1
|
Galstyan DS, Krotova NA, Lebedev AS, Kotova MM, Martynov DD, Golushko NI, Perederiy AS, Zhukov IS, Rosemberg DB, Lim LW, Yang L, de Abreu MS, Gainetdinov RR, Kalueff AV. Trace amine signaling in zebrafish models: CNS pharmacology, behavioral regulation and translational relevance. Eur J Pharmacol 2025; 991:177312. [PMID: 39870233 DOI: 10.1016/j.ejphar.2025.177312] [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: 09/04/2024] [Revised: 11/29/2024] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
Tyramine, β-phenylethylamine, octopamine and other trace amines are endogenous substances recently recognized as important novel neurotransmitters in the brain. Trace amines act via multiple selective trace amine-associated receptors (TAARs) of the G protein-coupled receptor family. TAARs are expressed in various brain regions and modulate neurotransmission, neuronal excitability, adult neurogenesis, cognition, mood, locomotor activity and olfaction. Disrupted trace amine circuits have been implicated in various clinical neuropsychiatric disorders, including schizophrenia, Parkinson's disease, addiction, depression and anxiety. Dysregulated TAAR signaling has been linked in rodents to altered dopamine and serotonin neurotransmission, known to be associated with these psychiatric conditions. Complementing rodent genetic and pharmacological evidence, zebrafish (Danio rerio) are rapidly becoming a novel powerful model system in translational neuropharmacology research. Here, we review trace amine/TAAR neurobiology in zebrafish and discuss their developing translational utility as pharmacological and genetic models for unraveling the role of trace amines in CNS processes and brain disorders.
Collapse
Affiliation(s)
- David S Galstyan
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Natalia A Krotova
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Andrey S Lebedev
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Maria M Kotova
- Neuroscience Program, Sirius University of Science and Technology, Sochi, Russia
| | - Daniil D Martynov
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Nikita I Golushko
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Alexander S Perederiy
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Ilya S Zhukov
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), New Olreans, USA
| | - Lee Wei Lim
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - LongEn Yang
- Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Murilo S de Abreu
- Western Caspian University, Baku, Azerbaijan; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), New Olreans, USA; Moscow Institute of Physics and Technology, Moscow, Russia.
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia
| | - Allan V Kalueff
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neuroscience Program, Sirius University of Science and Technology, Sochi, Russia; Department of Biosciences and Bioinformatics, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China; Moscow Institute of Physics and Technology, Moscow, Russia.
| |
Collapse
|
2
|
Fairooz T, McNamee SE, Finlay D, Ng KY, McLaughlin J. Enhancing Sensitivity of Point-of-Care Thyroid Diagnosis via Computational Analysis of Lateral Flow Assay Images Using Novel Textural Features and Hybrid-AI Models. BIOSENSORS 2024; 14:611. [PMID: 39727875 DOI: 10.3390/bios14120611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Lateral flow assays are widely used in point-of-care diagnostics but face challenges in sensitivity and accuracy when detecting low analyte concentrations, such as thyroid-stimulating hormone biomarkers. This study aims to enhance assay performance by leveraging textural features and hybrid artificial intelligence models. A modified Gray-Level Co-occurrence Matrix, termed the Averaged Horizontal Multiple Offsets Gray-Level Co-occurrence Matrix, was utilised to compute the textural features of the biosensor assay images. Significant textural features were selected for further analysis. A deep learning Convolutional Neural Network model was employed to extract features from these textural features. Both traditional machine learning models and hybrid artificial intelligence models, which combine Convolutional Neural Network features with traditional algorithms, were used to categorise these textural features based on the thyroid-stimulating hormone concentration levels. The proposed method achieved accuracy levels exceeding 95%. This pioneering study highlights the utility of textural aspects of assay images for accurate predictive disease modelling, offering promising advancements in diagnostics and management within biomedical research.
Collapse
Affiliation(s)
- Towfeeq Fairooz
- School of Engineering, Ulster University, Belfast BT15 1ED, UK
| | - Sara E McNamee
- School of Engineering, Ulster University, Belfast BT15 1ED, UK
| | - Dewar Finlay
- School of Engineering, Ulster University, Belfast BT15 1ED, UK
| | - Kok Yew Ng
- School of Engineering, Ulster University, Belfast BT15 1ED, UK
| | | |
Collapse
|
3
|
Jin Y, Zhai T, Wang Y, Li J, Wang T, Huang J. Recent advances in liquid chromatography-tandem mass spectrometry for the detection of thyroid hormones and thyroglobulin in clinical samples: A review. J Sep Sci 2024; 47:e2400466. [PMID: 39294846 DOI: 10.1002/jssc.202400466] [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: 06/23/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/21/2024]
Abstract
Thyroid hormones (THs), including triiodothyronine (T3), thyroxine (T4), and their metabolites, are essential for regulating development, growth, and energy metabolism. Thyroglobulin (Tg) produced by thyroid follicular cells acts as an essential substrate for TH synthesis. The combination of THs with Tg is a widely used serological laboratory test for thyroid function assessment. Early detection and timely intervention are significant for preventing and managing thyroid disease. In recent years, liquid chromatography-tandem mass spectrometry (LC-MS/MS) has emerged as a powerful tool for the precise detection of small molecular analytes and steroid hormones in clinical practice as a result of its high sensitivity and specificity. While LC-MS/MS has been increasingly used for detecting THs and Tg recently, its application in clinical practice is still in its early stages. Recent advances in the assessment of thyroid metabolism using LC-MS/MS in clinical samples published during 2004-2023 were reviewed, with a special focus on the use of this technique for quantifying molecules involved in thyroid diseases.
