermore, MRD improved thyroid morphological structure, promoted the synthesis and secretion of thyroid hormones, and enhanced the actions of thyroid hormones on its receptor organs (liver and adipose tissue). These findings suggested that MRD promoted fat browning and attenuated hepatic lipid accumulation in HCD mice associated with the improvement of thyroid function.
Collapse
Affiliation(s)
- Yuhui Yang
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China
| | - Manman Lu
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China
| | - Jing Qian
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China
| | - Yuncong Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Bowen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Guowei Le
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Yanli Xie
- National Engineering Laboratory/Key Laboratory of Henan Province, College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, Henan, China
| |
Collapse
|
5
|
Mudri D, Kizivat T, Smolić M, Mihaljević I, Smolić R, Raguž Lučić N, Bilić-Ćurčić I. IMPACT OF GRAVES' DISEASE AND ANTITHYROID DRUG THERAPY ON BONE MINERAL DENSITY - PATHOPHYSIOLOGICAL MECHANISMS AND CLINICAL RELEVANCE. Acta Clin Croat 2022; 61:496-504. [PMID: 37492357 PMCID: PMC10364114 DOI: 10.20471/acc.2022.61.03.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/11/2019] [Indexed: 07/27/2023] Open
Abstract
Graves' disease is an autoimmune disease characterized by excessive thyroid hormone production. One of the consequences of that state can be a decrease in bone mineral density (BMD). Graves' disease is often treated with antithyroid drugs (ATD) as first line therapy, which can lead to disease remission. Moreover, recent data show that improvement in BMD can be expected. However, vitamin D deficiency can coexist along with Graves' disease, which is also involved in the process of bone remodeling. It is still not known whether lower values of vitamin D can contribute to onset of Graves' disease and if its supplementation might be helpful in therapy for hyperthyroidism. In the past couple of decades, osteopenia and osteoporosis have become a major health burden not only in post-menopausal women but also as a result of other diseases, leading to extensive research into various pathophysiological mechanisms responsible for bone remodeling. The Wnt (wingless integrated) signaling pathway is a very important factor in bone homeostasis, especially the canonical pathway. Present data indicate that stimulation of the Wnt pathway leads to bone mass increase and, in contrast, its inhibition leads to bone mass decrease. Hence, inhibitors of the canonical Wnt pathway became the focus of interest, in particular sclerostin and dickkopf 1 (DKK1). Hyperthyroidism and osteopenia/osteoporosis are quite common today and can coexist together or as separate entities. In this article, we aimed to give an overview of possible associations and potential mutual pathophysiological mechanisms.
Collapse
Affiliation(s)
- Dunja Mudri
- Faculty of Medicine, Josip Juraj Strossmayer University, Osijek
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Centre Osijek
| | - Tomislav Kizivat
- Faculty of Medicine, Josip Juraj Strossmayer University, Osijek
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Centre Osijek
| | - Martina Smolić
- Faculty of Medicine, Josip Juraj Strossmayer University, Osijek
- Faculty of Dental Medicine and Health Osijek Josip Juraj Strossmayer University
| | - Ivica Mihaljević
- Faculty of Medicine, Josip Juraj Strossmayer University, Osijek
- Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Centre Osijek
| | - Robert Smolić
- Faculty of Dental Medicine and Health Osijek Josip Juraj Strossmayer University
| | - Nikola Raguž Lučić
- Faculty of Dental Medicine and Health Osijek Josip Juraj Strossmayer University
| | - Ines Bilić-Ćurčić
- Faculty of Medicine, Josip Juraj Strossmayer University, Osijek
- Clinical Department of Endocrinology and Metabolism Disorders, University Hospital Centre Osijek
| |
Collapse
|
6
|
Major E, Keller I, Horváth D, Tamás I, Erdődi F, Lontay B. Smoothelin-Like Protein 1 Regulates the Thyroid Hormone-Induced Homeostasis and Remodeling of C2C12 Cells via the Modulation of Myosin Phosphatase. Int J Mol Sci 2021; 22:10293. [PMID: 34638630 PMCID: PMC8508602 DOI: 10.3390/ijms221910293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 11/28/2022] Open
Abstract
The pathological elevation of the active thyroid hormone (T3) level results in the manifestation of hyperthyroidism, which is associated with alterations in the differentiation and contractile function of skeletal muscle (SKM). Myosin phosphatase (MP) is a major cellular regulator that hydrolyzes the phosphoserine of phosphorylated myosin II light chain. MP consists of an MYPT1/2 regulatory and a protein phosphatase 1 catalytic subunit. Smoothelin-like protein 1 (SMTNL1) is known to inhibit MP by directly binding to MP as well as by suppressing the expression of MYPT1 at the transcriptional level. Supraphysiological vs. physiological concentration of T3 were applied on C2C12 myoblasts and differentiated myotubes in combination with the overexpression of SMTNL1 to assess the role and regulation of MP under these conditions. In non-differentiated myoblasts, MP included MYPT1 in the holoenzyme complex and its expression and activity was regulated by SMTNL1, affecting the phosphorylation level of MLC20 assessed using semi-quantitative Western blot analysis. SMTNL1 negatively influenced the migration and cytoskeletal remodeling of myoblasts measured by high content screening. In contrast, in myotubes, the expression of MYPT2 but not MYPT1 increased in a T3-dependent and SMTNL1-independent manner. T3 treatment combined with SMTNL1 overexpression impeded the activity of MP. In addition, MP interacted with Na+/K+-ATPase and dephosphorylated its inhibitory phosphorylation sites, identifying this protein as a novel MP substrate. These findings may help us gain a better understanding of myopathy, muscle weakness and the disorder of muscle regeneration in hyperthyroid patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Beáta Lontay
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (E.M.); (I.K.); (D.H.); (I.T.); (F.E.)
| |
Collapse
|
7
|
Terasaki T. [Development of Novel Methodology and Its Application for Clarifying the Transport Function of the Blood-brain Barrier]. YAKUGAKU ZASSHI 2021; 141:447-462. [PMID: 33790111 DOI: 10.1248/yakushi.20-00232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The blood-brain barrier (BBB) consists of brain capillary endothelial cells linked by tight junctions and serves to regulate the transfer of endogenous compounds and xenobiotics between the circulating blood and brain interstitial fluid. We have developed a methodology to characterize brain-to-blood efflux transport in vivo, using the Brain Efflux Index and an in vitro culture model of the BBB, i.e., a conditionally immortalized cell line of the neurovascular unit. Employing these methods, we showed that the BBB plays an important role in protecting the brain by transporting neurotransmitters, neuromodulators, metabolites, uremic toxins, and xenobiotics together with atrial natriuretic peptide from the brain interstitial fluid to the circulating blood. We also developed a highly selective, sensitive LC-MS/MS method for simultaneous protein quantification. We found significant species differences in the expression amounts of various BBB transporter proteins among mice, rats, marmosets, cynomolgus monkeys, and humans. Among transporter proteins at the BBB, multidrug resistance protein 1 (Mdr1/Abcb1) is known to generate a concentration gradient of unbound substrate drugs between the blood and brain. Based on measurements of the intrinsic efflux transport rate of Mdr1 and the protein expression amounts of Mdr1 in mouse brain capillaries and Mdr1-expressing cell lines, we predicted the unbound drug concentration gradients of 7 drugs in the mouse brain in vivo. This was the first successful prediction of in vivo drug transport activity from in vitro experimental data and transporter protein concentration in tissues. This methodology and findings should greatly advance central nervous system barrier research.
Collapse
Affiliation(s)
- Tetsuya Terasaki
- Membrane Transport and Drug Targeing Laboratory, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
8
|
Pagnin M, Kondos-Devcic D, Chincarini G, Cumberland A, Richardson SJ, Tolcos M. Role of thyroid hormones in normal and abnormal central nervous system myelination in humans and rodents. Front Neuroendocrinol 2021; 61:100901. [PMID: 33493504 DOI: 10.1016/j.yfrne.2021.100901] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/07/2021] [Accepted: 01/16/2021] [Indexed: 12/13/2022]
Abstract
Thyroid hormones (THs) are instrumental in promoting the molecular mechanisms which underlie the complex nature of neural development and function within the central nervous system (CNS) in vertebrates. The key neurodevelopmental process of myelination is conserved between humans and rodents, of which both experience peak fetal TH concentrations concomitant with onset of myelination. The importance of supplying adequate levels of THs to the myelin producing cells, the oligodendrocytes, for promoting their maturation is crucial for proper neural function. In this review we examine the key TH distributor and transport proteins, including transthyretin (TTR) and monocarboxylate transporter 8 (MCT8), essential for supporting proper oligodendrocyte and myelin health; and discuss disorders with impaired TH signalling in relation to abnormal CNS myelination in humans and rodents. Furthermore, we explore the importance of using novel TH analogues in the treatment of myelination disorders associated with abnormal TH signalling.
Collapse
Affiliation(s)
- Maurice Pagnin
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Delphi Kondos-Devcic
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Ginevra Chincarini
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | - Angela Cumberland
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia
| | | | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia.
| |
Collapse
|
9
|
McNerney C, Johnston RJ. Thyroid hormone signaling specifies cone photoreceptor subtypes during eye development: Insights from model organisms and human stem cell-derived retinal organoids. VITAMINS AND HORMONES 2021; 116:51-90. [PMID: 33752828 DOI: 10.1016/bs.vh.2021.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Cones are the color-detecting photoreceptors of the vertebrate eye. Cones are specialized into subtypes whose functions are determined by the expression of color-sensitive opsin proteins. Organisms differ greatly in the number and patterning of cone subtypes. Despite these differences, thyroid hormone is an important regulator of opsin expression in most vertebrates. In this chapter, we outline how the timing of thyroid hormone signaling controls cone subtype fates during retinal development. We first examine our current understanding of cone subtype specification in model organisms and then describe advances in human stem cell-derived organoid technology that identified mechanisms controlling development of the human retina.
Collapse
Affiliation(s)
- Christina McNerney
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
10
|
Abstract
Hönes et al. have recently shown that in vivo interference with the apparatus of the nuclear receptor-mediated, gene-driven mechanism of triiodothyronine (T3) actions fails to eliminate all actions of T3. However, the investigators conducting that study provided little information regarding the mechanisms that might be responsible for conferring those implied gene-independent effects. Dratman has long ago suggested a system wherein such gene-free mechanisms might operate. Therefore, since news of that discovery was originally published in 1974, it seems appropriate to describe the progress made since then. We propose that thyroxine and triiodothyronine have many different structural properties that may confer a series of different capabilities on their functions. These conform with our proposal that a series of catecholamine analogs and their conversion to iodothyronamines, allows them to perform many of the functions that previously were attributed to nuclear receptors regulating gene expression. The actions of deiodinases and the differential distribution of iodine substituents are among the critical factors that allow catecholamine analogs to change their effects into ones that either activate their targets or block them. They do this by using two different deiodinases to vary the position of an iodide ion on the diphenylether backbones of thyroxine metabolites. A panoply of these structural features imparts major unique functional properties on the behavior of vertebrates in general and possibly on Homo sapiens in particular.
Collapse
Affiliation(s)
- Mary B Dratman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph V Martin
- Biology Department, Center for Computational and Integrative Biology, Rutgers University, 315 Penn St., Camden, NJ 08102, USA
| |
Collapse
|
11
|
Sinha P, Chakrabarti N, Ghosh N, Mitra S, Dalui S, Bhattacharyya A. Alterations of thyroidal status in brain regions and hypothalamo-pituitary-blood-thyroid-axis associated with dopaminergic depletion in substantia nigra and ROS formation in different brain regions after MPTP treatment in adult male mice. Brain Res Bull 2020; 156:131-140. [PMID: 31891753 DOI: 10.1016/j.brainresbull.2019.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 12/20/2022]
Abstract
MPTP produces oxidative stress, damages niagrostriatal dopaminergic neurons and develops Parkinsonism in rodents. Due to paucity of information, the thyroidal status in brain regions and peripheral tissues during different post-treatment days in MPTP-induced mice had been executed in the present study. MPTP depleted tyrosine hydroxylase protein expressions that signify the dopaminergic neuronal damage in substantia nigra. MPTP elevated ROS formation differentially in brain regions (cerebral cortex, hippocampus, substantia nigra) with maximal elevation at hippocampus. The changes in thyroid hormone (T4 and T3) levels indicate that brain regions might combat the adverse situation by keeping the levels of thyroid hormones either unchanged or in the elevated conditions in the latter phases (day-3 and day-7), apart from the depletion of thyroid hormones in certain brain regions (T4 in SN and hippocampus, T3 in hippocampus) as the immediate (day-1) effects after MPTP treatment. MPTP caused alterations of cellular morphology, RNA:Protein ratio and TPO protein expression, concomitantly depleted TPO mRNA expression and elevated TSH levels in the thyroid gland. Although T4 levels changed differentially, T3 levels remained unaltered in thyroid gland throughout the post-treatment days. Results have been discussed mentioning the putative role of T4 and TSH in apoptosis and/or proliferation/differentiation of thyrocytes. In blood, T4 levels remained unchanged while the changes in T3 and TSH levels did not signify the clinical feature of hypo/hyperthyroidism of animals. In the pituitary, both T4 and T3 levels remained elevated where TSH differentially altered (elevated followed by depletion) during post-treatment days. Notably, T4, T3 and TSH levels did not alter in hypothalamus except initial (day-1) depletion of the T4 level. Therefore, the feedback control mechanism of hypothalamo-pituitary-blood-thyroid-axis failed to occur after MPTP treatment. Overall, MPTP altered thyroidal status in the brain and peripheral tissues while both events might occur in isolation as well.
Collapse
Affiliation(s)
- Priyobrata Sinha
- Immunology Lab, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India; Department of Physiology, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata 700009, India
| | - Nilkanta Chakrabarti
- Department of Physiology, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata 700009, India; Centres with Potential for Excellence in Particular Areas (CPEPA, UGC), Centre for "Electrophysiology & Neuroimaging Studies Including Mathematical Modeling" India.
| | - Nabanita Ghosh
- Immunology Lab, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Soham Mitra
- Immunology Lab, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Shauryabrota Dalui
- Immunology Lab, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Arindam Bhattacharyya
- Immunology Lab, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India.
| |
Collapse
|
12
|
Zarif H, Paquet A, Lebrigand K, Arguel MJ, Heurteaux C, Glaichenhaus N, Chabry J, Guyon A, Petit-Paitel A. CD4+ T Cells Affect the Thyroid Hormone Transport at the Choroid Plexus in Mice Raised in Enriched Environment. Neuroimmunomodulation 2019; 26:59-66. [PMID: 30703773 DOI: 10.1159/000495987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/04/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Others and we have shown that T cells have an important role in hippocampal synaptic plasticity, including neurogenesis in the dentate gyrus, spinogenesis, and glutamatergic synaptic function in the CA of the hippocampus. Hippocampus plasticity is particularly involved in the brain effects of the enriched environment (EE), and interestingly CD4+ and CD8+ T cells play essential and differential roles in these effects. However, the precise mechanisms by which they act on the brain remain elusive. OBJECTIVES We searched for a putative mechanism of action by which CD4+ T cells could influence brain plasticity and hypothesized that they could regulate protein transport at the level of the blood-CSF barrier in the choroid plexus. METHOD We compared mice housed in EE and deprived of CD4+ T cells using a depleting antibody with a control group injected with the control isotype. We analyzed in the hippocampus the gene expression profiles using the Agilent system, and the expression of target proteins in plasma, CSF, and the choroid plexus using ELISA. RESULTS We show that CD4+ T cells may influence EE-induced hippocampus plasticity via thyroid hormone signaling by regulating in the choroid plexus the expression of transthyretin, the major transporter of thyroxine (T4) to the brain parenchyma. CONCLUSIONS Our study highlights the contribution of close interactions between the immune and neuroendocrine systems in brain plasticity and function.
Collapse
Affiliation(s)
- Hadi Zarif
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | - Agnès Paquet
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | | | | | | | | | - Joëlle Chabry
- Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France
| | - Alice Guyon
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France,
| | | |
Collapse
|
13
|
Andrade MN, Santos-Silva AP, Rodrigues-Pereira P, Paiva-Melo FD, de Lima Junior NC, Teixeira MP, Soares P, Dias GRM, Graceli JB, de Carvalho DP, Ferreira ACF, Miranda-Alves L. The environmental contaminant tributyltin leads to abnormalities in different levels of the hypothalamus-pituitary-thyroid axis in female rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 241:636-645. [PMID: 29902746 DOI: 10.1016/j.envpol.2018.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 06/08/2023]
Abstract
Tributyltin is a biocide used in nautical paints, aiming to reduce fouling of barnacles in ships. Despite the fact that many effects of TBT on marine species are known, studies in mammals have been limited, especially those evaluating its effect on the function of the hypothalamus-pituitary-thyroid (HPT) axis. The aim of this study was to investigate the effects of subchronic exposure to TBT on the HPT axis in female rats. Female Wistar rats received vehicle, TBT 200 ng kg-1 BW d-1 or 1000 ng kg-1 BW d-1 orally by gavage for 40 d. Hypothalamus, pituitary, thyroid, liver and blood samples were collected. TBT200 and TBT1000 thyroids showed vacuolated follicular cells, with follicular hypertrophy and hyperplasia. An increase in epithelial height and a decrease in the thyroid follicle and colloid area were observed in TBT1000 rats. Moreover, an increase in the epithelium/colloid area ratio was observed in both TBT groups. Lower TRH mRNA expression was observed in the hypothalami of TBT200 and TBT1000 rats. An increase in Dio1 mRNA levels was observed in the hypothalamus and thyroid in TBT1000 rats only. TSH serum levels were increased in TBT200 rats. In TBT1000 rats, there was a decrease in total T4 serum levels compared to control rats, whereas T3 serum levels did not show significant alterations. We conclude that TBT exposure can promote critical abnormalities in the HPT axis, including changes in TRH mRNA expression and serum TSH and T4 levels, in addition to affecting thyroid morphology. These findings demonstrate that TBT disrupts the HPT axis. Additionally, the changes found in thyroid hormones suggest that TBT may interfere with the peripheral metabolism of these hormones, an idea corroborated by the observed changes in Dio1 mRNA levels. Therefore, TBT exposition might interfere not only with the thyroid axis but also with thyroid hormone metabolism.