Collapse
Affiliation(s)
- Yuting Jin
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Taiyu Zhai
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ying Wang
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiuyan Li
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Tingting Wang
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jing Huang
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Jragh D, Yousif MHM, Oriowo MA. Vasoconstrictor Effects of Trace Amine-Associated Receptor Agonists in the Isolated Perfused Rat Kidney. Med Princ Pract 2024; 33:537-544. [PMID: 39047696 PMCID: PMC11631017 DOI: 10.1159/000540429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
INTRODUCTION Endogenous trace amines such as tryptamine and 3-iodothyronamine (T1AM) are present in mammalian tissues at very low concentrations. They produce their actions by activating surface G protein-coupled receptors known as trace amine-associated receptors (TAARs). OBJECTIVE The study was designed to investigate the possible vasoconstrictor effects of tryptamine, T1AM, and the selective TAAR1 agonist RO5263397 in isolated perfused rat kidney. METHODS Renal vascular reactivity experiment using male Wistar Kyoto (WKY, n = 76) and spontaneously hypertensive rats (SHRs, n = 81) were used in this study. RESULTS Tryptamine (1011-106 mole), T1AM (1011-106 mole), and RO5263397 (1011-106 mole) increased perfusion pressure in preparations from WKY rats and SHRs in a dose-dependent manner. EPPTB, a selective TAAR1 antagonist (10-6 M), significantly (p < 0.05) reduced agonist-induced increase in perfusion pressure in both WKY rats and SHRs, suggesting a role for TAAR1 activation in these responses. The vasoconstrictor responses in both groups were significantly reduced by L-type dihydropyridine calcium channel blocker, Rho-kinase, and protein kinase C (PKC) inhibitors suggesting the involvement of extracellular calcium and enhanced calcium sensitization. Reactive oxygen species (ROS) scavenger TEMPO significantly inhibited the agonist-induced increase in perfusion pressure in preparations from SHRs but not WKY. CONCLUSION The TAARs enhanced agonist-induced increase in perfusion causing vasoconstriction in the kidney involves the influx of extracellular calcium, increased calcium sensitization, and ROS in SHRs animals only.
Collapse
Affiliation(s)
- Dina Jragh
- Department of Pharmacology and Toxicology, College of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Mariam H M Yousif
- Department of Pharmacology and Toxicology, College of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Mabayoje A Oriowo
- Department of Pharmacology and Toxicology, College of Medicine, Kuwait University, Kuwait City, Kuwait
| |
Collapse
|
5
|
Han Y, Han Z, Huang X, Li S, Jin G, Feng J, Wu D, Liu H. An injectable refrigerated hydrogel for inducing local hypothermia and neuroprotection against traumatic brain injury in mice. J Nanobiotechnology 2024; 22:251. [PMID: 38750597 PMCID: PMC11095020 DOI: 10.1186/s12951-024-02454-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/01/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Hypothermia is a promising therapy for traumatic brain injury (TBI) in the clinic. However, the neuroprotective outcomes of hypothermia-treated TBI patients in clinical studies are inconsistent due to several severe side effects. Here, an injectable refrigerated hydrogel was designed to deliver 3-iodothyronamine (T1AM) to achieve a longer period of local hypothermia for TBI treatment. Hydrogel has four advantages: (1) It can be injected into injured sites after TBI, where it forms a hydrogel and avoids the side effects of whole-body cooling. (2) Hydrogels can biodegrade and be used for controlled drug release. (3) Released T1AM can induce hypothermia. (4) This hydrogel has increased medical value given its simple operation and ability to achieve timely treatment. METHODS Pol/T hydrogels were prepared by a low-temperature mixing method and characterized. The effect of the Pol/T hydrogel on traumatic brain injury in mice was studied. The degradation of the hydrogel at the body level was observed with a small animal imager. Brain temperature and body temperature were measured by brain thermometer and body thermometer, respectively. The apoptosis of peripheral nerve cells was detected by immunohistochemical staining. The protective effect of the hydrogels on the blood-brain barrier (BBB) after TBI was evaluated by the Evans blue penetration test. The protective effect of hydrogel on brain edema after injury in mice was detected by Magnetic resonance (MR) in small animals. The enzyme linked immunosorbent assay (ELISA) method was used to measure the levels of inflammatory factors. The effects of behavioral tests on the learning ability and exercise ability of mice after injury were evaluated. RESULTS This hydrogel was able to cool the brain to hypothermia for 12 h while maintaining body temperature within the normal range after TBI in mice. More importantly, hypothermia induced by this hydrogel leads to the maintenance of BBB integrity, the prevention of cell death, the reduction of the inflammatory response and brain edema, and the promotion of functional recovery after TBI in mice. This cooling method could be developed as a new approach for hypothermia treatment in TBI patients. CONCLUSION Our study showed that injectable and biodegradable frozen Pol/T hydrogels to induce local hypothermia in TBI mice can be used for the treatment of traumatic brain injury.
Collapse
Affiliation(s)
- Yuhan Han
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
- Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Head Trauma, Shanghai, 200127, China
| | - Zhengzhong Han
- Department of Neurosurgery, Xuzhou Children's Hospital, Xuzhou, 221000, Jiangsu, China
| | - Xuyang Huang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
- Department of Intensive Care Medicine, The Second Hospital of Jiaxing, Jiaxing, 314000, Zhejiang, China
| | - Shanshan Li
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Guoliang Jin
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Junfeng Feng
- Brain Injury Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Head Trauma, Shanghai, 200127, China.
| | - Decheng Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| | - Hongmei Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
6
|
Wang T, Li C, Ma Y, Zhou H, Du X, Li Y, Long S, Ding Y, Lu G, Chen W, Zhou Y, Yu L, Wang J, Wang Y. Metabolomics of cerebrospinal fluid reveals prognostic biomarkers in pediatric status epilepticus. CNS Neurosci Ther 2023; 29:3925-3934. [PMID: 37381696 PMCID: PMC10651953 DOI: 10.1111/cns.14312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/30/2023] Open
Abstract
AIMS Status epilepticus (SE) is the most common neurological emergency in pediatric patients. This study aimed to screen for prognostic biomarkers of SE in the cerebrospinal fluid (CSF) using metabolomics. METHODS Ultra-performance liquid chromatography quadrupole time-of-flight tandem mass spectrometry (UPLC-QTOF-MS) was conducted to identify prognostic biomarkers in CSF metabolomics by comparing the poor outcome group (N = 13) with the good outcome group (N = 15) of children with SE. Differentially expressed metabolites were identified using Mann-Whitney U test corrected by Benjamini-Hochberg and partial least squares discriminant analysis (PLS-DA). RESULTS The PLS-DA model identified and validated significant metabolic differences between the poor and good outcome groups of children with SE (PLS-DA with R2 Y = 0.992 and Q2 = 0.798). A total of 49 prognosis-related metabolites were identified. Of these metabolites, 20 including glutamyl-glutamine, 3-iodothyronamine, and L-fucose had an area under the curve (AUC) ≥ 80% in prognostic prediction of SE. The logistic regression model combining glutamyl-glutamine and 3-iodothyronamine produced an AUC value of 0.976, with a sensitivity of 0.863 and specificity of 0.956. Pathway analysis revealed that dysregulation of the citrate cycle (TCA) and arginine biosynthesis may contribute to poor SE prognosis. CONCLUSIONS This study highlighted the prognosis-related metabolomic disturbances in the CSF of children with SE and identified potential prognostic biomarkers. A prognostic prediction model combining glutamyl-glutamine and 3-iodothyronamine with high predictive value was established.