Collapse
Affiliation(s)
- Marcelle Novaes Andrade
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Ana Paula Santos-Silva
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Paula Rodrigues-Pereira
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Francisca Diana Paiva-Melo
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Niedson Correa de Lima Junior
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Mariana Pires Teixeira
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Patologia, Universidade Federal Fluminense, Brazil
| | - Paula Soares
- Institute for Research and Innovation in Health, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP) - Cancer Biology, Porto, Portugal; Medical Faculty, University of Porto, Porto, Portugal; Department of Pathology, Medical Faculty of Porto University, Porto, Portugal
| | - Glaecir Roseni Munstock Dias
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Jones Bernardes Graceli
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Endocrinologia e Toxicologia Celular, Departamento de Morfologia, Universidade Federal do Espírito Santo, Brazil
| | - Denise Pires de Carvalho
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Andrea Claudia Freitas Ferreira
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Polo de Xerém/NUMPEX, Universidade Federal do Rio de Janeiro, Brazil
| | - Leandro Miranda-Alves
- Grupo de Pesquisa, Desenvolvimento e Inovação em Endocrinologia Experimental-GPDIEEx, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil; Programa de Pós-graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Suzuki M, Banno K, Usui T, Funasaka N, Segawa T, Kirihata T, Kamisako H, Ueda K, Munakata A. Seasonal changes in plasma levels of thyroid hormones and the effects of the hormones on cellular ATP content in common bottlenose dolphin. Gen Comp Endocrinol 2018. [PMID: 29524524 DOI: 10.1016/j.ygcen.2018.03.008] [Citation(s) in RCA: 2] [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] [Indexed: 10/17/2022]
Abstract
In general, thyroid hormones (THs) stimulate cellular metabolism by inducing ATP utilization that collaterally leads to thermogenesis. However, in cetaceans, TH functions and the contribution of THs to cold adaptation are not fully understood. To investigate the role of THs in metabolism of cetaceans, seasonal changes in circulating levels in THs were investigated in the common bottlenose dolphin Tursiops truncatus that were monitored under two different conditions for two years, with routine measurements of body temperature (BT), water temperature (WT) and air temperature (AT). The effects of THs on ATP synthesis were determined using cultured cells. Blood samples were collected from the species kept in different conditions at the Taiji Whale Museum located in the temperate zone and at Okinawa Expo Park in the subtropical zone. Circulating levels in total thyroxine (T4) for the dolphins at both aquaria and total 3,5,3'-tri-iodothyronine (T3) levels in dolphins at Taiji were measured by enzyme-linked immunoassay methods, respectively, and average concentrations were compared among seasons. To confirm the effects of THs on ATP synthesis, T3 or T4 was administrated to cultured kidney cells from the same species and cellular ATP contents were quantified at 0, 24, 48, 96 and 192 h after administration. BT of common bottlenose dolphins in each aquarium was measured for health check by chance in Taiji and every morning in Okinawa. WT in pools and AT were also measured every morning. Circulating T4 levels in autumn and winter were lower than those in spring and summer in dolphins in Taiji where WT and AT varied greatly from season to season. T4 levels showed a small difference between spring and autumn in dolphins in Okinawa with warmer WT and AT in smaller amplitude ranges than in Taiji. Total T3 level in Taiji was highest in spring and lowest in autumn as T4 levels, but not significant. The BT of dolphins in Taiji was also lower in autumn and winter compared with those in spring and summer, whereas the BT of dolphins in Okinawa fell in autumn but rose in summer, albeit to a lesser extent than in Taiji. Cellular ATP was increased by administration of both T3 and T4 compared to control. Collectively, these results suggest that the cellular metabolic activities regulated by THs may be enhanced in dolphins exposed to increasing surrounding temperature for lipolysis and reduced in dolphins exposed to colder conditions for fat accumulation.
Collapse
Affiliation(s)
| | - Kaho Banno
- Nihon University, Kameino, Kanagawa, Japan
| | | | - Noriko Funasaka
- Mie University, Tsu, Mie, Japan; Taiji Whale Museum, Higashimurogun, Wakayama, Japan
| | - Takao Segawa
- Nihon University, Kameino, Kanagawa, Japan; Mie University, Tsu, Mie, Japan
| | | | | | - Keiichi Ueda
- Okinawa Churashima Foundation, Motobu, Okinawa, Japan
| | | |
Collapse
|
15
|
Abstract
As one of the most basal living vertebrates, lampreys represent an excellent model system to study the evolution of thyroid hormone (TH) signaling. The lamprey hypothalamic-pituitary-thyroid and reproductive axes overlap functionally. Lampreys have 3 gonadotropin-releasing hormones and a single glycoprotein hormone from the hypothalamus and pituitary, respectively, that regulate both the reproductive and thyroid axes. TH synthesis in larval lampreys takes place in an endostyle that transforms into typical vertebrate thyroid tissue during metamorphosis; both the endostyle and follicular tissue have all the typical TH synthetic components found in other vertebrates. Furthermore, lampreys also have the vertebrate suite of peripheral regulators including TH distributor proteins (THDPs), deiodinases and TH receptors (TRs). Although at the molecular level the components of the lamprey thyroid system are ancestral to other vertebrates, their functions have been largely conserved. TH signaling as it relates to lamprey metamorphosis represents a particularly interesting phenomenon. Unlike other metamorphosing vertebrates, lamprey THs increase throughout the larval period, peak prior to metamorphosis and decline rapidly at the onset of metamorphosis; patterns of deiodinase activity are consistent with these increases and declines. Moreover, goitrogens (which suppress TH levels) initiate precocious metamorphosis, and exogenous TH treatment blocks goitrogen-induced metamorphosis and disrupts natural metamorphosis. Despite this clear physiological difference, TH action via TRs is consistent with higher vertebrates. Based on observations that TRs are upregulated in a tissue-specific fashion during morphogenesis and the finding that lamprey TRs upregulate genes via THs in a fashion similar to higher vertebrates, we propose the following hypothesis for further testing. THs have a dual role in lampreys where high TH levels promote larval feeding and growth and then at the onset of metamorphosis TH levels decrease rapidly; at this time the relatively low TH levels function via TRs in a fashion similar to that of other metamorphosing vertebrates.
Collapse
Affiliation(s)
- Richard G Manzon
- Department of Biology, University of Regina, 3737 Wascana Parkway, Regina, SK S4S 0A2, Canada.
| | - Lori A Manzon
- Department of Biology, University of Regina, 3737 Wascana Parkway, Regina, SK S4S 0A2, Canada
| |
Collapse
|
16
|
Hülsmann S, Hagos L, Heuer H, Schnell C. Limitations of Sulforhodamine 101 for Brain Imaging. Front Cell Neurosci 2017; 11:44. [PMID: 28293173 PMCID: PMC5328990 DOI: 10.3389/fncel.2017.00044] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/09/2017] [Indexed: 11/19/2022] Open
Abstract
Since 2004, the red fluorescent dye Sulforhodamine 101 (SR101) has been boosting the functional analysis of astrocytes in a functional environment in an unprecedented way. However, two major limitations have been challenging the usefulness of this tool for cellular imaging: (i) SR101 is not as specific for astrocytes as previously reported; and (ii) discoveries of severe excitatory side effects of SR101 are bearing the risk of unwanted alteration of the system of interest. In this article, we summarize the current knowledge about SR101-labeling protocols and discuss the problems that arise from varying of the staining protocols. Furthermore, we provide a testable hypothesis for the observed hyper-excitability that can be observed when using SR101.
Collapse
Affiliation(s)
- Swen Hülsmann
- Clinic for Anesthesiology, University Hospital GöttingenGöttingen, Germany; DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen, Germany
| | - Liya Hagos
- Clinic for Anesthesiology, University Hospital GöttingenGöttingen, Germany; DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen, Germany
| | - Heike Heuer
- Leibniz-Institut für Umweltmedizinische Forschung GmbH Düsseldorf, Germany
| | - Christian Schnell
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB) Göttingen, Germany
| |
Collapse
|
17
|
Gigena N, Alamino VA, Montesinos MDM, Nazar M, Louzada RA, Wajner SM, Maia AL, Masini-Repiso AM, Carvalho DP, Cremaschi GA, Pellizas CG. Dissecting thyroid hormone transport and metabolism in dendritic cells. J Endocrinol 2017; 232:337-350. [PMID: 28052998 DOI: 10.1530/joe-16-0423] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 12/15/2016] [Indexed: 01/23/2023]
Abstract
We reported thyroid hormone (TH) receptor expression in murine dendritic cells (DCs) and 3,5,3'-triiodothyronine (T3)-dependent stimulation of DC maturation and ability to develop a Th1-type adaptive response. Moreover, an increased DC capacity to promote antigen-specific cytotoxic T-cell activity, exploited in a DC-based antitumor vaccination protocol, was revealed. However, putative effects of the main circulating TH, l-thyroxine (T4) and the mechanisms of TH transport and metabolism at DC level, crucial events for TH action at target cell level, were not known. Herein, we show that T4 did not reproduce those registered T3-dependent effects, finding that may reflect a homoeostatic control to prevent unspecific systemic activation of DCs. Besides, DCs express MCT10 and LAT2 TH transporters, and these cells mainly transport T3 with a favored involvement of MCT10 as its inhibition almost prevented T3 saturable uptake mechanism and reduced T3-induced IL-12 production. In turn, DCs express iodothyronine deiodonases type 2 and 3 (D2, D3) and exhibit both enzymatic activities with a prevalence towards TH inactivation. Moreover, T3 increased MCT10 and LAT2 expression and T3 efflux from DCs but not T3 uptake, whereas it induced a robust induction of D3 with a parallel slight reduction in D2. These findings disclose pivotal events involved in the mechanism of action of THs on DCs, providing valuable tools for manipulating the immunogenic potential of these cells. Furthermore, they broaden the knowledge of the TH mechanism of action at the immune system network.