Collapse
Affiliation(s)
- Tianqi Wang
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Chunpei Li
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yu Ma
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Hao Zhou
- Department of Developmental Behavioral Pediatrics, Guizhou Provincial People's HospitalMedical College of Guizhou UniversityGuiyangChina
| | - Xiaonan Du
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yingfeng Li
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Shasha Long
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yifeng Ding
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Guoping Lu
- Pediatric Intensive Care Unit, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Weiming Chen
- Pediatric Intensive Care Unit, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yuanfeng Zhou
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Lifei Yu
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Ji Wang
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yi Wang
- Department of Neurology, National Children's Medical CenterChildren's Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
7
|
Xu Z, Guo L, Yu J, Shen S, Wu C, Zhang W, Zhao C, Deng Y, Tian X, Feng Y, Hou H, Su L, Wang H, Guo S, Wang H, Wang K, Chen P, Zhao J, Zhang X, Yong X, Cheng L, Liu L, Yang S, Yang F, Wang X, Yu X, Xu Y, Sun JP, Yan W, Shao Z. Ligand recognition and G-protein coupling of trace amine receptor TAAR1. Nature 2023; 624:672-681. [PMID: 37935376 DOI: 10.1038/s41586-023-06804-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/31/2023] [Indexed: 11/09/2023]
Abstract
Trace-amine-associated receptors (TAARs), a group of biogenic amine receptors, have essential roles in neurological and metabolic homeostasis1. They recognize diverse endogenous trace amines and subsequently activate a range of G-protein-subtype signalling pathways2,3. Notably, TAAR1 has emerged as a promising therapeutic target for treating psychiatric disorders4,5. However, the molecular mechanisms underlying its ability to recognize different ligands remain largely unclear. Here we present nine cryo-electron microscopy structures, with eight showing human and mouse TAAR1 in a complex with an array of ligands, including the endogenous 3-iodothyronamine, two antipsychotic agents, the psychoactive drug amphetamine and two identified catecholamine agonists, and one showing 5-HT1AR in a complex with an antipsychotic agent. These structures reveal a rigid consensus binding motif in TAAR1 that binds to endogenous trace amine stimuli and two extended binding pockets that accommodate diverse chemotypes. Combined with mutational analysis, functional assays and molecular dynamic simulations, we elucidate the structural basis of drug polypharmacology and identify the species-specific differences between human and mouse TAAR1. Our study provides insights into the mechanism of ligand recognition and G-protein selectivity by TAAR1, which may help in the discovery of ligands or therapeutic strategies for neurological and metabolic disorders.
Collapse
Affiliation(s)
- Zheng Xu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Lulu Guo
- Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingjing Yu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Shen
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Wu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weifeng Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Chang Zhao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Deng
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowen Tian
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuying Feng
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hanlin Hou
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lantian Su
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Shuo Guo
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Heli Wang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kexin Wang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Peipei Chen
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Xiaoyu Zhang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xihao Yong
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Cheng
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shengyong Yang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Yang
- Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
- Beijing National Laboratory for Molecular Sciences, Beijing, China
| | - Xiao Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China.
| | - Jin-Peng Sun
- Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| | - Wei Yan
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|
8
|
Sato K, Yamauchi K, Ishihara A. Analysis of evolutionary and functional features of the bullfrog SULT1 family. Gen Comp Endocrinol 2023; 342:114349. [PMID: 37495023 DOI: 10.1016/j.ygcen.2023.114349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/17/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
We identified the bullfrog Rana catesbeiana sulfotransferase 1 (SULT1) family from the BLAST search tool of the public databases based on the SULT1 families of Nanorana parkeri, Xenopus laevis, and Xenopus tropicalis as queries, revealing the characteristics of the anuran SULT1 family. The results showed that the anuran SULT1 family comprises six subfamilies, four of which were related to the mammalian SULT1 subfamily. Additionally, the bullfrog has two SULT1Cc subfamily members that are consistent with the characteristics of the expanded Xenopus SULT1C subfamily. Several members of the bullfrog SULT1 family were suggested to play important roles in sulfation during metamorphosis. Among these, cDNAs encoding SULT1Cc1 and SULT1Y1 were cloned, and the sulfation activity was analyzed using recombinant proteins. The affinity for 2-naphthol and 3'-phosphoadenosine 5'-phosphosulfate (PAPS) and the enzymatic reaction rate were higher in SULT1Cc1 than in SULT1Y1. Both the enzymes showed inhibitory effect of many thyroid hormones (THs) analogs on the sulfation of 2-naphthol. The potency of sulfation activities of SULT1Cc1 and SULT1Y1 against T4 indicated their possible role in the intracellular T4 clearance during metamorphosis.
Collapse
Affiliation(s)
- Kosuke Sato
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka 422-8529, Japan.
| | - Kiyoshi Yamauchi
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka 422-8529, Japan.
| | - Akinori Ishihara
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka 422-8529, Japan.
| |
Collapse
|
9
|
Chakrabarti N, Sarkar PK, Ray AK, Martin JV. Unveiling the nongenomic actions of thyroid hormones in adult mammalian brain: The legacy of Mary B. Dratman. Front Endocrinol (Lausanne) 2023; 14:1240265. [PMID: 37842308 PMCID: PMC10570802 DOI: 10.3389/fendo.2023.1240265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
A comprehensive review was conducted to compile the contributions of Mary B. Dratman and studies by other researchers in the field of nongenomic actions of thyroid hormones in adult mammalian brain. Dratman and her collaborators authored roughly half of the papers in this area. It has been almost fifty years since Dratman introduced the novel concept of thyroid hormones as neurotransmitters for the first time. The characterization of unique brain-region specific accumulation of thyroid hormones within the nerve terminals in adult mammals was a remarkable contribution by Dratman. It suggested a neurotransmitter- or neuromodulator-like role of thyroid hormone and/or its derivative, 3-iodothyronamine within adrenergic systems in adult mammalian brain. Several studies by other researchers using synaptosomes as a model system, have contributed to the concept of direct nongenomic actions of thyroid hormones at synaptic regions by establishing that thyroid hormones or their derivatives can bind to synaptosomal membranes, alter membrane functions including enzymatic activities and ion transport, elicit Ca2+/NO-dependent signaling pathways and induce substrate-protein phosphorylation. Such findings can help to explain the physiological and pathophysiological roles of thyroid hormone in psychobehavioral control in adult mammalian brain. However, the exact mode of nongenomic actions of thyroid hormones at nerve terminals in adult mammalian brain awaits further study.