Collapse
Affiliation(s)
- Nicolás Gigena
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Vanina A Alamino
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Del Mar Montesinos
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Magalí Nazar
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ruy A Louzada
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - Simone M Wajner
- Thyroid UnitEndocrinology Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana L Maia
- Thyroid UnitEndocrinology Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana M Masini-Repiso
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Denise P Carvalho
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - Graciela A Cremaschi
- Laboratorio de Neuroinmunomodulación y Oncología MolecularInstituto de Investigaciones Biomédicas (BIOMED-CONICET), Universidad Católica Argentina, Ciudad Autónoma de Buenos Aires, Argentina
| | - Claudia G Pellizas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET)Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
18
|
Kato Y, Fujii A, Haraguchi K, Fujii Y, Atobe K, Endo T, Kimura O, Koga N, Ohta C, Yamada S, Degawa M. Possible mechanism for the polychlorinated bipheny-linduced liver-selective accumulation of thyroxine in rats. J Toxicol Sci 2017; 42:663-669. [DOI: 10.2131/jts.42.663] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Yoshihisa Kato
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University
| | - Aki Fujii
- School of Pharmaceutical Sciences, University of Shizuoka
| | | | | | - Kazutaka Atobe
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University
| | - Tetsuya Endo
- Faculty of Pharmaceutical Sciences, Health Science University of Hokkaido
| | - Osamu Kimura
- Faculty of Pharmaceutical Sciences, Health Science University of Hokkaido
| | - Nobuyuki Koga
- Faculty of Nutritional Sciences, Nakamura Gakuen University
| | - Chiho Ohta
- Faculty of Nutritional Sciences, Nakamura Gakuen University
| | - Shizuo Yamada
- School of Pharmaceutical Sciences, University of Shizuoka
| | | |
Collapse
|
19
|
Van den Berghe G. On the Neuroendocrinopathy of Critical Illness. Perspectives for Feeding and Novel Treatments. Am J Respir Crit Care Med 2016; 194:1337-1348. [DOI: 10.1164/rccm.201607-1516ci] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
20
|
|
21
|
Abstract
The thyroid hormones, thyroxine (T4) and triiodothyronine (T3), are essential for normal growth and development of the fetus. Their bioavailability in utero depends on development of the fetal hypothalamic-pituitary-thyroid gland axis and the abundance of thyroid hormone transporters and deiodinases that influence tissue levels of bioactive hormone. Fetal T4 and T3 concentrations are also affected by gestational age, nutritional and endocrine conditions in utero, and placental permeability to maternal thyroid hormones, which varies among species with placental morphology. Thyroid hormones are required for the general accretion of fetal mass and to trigger discrete developmental events in the fetal brain and somatic tissues from early in gestation. They also promote terminal differentiation of fetal tissues closer to term and are important in mediating the prepartum maturational effects of the glucocorticoids that ensure neonatal viability. Thyroid hormones act directly through anabolic effects on fetal metabolism and the stimulation of fetal oxygen consumption. They also act indirectly by controlling the bioavailability and effectiveness of other hormones and growth factors that influence fetal development such as the catecholamines and insulin-like growth factors (IGFs). By regulating tissue accretion and differentiation near term, fetal thyroid hormones ensure activation of physiological processes essential for survival at birth such as pulmonary gas exchange, thermogenesis, hepatic glucogenesis, and cardiac adaptations. This review examines the developmental control of fetal T4 and T3 bioavailability and discusses the role of these hormones in fetal growth and development with particular emphasis on maturation of somatic tissues critical for survival immediately at birth.
Collapse
Affiliation(s)
- A J Forhead
- Department of PhysiologyDevelopment and Neuroscience, University of Cambridge, Physiology Building, Downing Street, Cambridge CB2 3EG, UKDepartment of Biological and Medical SciencesOxford Brookes University, Oxford OX3 0BP, UKDepartment of PhysiologyDevelopment and Neuroscience, University of Cambridge, Physiology Building, Downing Street, Cambridge CB2 3EG, UKDepartment of Biological and Medical SciencesOxford Brookes University, Oxford OX3 0BP, UK
| | - A L Fowden
- Department of PhysiologyDevelopment and Neuroscience, University of Cambridge, Physiology Building, Downing Street, Cambridge CB2 3EG, UKDepartment of Biological and Medical SciencesOxford Brookes University, Oxford OX3 0BP, UK
| |
Collapse
|
22
|
Langouche L, Van den Berghe G. Hypothalamic-pituitary hormones during critical illness: a dynamic neuroendocrine response. HANDBOOK OF CLINICAL NEUROLOGY 2014; 124:115-26. [PMID: 25248583 DOI: 10.1016/b978-0-444-59602-4.00008-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Independent of the underlying condition, critical illness is characterized by a uniform dysregulation of the hypothalamic-pituitary-peripheral axes. In most axes a clear biphasic pattern can be distinguished. The acute phase of critical illness is characterized by low peripheral effector hormone levels such as T3, IGF-1 and testosterone, despite an actively secreting pituitary. The adrenal axis with high cortisol levels in the presence of low ACTH levels is a noteworthy exception. In the prolonged phase of critical illness, low peripheral effector hormone levels coincide with a uniform suppression of the neuroendocrine axes, predominantly of hypothalamic origin. The severity of the alterations in the different neuroendocrine axes is associated with a high risk of morbidity and mortality, but it remains unknown whether the observed changes are cause or consequence of adverse outcome. Several studies have identified therapeutic potential of hypothalamic releasing factors, but clinical outcome remains to be investigated with sufficiently powered randomized controlled trials.
Collapse
Affiliation(s)
- Lies Langouche
- Laboratory and Department of Intensive Care Medicine, University of Leuven, Leuven, Belgium.
| | - Greet Van den Berghe
- Laboratory and Department of Intensive Care Medicine, University of Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Dietrich JW, Landgrafe G, Fotiadou EH. TSH and Thyrotropic Agonists: Key Actors in Thyroid Homeostasis. J Thyroid Res 2012; 2012:351864. [PMID: 23365787 PMCID: PMC3544290 DOI: 10.1155/2012/351864] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 11/21/2012] [Indexed: 12/11/2022] Open
Abstract
This paper provides the reader with an overview of our current knowledge of hypothalamic-pituitary-thyroid feedback from a cybernetic standpoint. Over the past decades we have gained a plethora of information from biochemical, clinical, and epidemiological investigation, especially on the role of TSH and other thyrotropic agonists as critical components of this complex relationship. Integrating these data into a systems perspective delivers new insights into static and dynamic behaviour of thyroid homeostasis. Explicit usage of this information with mathematical methods promises to deliver a better understanding of thyrotropic feedback control and new options for personalised diagnosis of thyroid dysfunction and targeted therapy, also by permitting a new perspective on the conundrum of the TSH reference range.
Collapse
Affiliation(s)
- Johannes W. Dietrich
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
| | - Gabi Landgrafe
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
- Klinik für Allgemein- und Visceralchirurgie, Agaplesion Bethesda Krankenhaus Wuppertal gGmbH, Hainstraße 35, 42109 Wuppertal, NRW, Germany
| | - Elisavet H. Fotiadou
- Lab XU44, Medical Hospital I, Bergmannsheil University Hospitals, Ruhr University of Bochum (UK RUB), Bürkle-de-la-Camp-Platz 1, 44789 Bochum, NRW, Germany
| |
Collapse
|
24
|
Heuer H, Visser TJ. The pathophysiological consequences of thyroid hormone transporter deficiencies: Insights from mouse models. Biochim Biophys Acta Gen Subj 2012; 1830:3974-8. [PMID: 22543196 DOI: 10.1016/j.bbagen.2012.04.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/04/2012] [Accepted: 04/11/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND As a prerequisite for thyroid hormone (TH) metabolism and action TH has to be transported into cells where TH deiodinases and receptors are located. The trans-membrane passage of TH is facilitated by TH transporters of which the monocarboxylate transporter MCT8 has been most intensively studied. Inactivating mutations in the gene encoding MCT8 are associated with a severe form of psychomotor retardation and abnormal serum TH levels (Allan-Herndon-Dudley syndrome). In order to define the underlying pathogenic mechanisms, Mct8 knockout mice have been generated and intensively studied. Most surprisingly, Mct8 ko mice do not show any neurological symptoms but fully replicate the abnormal serum thyroid state. SCOPE OF REVIEW We will summarize the findings of these mouse studies that shed light on various aspects of Mct8 deficiency and unambiguously demonstrated the pivotal role of Mct8 in mediating TH transport in various tissues. These studies have also revealed the presence of the complex interplay between different pathogenic mechanisms that contribute to the generation of the abnormal TH serum profile. MAJOR CONCLUSIONS Most importantly, studies of Mct8 ko mice indicated the presence of additional TH transporters that act in concert with Mct8. Interesting candidates for such a function are the L-type amino acid transporters Lat1 and Lat2 as well as the organic anion transporting polypeptide Oatp1c1. GENERAL SIGNIFICANCE Overall, the analysis of Mct8 deficient mice has greatly expanded our knowledge about the (patho-) physiological function of this transporter and established a sound basis for the characterization of additional TH transporter candidates. This article is part of a Special Issue entitled Thyroid hormone signalling.