Collapse
Affiliation(s)
- Nilkanta Chakrabarti
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
- (CPEPA-UGC) Centre for Electro-Physiological and Neuroimaging studies including Mathematical Modelling, University of Calcutta, Kolkata, West Bengal, India
| | - Pradip K. Sarkar
- Department of Basic Sciences, Parker University, Dallas, TX, United States
| | - Arun K. Ray
- Department of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VII-M, Kolkata, India
| | - Joseph V. Martin
- Biology Department, Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, United States
| |
Collapse
|
10
|
Kim B, Ko YH, Si J, Na J, Ortore G, Chiellini G, Kim JH. Thyroxine metabolite-derived 3-iodothyronamine (T1AM) and synthetic analogs as efficient suppressors of transthyretin amyloidosis. Comput Struct Biotechnol J 2023; 21:4717-4728. [PMID: 37822560 PMCID: PMC10562617 DOI: 10.1016/j.csbj.2023.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/12/2023] [Accepted: 09/23/2023] [Indexed: 10/13/2023] Open
Abstract
Aggregation and fibrillization of transthyretin (TTR) is a fatal pathogenic process that can cause cardiomyopathic and polyneuropathic diseases in humans. Although several therapeutic strategies have been designed to prevent and treat related pathological events, there is still an urgent need to develop better strategies to improve potency and wider applicability. Here, we present our study demonstrating that 3-iodothyronamine (T1AM) and selected thyronamine-like compounds can effectively prevent TTR aggregation. T1AM is one of the thyroid hormone (TH) metabolites, and T1AM and its analogs, such as SG2, SG6, and SG12, are notable molecules for their beneficial activities against metabolic disorders and neurodegeneration. Using nuclear magnetic resonance (NMR) spectroscopy and biochemical analysis, we confirmed that T1AM analogs could bind to and suppress acid-induced aggregation of TTR. In addition, we employed computational approaches to further understand the detailed mechanisms of the interaction between T1AM analogs and TTR. This study demonstrates that T1AM analogs, whose beneficial effects against several pathological processes have already been proven, may have additional benefits against TTR aggregation and fibrillization. Moreover, we believe that our work provides invaluable insights to enhance the pleiotropic activity of T1AM and structurally related analogs, relevant for their therapeutic potential, with particular reference to the ability to prevent TTR aggregation.
Collapse
Affiliation(s)
- Bokyung Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Republic of Korea
| | - Young Ho Ko
- Center for Self-Assembly and Complexity, Institute for Basic Science, Pohang 37673, Republic of Korea
| | - Jinbeom Si
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jongbum Na
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Republic of Korea
| | | | | | - Jin Hae Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Republic of Korea
| |
Collapse
|
11
|
Martin JV, Sarkar PK. Nongenomic roles of thyroid hormones and their derivatives in adult brain: are these compounds putative neurotransmitters? Front Endocrinol (Lausanne) 2023; 14:1210540. [PMID: 37701902 PMCID: PMC10494427 DOI: 10.3389/fendo.2023.1210540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/02/2023] [Indexed: 09/14/2023] Open
Abstract
We review the evidence regarding the nongenomic (or non-canonical) actions of thyroid hormones (thyronines) and their derivatives (including thyronamines and thyroacetic acids) in the adult brain. The paper seeks to evaluate these compounds for consideration as candidate neurotransmitters. Neurotransmitters are defined by their (a) presence in the neural tissue, (b) release from neural tissue or cell, (c) binding to high-affinity and saturable recognition sites, (d) triggering of a specific effector mechanism and (e) inactivation mechanism. Thyronines and thyronamines are concentrated in brain tissue and show distinctive patterns of distribution within the brain. Nerve terminals accumulate a large amount of thyroid hormones in mature brain, suggesting a synaptic function. However, surprisingly little is known about the potential release of thyroid hormones at synapses. There are specific binding sites for thyroid hormones in nerve-terminal fractions (synaptosomes). A notable cell-membrane binding site for thyroid hormones is integrin αvβ3. Furthermore, thyronines bind specifically to other defined neurotransmitter receptors, including GABAergic, catecholaminergic, glutamatergic, serotonergic and cholinergic systems. Here, the thyronines tend to bind to sites other than the primary sites and have allosteric effects. Thyronamines also bind to specific membrane receptors, including the trace amine associated receptors (TAARs), especially TAAR1. The thyronines and thyronamines activate specific effector mechanisms that are short in latency and often occur in subcellular fractions lacking nuclei, suggesting nongenomic actions. Some of the effector mechanisms for thyronines include effects on protein phosphorylation, Na+/K+ ATPase, and behavioral measures such as sleep regulation and measures of memory retention. Thyronamines promptly regulate body temperature. Lastly, there are numerous inactivation mechanisms for the hormones, including decarboxylation, deiodination, oxidative deamination, glucuronidation, sulfation and acetylation. Therefore, at the current state of the research field, thyroid hormones and their derivatives satisfy most, but not all, of the criteria for definition as neurotransmitters.
Collapse
Affiliation(s)
- Joseph V. Martin
- Biology Department, Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, United States
| | - Pradip K. Sarkar
- Department of Basic Sciences, Parker University, Dallas, TX, United States
| |
Collapse
|
12
|
Polini B, Ricardi C, Bertolini A, Carnicelli V, Rutigliano G, Saponaro F, Zucchi R, Chiellini G. T1AM/TAAR1 System Reduces Inflammatory Response and β-Amyloid Toxicity in Human Microglial HMC3 Cell Line. Int J Mol Sci 2023; 24:11569. [PMID: 37511328 PMCID: PMC10380917 DOI: 10.3390/ijms241411569] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Microglial dysfunction is one of the hallmarks and leading causes of common neurodegenerative diseases (NDDs), including Alzheimer's disease (AD) and Parkinson's disease (PD). All these pathologies are characterized by aberrant aggregation of disease-causing proteins in the brain, which can directly activate microglia, trigger microglia-mediated neuroinflammation, and increase oxidative stress. Inhibition of glial activation may represent a therapeutic target to alleviate neurodegeneration. Recently, 3-iodothyronamine (T1AM), an endogenous derivative of thyroid hormone (TH) able to interact directly with a specific GPCR known as trace amine-associated receptor 1 (TAAR1), gained interest for its ability to promote neuroprotection in several models. Nevertheless, T1AM's effects on microglial disfunction remain still elusive. In the present work we investigated whether T1AM could inhibit the inflammatory response of human HMC3 microglial cells to LPS/TNFα or β-amyloid peptide 25-35 (Aβ25-35) stimuli. The results of ELISA and qPCR assays revealed that T1AM was able to reduce microglia-mediated inflammatory response by inhibiting the release of proinflammatory factors, including IL-6, TNFα, NF-kB, MCP1, and MIP1, while promoting the release of anti-inflammatory mediators, such as IL-10. Notably, T1AM anti-inflammatory action in HMC3 cells turned out to be a TAAR1-mediated response, further increasing the relevance of the T1AM/TAAR1 system in the management of NDDs.
Collapse
Affiliation(s)
- Beatrice Polini
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Caterina Ricardi
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Andrea Bertolini
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Vittoria Carnicelli
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Grazia Rutigliano
- Institute of Clinical Sciences, Imperial College London, London SW7 2AZ, UK;
| | - Federica Saponaro
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Riccardo Zucchi
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| | - Grazia Chiellini
- Department of Pathology, University of Pisa, 56100 Pisa, Italy; (C.R.); (A.B.); (V.C.); (F.S.); (R.Z.)
| |
Collapse
|
13
|
Elsammak GA, Talaat A, Reda S. The possible ameliorative role of Lycopene on Tributyltin induced thyroid damage in adult male albino rats (histological, immunohistochemical and biochemical study). Ultrastruct Pathol 2023; 47:324-338. [PMID: 37125846 DOI: 10.1080/01913123.2023.2205922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/19/2023] [Indexed: 05/02/2023]
Abstract
Tributyltin is used in industrial applications. This current research aimed to study the effect of Tributyltin on the thyroid gland structure and function of adult male albino rats and the protective effect of Lycopene. Twenty-one male adult albino rats were classified into three groups: Control, treated that received Tributyltin, and protective that received Lycopene with Tributyltin. At the end of the experiment, blood samples were collected and T4, T3, and (TSH) were measured. Tissue superoxide dismutase (SOD) and malondialdehyde (MDA) were estimated. Thyroid gland specimens were processed for histological and immunohistochemical examination. Then morphometric and statistical analyses were done. The treated group showed affection in thyroid function and histological structure as vacuolated colloid and cytoplasm and dark nuclei. Ultrastructurally, follicular cells showed irregular shrunken nuclei, atrophied apical microvilli, vacuoles, multiple lysosomal granules, mitochondria with destructed cristae, and extensively dilated rough endoplasmic reticulum. There was increase in Caspase-3 immunoexpression and decrease in Beclin-1 immunoexpression. The thyroid structure and biochemical markers improved after Lycopene administration. The thyroid gland damage caused by Tributyltin is ameliorated by Lycopene.
Collapse
Affiliation(s)
- Ghada A Elsammak
- Medical Histology and cell biology Department, Zagazig University Faculty of Human Medicine, Zagazig, Egypt
| | - Aliaa Talaat
- Medical Biochemistry Department, Zagazig University Faculty of Human Medicine, Zagazig, Egypt
| | - Samar Reda
- Medical Histology and cell biology Department, Zagazig University Faculty of Human Medicine, Zagazig, Egypt
| |
Collapse
|
14
|
Hegazy W, Abdul-Hamid M, Abdel-Rehiem ES, Abdel-Moneim A, Salah M. The protective impact of hesperidin against carbimazole-induced hypothyroidism, via enhancement of inflammatory cytokines, histopathological alterations, and Nrf2/HO-1. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:53589-53604. [PMID: 36862292 DOI: 10.1007/s11356-023-26103-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
The aim of this study was to evaluate the anti-inflammatory, antioxidant, and antiproliferative effects of hesperidin (HSP) and eltroxin (ELT) on hypothyroidism (HPO) induced by carbimazole (CBZ) in white male albino rats. Thirty-two adult rats were categorized into four groups: Group 1, no treatment (control); Group II, treated with CBZ (20 mg/kg); Group III, treated with HSP (200 mg/kg) + CBZ; and Group IV, treated with ELT (0.045 mg/kg) + CBZ. All treatments were provided as oral daily doses for 90 days. Thyroid hypofunction was significantly manifested in Group II. However, increased levels of thyroid hormones, antioxidant enzymes, nuclear factor erythroid 2-related factor 2, heme oxygenase 1, and interleukin (IL)-10, and a decrease in the level of the thyroid-stimulating hormone were observed in Groups III and IV. On the contrary, decreased levels of lipid peroxidation, inducible nitric oxide synthase, tumor necrosis factor α, IL-17, and cyclooxygenase 2 were detected in groups III and IV. The histopathological and ultrastructural findings were ameliorated in Groups III and IV; on the contrary, Group II presented with significant increases in the height and number of layers of the follicular cells. Immunohistochemistry demonstrated a marked increase in thyroglobulin and significant decreases in the levels of nuclear factor kappa B and proliferating cell nuclear antigen in Groups III and IV. These results confirmed the effectiveness of HSP as an anti-inflammatory, antioxidant, and antiproliferative agent in rats with hypothyroidism. Additional studies are required to assess its potential as a novel agent against HPO.
Collapse
Affiliation(s)
- Walaa Hegazy
- Histology Division, Basic Science Department, Faculty of physical therapy, Nahda University, Beni-Suef, Egypt
| | - Manal Abdul-Hamid
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, Salah Salem St, P.O. Box 62521, Beni-Suef, Egypt.
| | - Eman S Abdel-Rehiem
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Salah Salem St, Box, Beni-Suef, 62521, Egypt
| | - Adel Abdel-Moneim
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Salah Salem St, Box, Beni-Suef, 62521, Egypt
| | - Marwa Salah
- Cell Biology, Histology and Genetics Division, Department of Zoology, Faculty of Science, Beni-Suef University, Salah Salem St, P.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
15
|
Abstract
In this article, starting with the recognition that iodine is essential for normal thyroid function and is a component of thyroid hormone (TH) molecules, we discuss the many seminal observations and discoveries that have led to identification of various pathways of TH metabolism and their potential roles in TH economy and action. We then recount evidence that TH metabolism participates in maintaining the appropriate content of active hormone in a TH-responsive tissue or cell. Thus, metabolism of the TH is not merely a means by which it is degraded and eliminated from the body, but an essential component of an intricate system by which the thyroid exerts its multiple regulatory effects on almost all organs and tissues. The article ends with a summary of the current concepts and some outstanding questions that are awaiting answers.