Collapse
Affiliation(s)
- Heike Heuer
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
| | | |
Collapse
|
25
|
Raldúa D, Thienpont B, Babin PJ. Zebrafish eleutheroembryos as an alternative system for screening chemicals disrupting the mammalian thyroid gland morphogenesis and function. Reprod Toxicol 2012; 33:188-97. [DOI: 10.1016/j.reprotox.2011.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 08/16/2011] [Accepted: 09/08/2011] [Indexed: 01/09/2023]
|
26
|
Tóth G, Hosztafi S, Kovács Z, Noszál B. The site-specific basicity of thyroid hormones and their precursors as regulators of their biological functions. J Pharm Biomed Anal 2012; 61:156-64. [DOI: 10.1016/j.jpba.2011.11.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Revised: 11/18/2011] [Accepted: 11/20/2011] [Indexed: 10/14/2022]
|
27
|
Arjona FJ, de Vrieze E, Visser TJ, Flik G, Klaren PHM. Identification and functional characterization of zebrafish solute carrier Slc16a2 (Mct8) as a thyroid hormone membrane transporter. Endocrinology 2011; 152:5065-73. [PMID: 21952246 DOI: 10.1210/en.2011-1166] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Most components of the thyroid system in bony fish have been described and characterized, with the notable exception of thyroid hormone membrane transporters. We have cloned, sequenced, and expressed the zebrafish solute carrier Slc16a2 (also named monocarboxylate transporter Mct8) cDNA and established its role as a thyroid hormone transport protein. The cloned cDNA shares 56-57% homology with its mammalian orthologs. The 526-amino-acid sequence contains 12 predicted transmembrane domains. An intracellular N-terminal PEST domain, thought to be involved in proteolytic processing of the protein, is present in the zebrafish sequence. Measured at initial rate and at the body/rearing temperature of zebrafish (26 C), T(3) uptake by zebrafish Slc16a2 is a saturable process with a calculated Michaelis-Menten constant of 0.8 μM T(3). The rate of T(3) uptake is temperature dependent and Na(+) independent. Interestingly, at 26 C, zebrafish Slc16a2 does not transport T(4). This implies that at a normal body temperature in zebrafish, Slc16a2 protein is predominantly involved in T(3) uptake. When measured at 37 C, zebrafish Slc16a2 transports T(4) in a Na(+)-independent manner. In adult zebrafish, the Slc16a2 gene is highly expressed in brain, gills, pancreas, liver, pituitary, heart, kidney, and gut. Beginning from the midblastula stage, Slc16a2 is also expressed during zebrafish early development, the highest expression levels occurring 48 h after fertilization. This is the first direct evidence for thyroid hormone membrane transporters in fish. We suggest that Slc16a2 plays a key role in the local availability of T(3) in adult tissues as well as during the completion of morphogenesis of primary organ systems.
Collapse
Affiliation(s)
- Francisco J Arjona
- Department of Organismal Animal Physiology, Institute for Water and Wetland Research, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
28
|
Abstract
Thyroid hormones (TH) are essential for the development of the human brain, growth and cellular metabolism. Investigation of TH transporters became one of the emerging fields in thyroid research after the discovery of inactivating mutations in the Monocarboxylate transporter 8 (MCT8), which was found to be highly specific for TH transport. However, additional transmembrane transporters are also very important for TH uptake and efflux in different cell types. They transport TH as secondary substrates and include the aromatic amino acid transporting MCT10, the organic anion transporting polypeptides (e.g. OATP1C1, OATP1A2, OPTP1A4) and the large neutral amino acid transporters (LAT1 and LAT2). These TH transporters characteristically possess 12 transmembrane spanners but due to the strong differing sequences between the three transporter families we assume an identical conformation is not very likely. In contrast to the others, the LAT family members form a heterodimer with the escort protein 4F2hc/CD98. A comparison of sequence proportions, locations and types of functional sensitive features for TH transport discovered by mutations, revealed that transport sensitive charged residues occur as conserved amino acids only within each family of the transporter types but not in all putative TH transporters. Based on the lack of highly conserved sensitive charged residues throughout the three transporter families as a common counterpart for the amino acid moiety of the substrates, we conclude that the molecular transport mechanism is likely organized either a) by different molecular determinants in the divergent transporter types or b) the counterparts for the substrates` amino acid moiety at the transporter are not any charged side chains but other proton acceptors or donators. However, positions of transport sensitive residues coincide at transmembrane helix 8 in the TH transporter MCT8, OATP1C1 and another amino acid transporter, the L-cystine and L-glutamate exchanger xCT, which is highly homologous to LAT1 and LAT2. Here we review the data available and compare similarities and differences between these primary and secondary TH transporters regarding sequences, topology, potential structures, trafficking to the plasma membrane, molecular features and locations of transport sensitive functionalities. Thereby, we focus on TH transporters occurring in the blood-brain barrier.
Collapse
Affiliation(s)
- Anita Kinne
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Roessle-Str, 10, 13125 Berlin, Germany.
| | | | | |
Collapse
|
29
|
Ahmed R. Perinatal TCDD exposure alters developmental neuroendocrine system. Food Chem Toxicol 2011; 49:1276-84. [DOI: 10.1016/j.fct.2011.03.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 02/26/2011] [Accepted: 03/09/2011] [Indexed: 10/18/2022]
|
30
|
A possible mechanism for 2,2',4,4',5,5'-hexachlorobiphenyl-mediated decrease in serum thyroxine level in mice. Toxicol Appl Pharmacol 2011; 254:48-55. [PMID: 21569787 DOI: 10.1016/j.taap.2011.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 04/23/2011] [Accepted: 04/25/2011] [Indexed: 11/22/2022]
Abstract
Serum total thyroxine (T₄) level was markedly decreased, without significant increases in the levels of hepatic T₄-UDP-glucuronosyltransferase (T₄-UGT) and serum thyroid-stimulating hormone, 3 days after treatment with 2,2',4,4',5,5'-hexachlorobiphenyl (CB153) (100mg/kg, ip) in both 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-sensitive C57BL/6 and TCDD-resistant DBA/2 mice. Likewise, in either strain of mice, no CB153-mediated changes in the binding levels of [(125)I]T₄ to serum proteins, such as transthyretin, albumin, and thyroxine binding globulin, were observed, while in CB153-pretreated C57BL/6 mice, but not in CB153-pretreated DBA/2 mice, the levels of biliary [(125)I]₄T and [(125)I]T₄-glucuronide at 90-120 min after injection of [(125)I]T₄ slightly increased, as compared with those in the corresponding control mice. Concerning tissue distribution of [(125)I]T₄, liver-selective increases in the [(125)I]T₄ accumulation by CB153-pretreatment were observed in both C57BL/6 and DBA/2 mice, and the hepatic levels of [(125)I]T₄ in the C57BL/6 and DBA/2 mice became more than 44% and 34% of the [(125)I]T₄ dosed, respectively. The present findings indicated that the CB153-mediated decreases in the level of serum total T₄in C57BL/6 and DBA/2 mice occur mainly through an increase in the accumulation of T₄ in the liver.
Collapse
|
31
|
Ianculescu AG, Friesema ECH, Visser TJ, Giacomini KM, Scanlan TS. Transport of thyroid hormones is selectively inhibited by 3-iodothyronamine. MOLECULAR BIOSYSTEMS 2010; 6:1403-10. [PMID: 20358049 PMCID: PMC3257980 DOI: 10.1039/b926588k] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Thyroid hormone transporters are responsible for the cellular uptake of thyroid hormones, which is a prerequisite for their subsequent metabolism and action at nuclear thyroid hormone receptors. A recently discovered thyroid hormone derivative, 3-iodothyronamine (T(1)AM), has distinct biological effects that are opposite those of thyroid hormone. Here we investigate the effects of T(1)AM on thyroid hormone transporters using COS-1 cells transfected with the multispecific organic anion transporting polypeptides (OATPs) 1A2, 1B3, and 1C1, as well as the specific thyroid hormone transporters MCT8 and MCT10, and show that T(1)AM displays differential inhibition of T(3) and T(4) cellular uptake by these transporters. T(1)AM inhibits T(3) and T(4) transport by OATP1A2 with IC(50) values of 0.27 and 2.1 microM, respectively. T(4) transport by OATP1C1, which is thought to play a key role in thyroid hormone transport across the blood-brain barrier, is inhibited by T(1)AM with an IC(50) of 4.8 microM. T(1)AM also inhibits both T(3) and T(4) uptake via MCT8, the most specific thyroid hormone transporter identified to date, with IC(50) values of 95 and 31 microM, respectively. By contrast, T(1)AM has no effect on thyroid hormone transport by OATP1B3 and MCT10. Given that OATP1A2, OATP1C1, and MCT8 are all present in the brain, T(1)AM may play an important role in modulating thyroid hormone delivery and activity in specific target regions in the central nervous system.
Collapse
Affiliation(s)
- Alexandra G. Ianculescu
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California, USA
| | - Edith C. H. Friesema
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, The Netherlands
| | - Theo J. Visser
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, The Netherlands
| | - Kathleen M. Giacomini
- Department of Biopharmaceutical Sciences, University of California at San Francisco, San Francisco, California, USA
| | - Thomas S. Scanlan
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
32
|
Richardson TA, Klaassen CD. Disruption of thyroid hormone homeostasis in Ugt1a-deficient Gunn rats by microsomal enzyme inducers is not due to enhanced thyroxine glucuronidation. Toxicol Appl Pharmacol 2010; 248:38-44. [PMID: 20655938 DOI: 10.1016/j.taap.2010.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 07/10/2010] [Accepted: 07/14/2010] [Indexed: 11/25/2022]
Abstract
Microsomal enzyme inducers (MEI) that increase UDP-glucuronosyltransferases (UGTs) are thought to increase glucuronidation of thyroxine (T(4)), thus reducing serum T(4), and subsequently increasing thyroid stimulating hormone (TSH). Ugt1a1 and Ugt1a6 mediate T(4) glucuronidation. Therefore, this experiment determined the involvement of Ugt1a enzymes in increased T(4) glucuronidation, decreased serum T(4), and increased TSH after MEI treatment. Male Wistar and Ugt1a-deficient Wistar (Gunn) rats were fed a control diet or diet containing pregnenolone-16α-carbonitrile (PCN; 800 ppm), 3-methylcholanthrene (3-MC; 200 ppm), or Aroclor 1254 (PCB; 100 ppm) for 7 days. Serum T(4), triiodothyronine (T(3)), and TSH concentrations, hepatic T(4)/T(3) glucuronidation, and thyroid histology and follicular cell proliferation were investigated. PCN, 3-MC, and PCB treatments decreased serum T(4), whereas serum T(3) was maintained in both Gunn and Wistar rats (except for PCB treatment). TSH was increased in Wistar and Gunn rats after PCN (130 and 277%) or PCB treatment (72 and 60%). T(4) glucuronidation in Wistar rats was increased after PCN (298%), 3-MC (85%), and PCB (450%), but was extremely low in Gunn rats, and unchanged after MEI. T(3) glucuronidation was increased after PCN (121%) or PCB (58%) in Wistar rats, but only PCN increased T(3) glucuronidation in Gunn rats (43%). PCN treatment induced thyroid morphological changes and increased follicular cell proliferation in both strains. These data demonstrate that T(4) glucuronidation cannot be increased in Ugt1a-deficient Gunn rats. Thus, the decrease in serum T(4), increase in TSH, and increase in thyroid cell proliferation after MEI are not dependent on increased T(4) glucuronidation, and cannot be attributed to Ugt1a enzymes.
Collapse
Affiliation(s)
- Terrilyn A Richardson
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | |
Collapse
|
33
|
Ucán-Marin F, Arukwe A, Mortensen AS, Gabrielsen GW, Letcher RJ. Recombinant albumin and transthyretin transport proteins from two gull species and human: chlorinated and brominated contaminant binding and thyroid hormones. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2010; 44:497-504. [PMID: 20039755 DOI: 10.1021/es902691u] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Environmentally relevant concentrations of selected polychlorinated biphenyl (PCB) and polybrominated diphenyl ether (PBDE) flame retardant congeners and their hydroxylated (OH) and methoxylated (MeO) analogues that can perturb thyroid hormone-dependent processes were comparatively examined with respect to competitive binding with thyroxine (T(4)) and 3,5,3'-triiodothyronine (T(3)) thyroid hormones (THs) on recombinant human and gull albumin and transthyretin transport proteins. The liver tissue was from glaucous gulls (Larus hyperboreus) from Norway and herring gulls (Larus argentatus) from the Great Lakes of North America. We isolated, cloned, sequenced, purified, and expressed the cDNA (cDNA) of albumin from liver of herring and glaucous gull. Albumin amino acid sequences were identical for both gull species. Concentration-dependent, competitive binding curves were generated for T(4) and T(3) binding alone and for selected substrates using gull and human recombinant albumin (recALB). Human recALB had high preference for T(4) relative to T(3), whereas it was reversed for gull recALB. Binding assays with recALB and recTTR gull proteins showed that relative to 2,2',4,4'-tetrabromoDE (BDE-47) and 2,2',3,4',5,5',6-heptaCB (CB-187) and the MeO-substituted (4-MeO-CB187 and 6-MeO-BDE47) analogues, 4-OH-CB187, 6-OH-BDE47, and 4'-OH-BDE49 had the greatest binding affinity and potency, and that competitive binding was greater for T(3) relative to T(4). These results indicate that xenobiotic ligand binding to human ALB or TTR cannot be used as a surrogate for gull binding interactions. The combination of TH-like brominated diphenyl ether backbone (relative to the chlorinated biphenyl backbone), and the presence of OH-group produced a more effective competitive ligand on human and gull recALB and recTTR relative to both T(3) and T(4). This suggests the possibility that OH-substituted organohalogen contaminants may be an exposure concern to the thyroid system in free-ranging gulls as well as for humans.
Collapse
Affiliation(s)
- Francisco Ucán-Marin
- Wildlife and Landscape Science Directorate, Science and Technology Branch, Environment Canada, National Wildlife Research Centre, Carleton University, Ottawa, ON, K1A 0H3, Canada
| | | | | | | | | |
Collapse
|
34
|
Lago-Lestón R, Iglesias MJ, San-José E, Areal C, Eiras A, Araújo-Vilar D, Lado-Abeal J, Domínguez-Gerpe L. Prevalence and functional analysis of the S107P polymorphism (rs6647476) of the monocarboxylate transporter 8 (SLC16A2) gene in the male population of north-west Spain (Galicia). Clin Endocrinol (Oxf) 2009; 70:636-43. [PMID: 18710470 DOI: 10.1111/j.1365-2265.2008.03377.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Mutations in SLC16A2, the gene encoding the thyroid hormone (TH)-specific transporter monocarboxylate transporter 8 (MCT8), result in a thyroid phenotype and severe mental retardation caused by neuronal TH deficiency. These mutational effects raise the question of whether polymorphic variation in SLC16A2 may also be associated with differences in serum levels of TH and/or TSH. DESIGN This is the first major study of the frequency of the SLC16A2 rs6647476 single nucleotide polymorphism (SNP) (amino acid change Ser107Pro). We also studied the relationships of SLC16A2 genetic variants with serum levels of TSH, T4 and T3, with their mRNA expression and with expression of the TH-responsive genes ZAKI-4 and BTEB in white blood cells. Experiments in cultured fibroblasts were carried out to ascertain the dynamics of the T3 response. METHODS A total of 276 men were studied. Genotyping of the S107P SNP was carried out using polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP); serum hormone levels were determined by chemiluminescence; expression of mRNA was quantified by real-time PCR. RESULTS The SLC16A2 S107P SNP was found in 36% of Galician males. With the present sample size we did not find any association of this polymorphism with variability in serum levels of TSH, free T4 (fT4) or fT3, or with basal expression of mRNA for SLC16A2 or the two TH-responsive genes ZAKI-4 and BTEB, either in white blood cells or in cultured human fibroblasts from either Ser107 or Pro107 genotypes under T3 stimulation. CONCLUSIONS The S107P change in MCT8 is frequent in the male population in Galicia. In the population studied in this report an association with a thyroid phenotype was not demonstrated, even though the S107P SNP causes an important amino acid change.
Collapse
Affiliation(s)
- Ramón Lago-Lestón
- Departamento de Medicina, Unidade de Enfermedades Tiroideas e Metabólicas, Universidad de Santiago de Compostela, Spain
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Ianculescu AG, Giacomini KM, Scanlan TS. Identification and characterization of 3-iodothyronamine intracellular transport. Endocrinology 2009; 150:1991-9. [PMID: 19074582 PMCID: PMC2659270 DOI: 10.1210/en.2008-1339] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
3-Iodothyronamine (T(1)AM) is a naturally occurring thyroid hormone metabolite with distinct biological effects that are opposite those of thyroid hormone. The known molecular targets of T(1)AM include both plasma membrane and intracellular proteins, suggesting that intracellular transport of T(1)AM may be an important component of its action, although no uptake mechanism has yet been described. Using various human cell lines, we show that, indeed, cellular uptake of T(1)AM occurs in multiple cell types and that this process involves specific, saturable, and inhibitable transport mechanisms. These mechanisms are sodium and chloride independent, pH dependent, thyronamine specific, and do not involve the likely candidate transporters of other monoamines, organic cations, or thyroid hormones. A large-scale RNA interference screen targeting the entire solute carrier superfamily of transporter genes reveals that the transport of T(1)AM into cells involves multiple transporters, and we identify eight transporters that may contribute to the uptake of T(1)AM in HeLa cells. This type of transporter small interfering RNA screening approach can be used in general to identify the constellation of transporters that participate in the intracellular disposition of compounds.
Collapse
Affiliation(s)
- Alexandra G Ianculescu
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California 94143, USA
| | | | | |
Collapse
|
36
|
Fuchs O, Pfarr N, Pohlenz J, Schmidt H. Elevated serum triiodothyronine and intellectual and motor disability with paroxysmal dyskinesia caused by a monocarboxylate transporter 8 gene mutation. Dev Med Child Neurol 2009; 51:240-4. [PMID: 19018842 DOI: 10.1111/j.1469-8749.2008.03125.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Monocarboxylate transporter 8 (MCT8 or SLC16A2) is important for the neuronal uptake of triiodothyronine (T3) in its function as a specific and active transporter of thyroid hormones across the cell membrane, thus being essential for human brain development. We report on a German male with Allan-Herndon-Dudley syndrome presenting with severe intellectual and motor disability, paroxysmal dyskinesia combined with truncal muscular hypotonia, and peripheral muscular hypertonia at his current age of 9 years. Additionally, the patient has a lesion in the left putamen region revealed by magnetic resonance imaging and elevated serum T3 levels. The male appeared to have a hemizygous mutation (R271H) in the MCT8 gene that was sequenced directly from genomic DNA and occurred de novo in the maternal germline, as both his mother and his sister were not carriers of the mutation. Ruling out a common polymorphism, 50 normal individuals of the same ethnic background did not harbour the mutation. The identified MCT8 gene mutation (R271H) is very likely to be the genetic cause for neuronal hypothyroidism despite elevated serum T3 levels.
Collapse
Affiliation(s)
- Oliver Fuchs
- Dr von Hauner University Children's Hospital, Pediatric Endocrinology, Munich, Germany
| | | | | | | |
Collapse
|
37
|
Davis PJ, Davis FB, Mousa SA. Thyroid hormone-induced angiogenesis. Curr Cardiol Rev 2009; 5:12-6. [PMID: 20066142 PMCID: PMC2803282 DOI: 10.2174/157340309787048158] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Revised: 07/24/2008] [Accepted: 07/24/2008] [Indexed: 11/22/2022] Open
Abstract
A series of reports in the past decade have ascribed pro-angiogenic activity to several thyroid hormone analogues, including L-thyroxine (T(4)), 3,5,3-triiodo-L-thyronine (T(3)) and diiodothyropropionic acid (DITPA). Model systems of angiogenesis have demonstrated that thyroid hormone-induced neovascularization is initiated at a cell surface receptor for the hormone on an integrin. The hormone signal is transduced within the cell by extracellular regulated kinase 1/2 (ERK1/2) into secretion of basic fibroblast growth factor (bFGF) and other vascular growth factors and consequent angiogenesis. Intact animal studies have shown that endogenous thyroid hormone supports blood vessel density in heart and brain and that thyroid hormone administration can induce angiogenesis in ischemic limbs.