Collapse
Affiliation(s)
- Valerie Anne Galton
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Arturo Hernandez
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| |
Collapse
|
16
|
Gribble GW. Naturally Occurring Organohalogen Compounds-A Comprehensive Review. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2023; 121:1-546. [PMID: 37488466 DOI: 10.1007/978-3-031-26629-4_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
The present volume is the third in a trilogy that documents naturally occurring organohalogen compounds, bringing the total number-from fewer than 25 in 1968-to approximately 8000 compounds to date. Nearly all of these natural products contain chlorine or bromine, with a few containing iodine and, fewer still, fluorine. Produced by ubiquitous marine (algae, sponges, corals, bryozoa, nudibranchs, fungi, bacteria) and terrestrial organisms (plants, fungi, bacteria, insects, higher animals) and universal abiotic processes (volcanos, forest fires, geothermal events), organohalogens pervade the global ecosystem. Newly identified extraterrestrial sources are also documented. In addition to chemical structures, biological activity, biohalogenation, biodegradation, natural function, and future outlook are presented.
Collapse
Affiliation(s)
- Gordon W Gribble
- Department of Chemistry, Dartmouth College, Hanover, NH, 03755, USA.
| |
Collapse
|
17
|
Sakanoue W, Yokoyama T, Hirakawa M, Maesawa S, Sato K, Saino T. 3-Iodothyronamine, a trace amine-associated receptor agonist, regulates intracellular Ca2+ increases via CaMK II through Epac2 in rat cerebral arterioles. Biomed Res 2023; 44:219-232. [PMID: 37779034 DOI: 10.2220/biomedres.44.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Trace amines (TAs) in the nervous system bind to TA-associated receptors (TAARs) and are involved in the regulation of monoaminergic functions. Among TAAR subtypes, TAAR1 has been implicated in the development of neurological disorders, such as schizophrenia. The present study investigated the effects of the TAAR1 agonist, 3-iodothyronamine (T1AM) on cerebral arterioles using fluctuations in the intracellular concentration of Ca2+ ([Ca2+]i) as an index of contractile responses. In cerebral arterioles, most of the TAAR agonists did not increase [Ca2+]i, while only T1AM elevated [Ca2+]i in vascular smooth muscle cells. This increase involved extracellular Ca2+ influx through T-type Ca2+ channels and inositol trisphosphate- and ryanodine-receptor-mediated Ca2+ release from intracellular stores. The inhibition of the cAMP sensor, exchange protein directly activated by cAMP (Epac) 2, and calmodulin kinase (CaMK) II strongly inhibited Ca2+ elevations. The present study revealed that T1AM acted not only on the TAAR1 receptor as previously suggested, but also on other G-protein coupled receptors and/or signal transduction systems to increase intracellular Ca2+ in cerebral arteriole smooth muscle cells. These results suggest that when using T1AM in clinical practice, attention should be paid to the early rise in blood pressure.
Collapse
Affiliation(s)
- Wakana Sakanoue
- Division of Dental Anesthesiology, Department of Reconstructive Oral and Maxillofacial Surgery, School of Dentistry, Iwate Medical University, Yahaba, Japan
| | - Takuya Yokoyama
- Laboratory of Veterinary Anatomy and Cell Biology, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Masato Hirakawa
- Department of Anatomy (Cell Biology), Iwate Medical University, Yahaba, Japan
| | - Satsuki Maesawa
- Division of Dental Anesthesiology, Department of Reconstructive Oral and Maxillofacial Surgery, School of Dentistry, Iwate Medical University, Yahaba, Japan
| | - Kenichi Sato
- Division of Dental Anesthesiology, Department of Reconstructive Oral and Maxillofacial Surgery, School of Dentistry, Iwate Medical University, Yahaba, Japan
| | - Tomoyuki Saino
- Department of Anatomy (Cell Biology), Iwate Medical University, Yahaba, Japan
| |
Collapse
|
18
|
Huang S, Liu L, Tang X, Xie S, Li X, Kang X, Zhu S. Research progress on the role of hormones in ischemic stroke. Front Immunol 2022; 13:1062977. [PMID: 36569944 PMCID: PMC9769407 DOI: 10.3389/fimmu.2022.1062977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is a major cause of death and disability around the world. However, ischemic stroke treatment is currently limited, with a narrow therapeutic window and unsatisfactory post-treatment outcomes. Therefore, it is critical to investigate the pathophysiological mechanisms following ischemic stroke brain injury. Changes in the immunometabolism and endocrine system after ischemic stroke are important in understanding the pathophysiological mechanisms of cerebral ischemic injury. Hormones are biologically active substances produced by endocrine glands or endocrine cells that play an important role in the organism's growth, development, metabolism, reproduction, and aging. Hormone research in ischemic stroke has made very promising progress. Hormone levels fluctuate during an ischemic stroke. Hormones regulate neuronal plasticity, promote neurotrophic factor formation, reduce cell death, apoptosis, inflammation, excitotoxicity, oxidative and nitrative stress, and brain edema in ischemic stroke. In recent years, many studies have been done on the role of thyroid hormone, growth hormone, testosterone, prolactin, oxytocin, glucocorticoid, parathyroid hormone, and dopamine in ischemic stroke, but comprehensive reviews are scarce. This review focuses on the role of hormones in the pathophysiology of ischemic stroke and discusses the mechanisms involved, intending to provide a reference value for ischemic stroke treatment and prevention.