Collapse
Affiliation(s)
- Paul J Davis
- Address for correspondence to this author at the Signal Transduction Laboratory, Ordway Research Institute, Inc., 150 New Scotland Avenue, Albany, NY 12208 USA; Tel: 518 641 6410; Fax: 518 641 6303; E-mail:
| | | | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy, Albany, NY, USAOrdway Research Institute, Inc., Albany, New York
| |
Collapse
|
38
|
Marchesini GR, Meimaridou A, Haasnoot W, Meulenberg E, Albertus F, Mizuguchi M, Takeuchi M, Irth H, Murk AJ. Biosensor discovery of thyroxine transport disrupting chemicals. Toxicol Appl Pharmacol 2008; 232:150-60. [DOI: 10.1016/j.taap.2008.06.014] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Revised: 06/16/2008] [Accepted: 06/25/2008] [Indexed: 11/29/2022]
|
39
|
Abstract
Thyroid hormones play a critical role in the metabolic activity of the adult brain, and neuropsychiatric manifestations of thyroid disease have long been recognised. However, it is only recently that methodology such as functional neuroimaging has been available to facilitate investigation of thyroid hormone metabolism. Although the role of thyroid hormones in the adult brain is not yet specified, it is clear that without optimal thyroid function, mood disturbance, cognitive impairment and other psychiatric symptoms can emerge. Additionally, laboratory measurements of peripheral thyroid function may not adequately characterise central thyroid metabolism. Here, we review the relationship between thyroid hormone and neuropsychiatric symptoms in patients with primary thyroid disease and primary mood disorders.
Collapse
Affiliation(s)
- M Bauer
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | | | | | | |
Collapse
|
40
|
Namba N, Etani Y, Kitaoka T, Nakamoto Y, Nakacho M, Bessho K, Miyoshi Y, Mushiake S, Mohri I, Arai H, Taniike M, Ozono K. Clinical phenotype and endocrinological investigations in a patient with a mutation in the MCT8 thyroid hormone transporter. Eur J Pediatr 2008; 167:785-91. [PMID: 17899191 DOI: 10.1007/s00431-007-0589-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Accepted: 08/07/2007] [Indexed: 11/29/2022]
Abstract
UNLABELLED Thyroid hormones are known to be essential for growth, development, and metabolism. Recently, the monocarboxylate transporter 8 (MCT8) was identified as a thyroid hormone transporter, and MCT8 mutations have been associated with Allan-Herndon-Dudley syndrome, an X linked condition characterized by severe mental retardation, dysarthria, athetoid movements, muscle hypoplasia, and spastic paraplegia. Here we describe in detail the clinical and biochemical features and the response to thyroid hormone (L-thyroxine (LT4)) administration in a boy with an MCT8 mutation (c.1649delA) that truncates the protein in the twelfth transmembrane domain. It is of note that brain magnetic resonance imaging (MRI) revealed delayed myelination from infancy. Endocrine functions other than thyroid hormone regulation and metabolism were intact, resulting in normal hypothalamic/pituitary function tests. While LT4 administration suppressed thyrotropin (TSH) secretion, no significant changes in thyroid hormone values or clinical symptoms were observed. CONCLUSION the characteristic thyroid hormone function tests and brain MRI findings may allow screening of high-risk populations for a better understanding of MCT8 pathophysiology.
Collapse
Affiliation(s)
- Noriyuki Namba
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Seasonal rhythms of body weight, reflecting altered food intake, energy storage and expenditure, are a common feature of mammals inhabiting temperate and arctic latitudes. They have evolved so that predictable annual changes in the external environment can be anticipated and animals can adjust their physiology and behaviour in preparation for the changing demands of the seasons. These long-term changes in energy balance are not simply effected by the brain centres and peptidergic pathways known to underlie short-term homeostatic regulation. Screens of altered gene expression in Siberian hamsters comparing the anabolic summer state in long photoperiods and the catabolic 'winter' state in short photoperiods have identified differential gene expression in the hypothalamus. The majority of gene expression changes are confined to two restricted areas: the dorsomedial posterior arcuate nucleus, and the ventral ependymal layer of the third ventricle. Functions encoded by these 'seasonal' genes include thyroid hormone metabolism, retinoic acid and histaminergic signalling, and VGF and secretogranin production. The changes in thyroid hormone availability that are brought about by differential activity of deiodinase enzymes are of particular importance because experimental manipulation of central thyroid levels can prevent seasonal cyclicity. Given the importance of thyroid hormone in the initial development of the brain, we hypothesise that thyroid hormone-dependent plasticity of hypothalamic connections and neurogenesis underlie seasonal cycles of food intake and body weight.
Collapse
Affiliation(s)
- F J P Ebling
- School of Biomedical Sciences and Institute of Neuroscience, University of Nottingham Medical School, Nottingham, UK
| | | |
Collapse
|
42
|
Hawkes WC, Keim NL, Diane Richter B, Gustafson MB, Gale B, Mackey BE, Bonnel EL. High-selenium yeast supplementation in free-living North American men: no effect on thyroid hormone metabolism or body composition. J Trace Elem Med Biol 2008; 22:131-42. [PMID: 18565425 DOI: 10.1016/j.jtemb.2007.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Revised: 10/05/2007] [Accepted: 11/29/2007] [Indexed: 11/30/2022]
Abstract
In a prior study, we observed decreased serum 3,3',5-triiodothyronine (T(3)), increased serum thyrotropin and increased body weight in five men fed 297 microg/d of selenium (Se) in foods naturally high in Se while confined in a metabolic research unit. In an attempt to replicate and confirm those observations, we conducted a randomized study of high-Se yeast supplements (300 microg/d) or placebo yeast administered to 42 healthy free-living men for 48 weeks. Serum thyroxine, T(3) and thyrotropin did not change in supplemented or control subjects. Body weight increased in both groups during the 48-week treatment period and remained elevated for the 48-week follow-up period. Body fat increased by 1.2 kg in both groups. Energy intake and voluntary activity levels were not different between the groups and remained unchanged during the treatment period. Dietary intakes of Se, macronutrients and micronutrients were not different between groups and remained unchanged during the treatment period. These results suggest that our previous observation of a hypothyroidal response to high-Se foods was confounded by some aspect of the particular foods used, or were merely chance observations. Because of the high dose and long administration period, the present study suggests that the effects of Se supplements on thyroid hormone metabolism and energy metabolism in healthy North American men with adequate Se status do not represent a significant risk for unhealthy weight gain.
Collapse
Affiliation(s)
- Wayne Chris Hawkes
- Western Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, University of California at Davis, 430 West Health Sciences Drive, Davis, CA 95616, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Haase H, Mazzatti DJ, White A, Ibs KH, Engelhardt G, Hebel S, Powell JR, Rink L. Differential gene expression after zinc supplementation and deprivation in human leukocyte subsets. Mol Med 2007. [PMID: 17622302 DOI: 10.2119/2007-00049.haase] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
An individual's zinc status has a significant impact on the immune system, and zinc deficiency, as well as supplementation, modulates immune function. To investigate the effects of zinc on different leukocyte subsets, we used microarray technology to analyze and compare the changes in mRNA expression in cell culture models of monocytes (THP-1), T cells (Jurkat), and B cells (Raji), in response to supplementation for 40 h with 50 microM zinc or 2.5 microM of the membrane-permeant zinc chelator TPEN [N,N,N',N'-tetrakis-(2-pyridyl-methyl)ethylenediamine], respectively. In each cell type, several hundred genes were identified to be zinc sensitive, but only a total of seven genes were commonly regulated in all three cell lines. The majority of those genes were involved in zinc homeostasis, and none in immune function. Nevertheless, further analysis revealed that zinc affects entire functional networks of genes that are related to proinflammatory cytokines and cellular survival. Although the zinc-regulated activities are similar throughout the gene networks, the specific genes that are affected vary significantly between different cell types, a situation that helps to elucidate the disparity of the effects that zinc has on different leukocyte populations.
Collapse
Affiliation(s)
- Hajo Haase
- Institute of Immunology, University Hospital, RWTH Aachen University, Germany
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Erion MD, Cable EE, Ito BR, Jiang H, Fujitaki JM, Finn PD, Zhang BH, Hou J, Boyer SH, van Poelje PD, Linemeyer DL. Targeting thyroid hormone receptor-beta agonists to the liver reduces cholesterol and triglycerides and improves the therapeutic index. Proc Natl Acad Sci U S A 2007; 104:15490-5. [PMID: 17878314 PMCID: PMC1978486 DOI: 10.1073/pnas.0702759104] [Citation(s) in RCA: 173] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Despite efforts spanning four decades, the therapeutic potential of thyroid hormone receptor (TR) agonists as lipid-lowering and anti-obesity agents remains largely unexplored in humans because of dose-limiting cardiac effects and effects on the thyroid hormone axis (THA), muscle metabolism, and bone turnover. TR agonists selective for the TRbeta isoform exhibit modest cardiac sparing in rodents and primates but are unable to lower lipids without inducing TRbeta-mediated suppression of the THA. Herein, we describe a cytochrome P450-activated prodrug of a phosphonate-containing TR agonist that exhibits increased TR activation in the liver relative to extrahepatic tissues and an improved therapeutic index. Pharmacokinetic studies in rats demonstrated that the prodrug (2R,4S)-4-(3-chlorophenyl)-2-[(3,5-dimethyl-4-(4'-hydroxy-3'-isopropylbenzyl)phenoxy)methyl]-2-oxido-[1,3,2]-dioxaphosphonane (MB07811) undergoes first-pass hepatic extraction and that cleavage of the prodrug generates the negatively charged TR agonist (3,5-dimethyl-4-(4'-hydroxy-3'-isopropylbenzyl)phenoxy)methylphosphonic acid (MB07344), which distributes poorly into most tissues and is rapidly eliminated in the bile. Enhanced liver targeting was further demonstrated by comparing the effects of MB07811 with 3,5,3'-triiodo-l-thyronine (T(3)) and a non-liver-targeted TR agonist, 3,5-dichloro-4-(4-hydroxy-3-isopropylphenoxy)phenylacetic acid (KB-141) on the expression of TR agonist-responsive genes in the liver and six extrahepatic tissues. The pharmacologic effects of liver targeting were evident in the normal rat, where MB07811 exhibited increased cardiac sparing, and in the diet-induced obese mouse, where, unlike KB-141, MB07811 reduced cholesterol and both serum and hepatic triglycerides at doses devoid of effects on body weight, glycemia, and the THA. These results indicate that targeting TR agonists to the liver has the potential to lower both cholesterol and triglyceride levels with an acceptable safety profile.
Collapse
Affiliation(s)
- Mark D Erion
- Metabasis Therapeutics, Inc., 11119 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
A class of thyroid hormone metabolites has dramatic physiological effects on metabolism and heart rate by still-unknown mechanisms of action. A recent study has discovered that thyronamines can inhibit neuronal reuptake of neurotransmitters and prevent the intracellular transport of monoamines for release. This discovery presents a third signaling pathway for thyroid hormone, expands the role that thyroid plays in the central nervous system, and suggests mechanisms of action for the effects of thyronamine-derived neuromodulators.
Collapse
Affiliation(s)
- Ross V Weatherman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, USA.
| |
Collapse
|
46
|
Abstract
Thyroid hormones are vital for fetal development and can act directly on placental tissues to modify their metabolism, differentiation and development. There is evidence that maternal thyroid hormones can cross the human placenta and act to modulate fetal development before the onset of the fetus's own thyroid hormone production. Plasma membrane transport of thyroid hormones has now been shown to require specific transporter proteins. Several proteins have recently been identified as specific thyroid hormone transporters. However, as yet few data are available to define the functionally important transporter proteins in the human placenta. To date, members of the organic anion-transporting polypeptide, L-type amino acid, and of the monocarboxylate transporter families, have been identified as thyroid hormone transporters that are active in a variety of placental cell types. However, further research is necessary to determine the role of these and other proteins in placental transport of thyroid hormone, and to investigate how modulations of their function could affect fetal pathologies such as intrauterine growth restriction.
Collapse
Affiliation(s)
- Sally R James
- Division of Reproductive and Child Health, University of Birmingham, Birmingham, UK
| | | | | |
Collapse
|
47
|
Trajkovic M, Visser TJ, Mittag J, Horn S, Lukas J, Darras VM, Raivich G, Bauer K, Heuer H. Abnormal thyroid hormone metabolism in mice lacking the monocarboxylate transporter 8. J Clin Invest 2007; 117:627-35. [PMID: 17318265 PMCID: PMC1797602 DOI: 10.1172/jci28253] [Citation(s) in RCA: 260] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2006] [Accepted: 01/02/2007] [Indexed: 11/17/2022] Open
Abstract
In humans, inactivating mutations in the gene of the thyroid hormone transporter monocarboxylate transporter 8 (MCT8; SLC16A2) lead to severe forms of psychomotor retardation combined with imbalanced thyroid hormone serum levels. The MCT8-null mice described here, however, developed without overt deficits but also exhibited distorted 3,5,3'-triiodothyronine (T3) and thyroxine (T4) serum levels, resulting in increased hepatic activity of type 1 deiodinase (D1). In the mutants' brains, entry of T4 was not affected, but uptake of T3 was diminished. Moreover, the T4 and T3 content in the brain of MCT8-null mice was decreased, the activity of D2 was increased, and D3 activity was decreased, indicating the hypothyroid state of this tissue. In the CNS, analysis of T3 target genes revealed that in the mutants, the neuronal T3 uptake was impaired in an area-specific manner, with strongly elevated thyrotropin-releasing hormone transcript levels in the hypothalamic paraventricular nucleus and slightly decreased RC3 mRNA expression in striatal neurons; however, cerebellar Purkinje cells appeared unaffected, since they did not exhibit dendritic outgrowth defects and responded normally to T3 treatment in vitro. In conclusion, the circulating thyroid hormone levels of MCT8-null mice closely resemble those of humans with MCT8 mutations, yet in the mice, CNS development is only partially affected.
Collapse
Affiliation(s)
- Marija Trajkovic
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
Max Planck Institute for Experimental Endocrinology, Hannover, Germany.
Laboratory of Comparative Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium.
Perinatal Brain Repair Group, University College London, London, United Kingdom
| | - Theo J. Visser
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
Max Planck Institute for Experimental Endocrinology, Hannover, Germany.
Laboratory of Comparative Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium.
Perinatal Brain Repair Group, University College London, London, United Kingdom
| | - Jens Mittag
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
Max Planck Institute for Experimental Endocrinology, Hannover, Germany.
Laboratory of Comparative Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium.
Perinatal Brain Repair Group, University College London, London, United Kingdom
| | - Sigrun Horn
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
Max Planck Institute for Experimental Endocrinology, Hannover, Germany.
Laboratory of Comparative Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium.
Perinatal Brain Repair Group, University College London, London, United Kingdom
| | - Jan Lukas
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
Max Planck Institute for Experimental Endocrinology, Hannover, Germany.
Laboratory of Comparative Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium.
Perinatal Brain Repair Group, University College London, London, United Kingdom
| | - Veerle M. Darras
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
Max Planck Institute for Experimental Endocrinology, Hannover, Germany.
Laboratory of Comparative Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium.
Perinatal Brain Repair Group, University College London, London, United Kingdom
| | - Genadij Raivich
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
Max Planck Institute for Experimental Endocrinology, Hannover, Germany.
Laboratory of Comparative Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium.
Perinatal Brain Repair Group, University College London, London, United Kingdom
| | - Karl Bauer
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
Max Planck Institute for Experimental Endocrinology, Hannover, Germany.
Laboratory of Comparative Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium.
Perinatal Brain Repair Group, University College London, London, United Kingdom
| | - Heike Heuer
- Leibniz Institute for Age Research/Fritz Lipmann Institute, Jena, Germany.
Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
Max Planck Institute for Experimental Endocrinology, Hannover, Germany.
Laboratory of Comparative Endocrinology, Katholieke Universiteit Leuven, Leuven, Belgium.
Perinatal Brain Repair Group, University College London, London, United Kingdom
| |
Collapse
|
48
|
van der Heide SM, Joosten BJLJ, Dragt BS, Everts ME, Klaren PHM. A physiological role for glucuronidated thyroid hormones: preferential uptake by H9c2(2-1) myotubes. Mol Cell Endocrinol 2007; 264:109-17. [PMID: 17118529 DOI: 10.1016/j.mce.2006.10.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2006] [Revised: 10/12/2006] [Accepted: 10/13/2006] [Indexed: 02/07/2023]
Abstract
Conjugation reactions are important pathways in the peripheral metabolism of thyroid hormones. Rat cardiac fibroblasts produce and secrete glucuronidated thyroxine (T4G) and 3,3',5-triiodothyronine (T3G). We here show that, compared to fibroblasts from other anatomical locations, the capacity of cardiofibroblasts to secrete T4G and T3G is highest. H9c2(2-1) myotubes, a model system for cardiomyocytes, take up T4G and T3G at a rate that is 10-15 times higher than that for the unconjugated thyroid hormones. T3 and T4, and their glucuronides, stimulate H9c2(2-1) myoblast-to-myotube differentiation. A substantial beta-glucuronidase activity was measured in H9c2(2-1) myotubes, and this confers a deconjugating capacity to these cells, via which native thyroid hormones can be regenerated from glucuronidated precursors. This indicates that the stimulatory effects on myoblast differentiation are exerted by the native hormones. We suggest that glucuronidation represents a mechanism to uncouple local thyroid hormone action in the heart from that in other peripheral tissues and in the systemic circulation. This could represent a mechanism for the local fine-tuning of cardiac thyroid hormone action.
Collapse
Affiliation(s)
- Sabine M van der Heide
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
49
|
Langouche L, Van den Berghe G. The dynamic neuroendocrine response to critical illness. Endocrinol Metab Clin North Am 2006; 35:777-91, ix. [PMID: 17127146 DOI: 10.1016/j.ecl.2006.09.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The severity of striking alterations in the hypothalamic-anterior pituitary-peripheral hormone axes, which are the hallmark of severity of critical illness, is associated with a high risk for morbidity and mortality. Most attempts to correct the hormone balance are ineffective or harmful because of lack of pathophysiologic understanding. Extensive research has provided more insight in the biphasic neuroendocrine response to critical illness: the acute phase is characterized by an actively secreting pituitary but low peripheral effector hormone levels. In contrast, in prolonged critical illness, uniform suppression of the neuroendocrine axes, predominantly of hypothalamic origin, contributes to low serum levels of the respective target-organ hormones.
Collapse
Affiliation(s)
- Lies Langouche
- Department of Intensive Care Medicine, Katholieke Universiteit Leuven, Herestraat 49, B-300 Leuven, Belgium
| | | |
Collapse
|
50
|
Hamann I, Seidlova-Wuttke D, Wuttke W, Köhrle J. Effects of isoflavonoids and other plant-derived compounds on the hypothalamus–pituitary–thyroid hormone axis. Maturitas 2006. [DOI: 10.1016/j.maturitas.2006.06.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|