Collapse
Affiliation(s)
- Shuyuan Huang
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lu Liu
- Department of Anesthesiology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiaodong Tang
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shulan Xie
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinrui Li
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xianhui Kang
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,*Correspondence: Xianhui Kang, ; Shengmei Zhu,
| | - Shengmei Zhu
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,*Correspondence: Xianhui Kang, ; Shengmei Zhu,
| |
Collapse
|
19
|
De Angelis M, Maity-Kumar G, Schriever SC, Kozlova EV, Müller TD, Pfluger PT, Curras-Collazo MC, Schramm KW. Development and validation of an LC-MS/MS methodology for the quantification of thyroid hormones in dko MCT8/OATP1C1 mouse brain. J Pharm Biomed Anal 2022; 221:115038. [PMID: 36152487 PMCID: PMC7613747 DOI: 10.1016/j.jpba.2022.115038] [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: 05/27/2022] [Revised: 08/19/2022] [Accepted: 09/07/2022] [Indexed: 11/18/2022]
Abstract
The Allan-Herndon Dudley Syndrome (AHDS) is a rare disease caused by the progressive loss of monocarboxylate transporter 8 (MCT8). In patients with AHDS, the absence of MCT8 impairs transport of thyroid hormones (TH) through the blood brain barrier, leading to a central state of TH deficiency. In mice, the AHDS is mimicked by simultaneous deletion of the TH transporters MCT8 and the solute carrier organic anion transporter family member 1c1 (OATP1C1). To support preclinical mouse studies, an analytical methodology was developed and successfully applied for quantifying selected thyroid hormones in mouse whole brain and in specific regions using liquid chromatography tandem mass-spectrometry (LC-MS/MS). An important requirement for the methodology was its high sensitivity since a very low concentration of THs was expected in MCT8/OATP1C1 double-knockout (dko) mouse brain. Seven THs were targeted: L-thyroxine (T4), 3,3,5-triiodo-L-thyronine-thy-ronine (T3), 3,3’,5’-triiodo-L-thyronine-thyronine (rT3), 3,3-diiodo-L-thyronine (3,3’-T2, T2), 3,5-diiodo-L-thyro-nine (rT2, 3,5-T2), 3-iodo-L-thyronine (T1), 3-iodothyronamine (T1AM). Isotope dilution liquid chromatography triple-quadrupole mass spectrometry methodology was applied for detection and quantification. The method was validated in wild-type animals for mouse whole brain and for five different brain regions (hypothalamus, hippocampus, prefrontal cortex, brainstem and cortex). Instrumental calibration curves ranged from 0.35 to 150 pg/μL with good linearity (r2 >0.996). The limit of quantification was from 0.08 to 0.6 pg/mg, with an intra- and inter-day precision of 4.2−14.02% and 0.4−17.9% respectively, and accuracies between 84.9% and 114.8% when the methodology was validated for the whole brain. In smaller, distinct brain regions, intra- and inter-day precision were 0.6−20.7% and 2.5−15.6% respectively, and accuracies were 80.2−128.6%. The new methodology was highly sensitive and allowed for the following quantification in wild-type mice: (i) for the first time, four distinct thyroid hormones (T4, T3, rT3 and 3,3’-T2) in only approximately 100 mg of mouse brain were detected; (ii) the quantification of T4 and T3 for the first time in distinct mouse brain regions were reported. Further, application of our method to MCT8/OATP1C1 dko mice revealed the expected, relative lack of T3 and T4 uptake into the brain, and confirmed the utility of our analytical method to study TH transport across the blood brain barrier in a preclinical model of central TH deficiency.
Collapse
Affiliation(s)
- Meri De Angelis
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Molecular EXposomics, Ingolstädter Landstr. 1, Neuherberg, Germany.
| | - Gandhari Maity-Kumar
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany
| | - Sonja C Schriever
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
| | - Elena V Kozlova
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, USA
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Paul T Pfluger
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, Neuherberg, Germany; TUM School of Medicine, Neurobiology of Diabetes, Technical University Munich, Germany
| | | | - Karl-Werner Schramm
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Molecular EXposomics, Ingolstädter Landstr. 1, Neuherberg, Germany; Department für Biowissenschaftliche Grundlagen, Technische Universität München, Weihenstephaner Steig 23, Freising, Germany
| |
Collapse
|
20
|
Exogenous 3-Iodothyronamine (T1AM) Can Affect Phosphorylation of Proteins Involved on Signal Transduction Pathways in In Vitro Models of Brain Cell Lines, but These Effects Are Not Strengthened by Its Catabolite, 3-Iodothyroacetic Acid (TA1). Life (Basel) 2022; 12:life12091352. [PMID: 36143389 PMCID: PMC9502970 DOI: 10.3390/life12091352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 12/02/2022] Open
Abstract
T1AM, a derivative of thyroid hormones, and its major catabolite, TA1, produce effects on memory acquisition in rodents. In the present study, we compared the effects of exogenous T1AM and TA1 on protein belonging to signal transduction pathways, assuming that TA1 may strengthen T1AM’s effects in brain tissue. A hybrid line of cancer cells of mouse neuroblastoma and rat glioma (NG 108-15), as well as a human glioblastoma cell line (U-87 MG) were used. We first characterized the in vitro model by analyzing gene expression of proteins involved in the glutamatergic cascade and cellular uptake of T1AM and TA1. Then, cell viability, glucose consumption, and protein expression were assessed. Both cell lines expressed receptors implicated in glutamatergic pathway, namely Nmdar1, Glur2, and EphB2, but only U-87 MG cells expressed TAAR1. At pharmacological concentrations, T1AM was taken up and catabolized to TA1 and resulted in more cytotoxicity compared to TA1. The major effect, highlighted in both cell lines, albeit on different proteins involved in the glutamatergic signaling, was an increase in phosphorylation, exerted by T1AM but not reproduced by TA1. These findings indicate that, in our in vitro models, T1AM can affect proteins involved in the glutamatergic and other signaling pathways, but these effects are not strengthened by TA1.
Collapse
|
21
|
Substantial impact of 3-iodothyronamine (T1AM) on the regulations of fluorescent thermoprobe-measured cellular temperature and natriuretic peptide expression in cardiomyocytes. Sci Rep 2022; 12:12740. [PMID: 35882940 PMCID: PMC9325765 DOI: 10.1038/s41598-022-17086-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/20/2022] [Indexed: 11/11/2022] Open
Abstract
There is growing interest in 3-iodothyronamine (T1AM), an active thyroid hormone metabolite, that induces negative inotropic and chronotropic actions in the heart and exerts systemic hypothermic action. We explored the direct impact of T1AM on cardiomyocytes with a focus on the regulation of the intracellular temperature and natriuretic peptide (NP) expression. A thermoprobe was successfully introduced into neonatal rat cardiomyocytes, and the temperature-dependent changes in the fluorescence intensity ratio were measured using a fluorescence microscope. After one-hour incubation with T1AM, the degree of change in the fluorescence intensity ratio was significantly lower in T1AM-treated cardiomyocytes than in equivalent solvent-treated controls (P < 0.01), indicating the direct hypothermic action of T1AM on cardiomyocytes. Furthermore, T1AM treatment upregulated B-type NP (BNP) gene expression comparable to treatment with endothelin-1 or phenylephrine. Of note, ERK phosphorylation was markedly increased after T1AM treatment, and inhibition of ERK phosphorylation by an MEK inhibitor completely cancelled both T1AM-induced decrease in thermoprobe-measured temperature and the increase in BNP expression. In summary, T1AM decreases fluorescent thermoprobe-measured temperatures (estimated intracellular temperatures) and increases BNP expression in cardiomyocytes by activating the MEK/ERK pathway. The present findings provide new insight into the direct myocardial cellular actions of T1AM in patients with severe heart failure.
Collapse
|
22
|
Liao CJ, Huang PS, Chien HT, Lin TK, Yeh CT, Lin KH. Effects of Thyroid Hormones on Lipid Metabolism Pathologies in Non-Alcoholic Fatty Liver Disease. Biomedicines 2022; 10:biomedicines10061232. [PMID: 35740254 PMCID: PMC9219876 DOI: 10.3390/biomedicines10061232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
The typical modern lifestyle contributes to the development of many metabolic-related disorders, as exemplified by metabolic syndrome. How to prevent, resolve, or avoid subsequent deterioration of metabolic disturbances and the development of more serious diseases has become an important and much-discussed health issue. Thus, the question of the physiological and pathological roles of thyroid hormones (THs) in metabolism has never gone out of fashion. Although THs influence almost all organs, the liver is one of the most important targets as well as the hub of metabolic homeostasis. When this homeostasis is out of balance, diseases may result. In the current review, we summarize the common features and actions of THs, first focusing on their effects on lipid metabolism in the liver. In the second half of the review, we turn to a consideration of non-alcoholic fatty liver disease (NAFLD), a disease characterized by excessive accumulation of fat in the liver that is independent of heavy alcohol consumption. NAFLD is a growing health problem that currently affects ~25% of the world’s population. Unfortunately, there are currently no approved therapies specific for NAFLD, which, if left uncontrolled, may progress to more serious diseases, such as cirrhosis or liver cancer. This absence of effective treatment can also result in the development of non-alcoholic steatohepatitis (NASH), an aggressive form of NAFLD that is the leading cause of liver transplantation in the United States. Because THs play a clear role in hepatic fat metabolism, their potential application in the prevention and treatment of NAFLD has attracted considerable research attention. Studies that have investigated the use of TH-related compounds in the management of NAFLD are also summarized in the latter part of this review. An important take-home point of this review is that a comprehensive understanding of the physiological and pathological roles of THs in liver fat metabolism is possible, despite the complexities of this regulatory axis—an understanding that has clinical value for the specific management of NAFLD.
Collapse
Affiliation(s)
- Chia-Jung Liao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-J.L.); (P.-S.H.)
| | - Po-Shuan Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-J.L.); (P.-S.H.)
| | - Hui-Tzu Chien
- Department of Nutrition and Health Sciences, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan;
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| | - Tzu-Kang Lin
- Neurosurgery, Fu Jen Catholic University Hospital School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan;
| | - Chau-Ting Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan;
| | - Kwang-Huei Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-J.L.); (P.-S.H.)
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan;
- Department of Biochemistry, Chang Gung University, 259 Wen-Hwa 1 Road, Taoyuan 333, Taiwan
- Correspondence: ; Tel./Fax: +886-3-2118263
| |
Collapse
|
23
|
Borsò M, Agretti P, Zucchi R, Saba A. Mass spectrometry in the diagnosis of thyroid disease and in the study of thyroid hormone metabolism. MASS SPECTROMETRY REVIEWS 2022; 41:443-468. [PMID: 33238065 DOI: 10.1002/mas.21673] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 06/11/2023]
Abstract
The importance of thyroid hormones in the regulation of development, growth, and energy metabolism is well known. Over the last decades, mass spectrometry has been extensively used to investigate thyroid hormone metabolism and to discover and characterize new molecules involved in thyroid hormones production, such as thyrotropin-releasing hormone. In the earlier period, the quantification methods, usually based on gas chromatography-mass spectrometry, were complicated and time consuming. They were mainly focused on basic research, and were not suitable for clinical diagnostics on a routine basis. The development of the modern mass spectrometers, mainly coupled to liquid chromatography, enabled simpler sample preparation procedures, and the accurate quantification of thyroid hormones, of their precursors, and of their metabolites in biological fluids, tissues, and cells became feasible. Nowadays, molecules of physiological and pathological interest can be assayed also for diagnostic purposes on a routine basis, and mass spectrometry is slowly entering the clinical laboratory. This review takes stock of the advancements in the field of thyroid metabolism that were carried out with mass spectrometry, with special focus on the use of this technique for the quantification of molecules involved in thyroid diseases.
Collapse
Affiliation(s)
- Marco Borsò
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Patrizia Agretti
- Department of Laboratory Medicine, Laboratory of Chemistry and Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Riccardo Zucchi
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Alessandro Saba
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
- Department of Laboratory Medicine, Laboratory of Clinical Pathology, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
24
|
Bikkarolla SK, McNamee SE, Vance P, McLaughlin J. High-Sensitive Detection and Quantitative Analysis of Thyroid-Stimulating Hormone Using Gold-Nanoshell-Based Lateral Flow Immunoassay Device. BIOSENSORS 2022; 12:182. [PMID: 35323452 PMCID: PMC8946628 DOI: 10.3390/bios12030182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/22/2022] [Accepted: 03/17/2022] [Indexed: 06/14/2023]
Abstract
Au nanoparticles (AuNPs) have been used as signal reporters in colorimetric lateral flow immunoassays (LFAs) for decades. However, it remains a major challenge to significantly improve the detection sensitivity of traditional LFAs due to the low brightness of AuNPs. As an alternative approach, we overcome this problem by utilizing 150 nm gold nanoshells (AuNSs) that were engineered by coating low-density silica nanoparticles with a thin layer of gold. AuNSs are dark green, have 14 times larger surface area, and are approximately 35 times brighter compared to AuNPs. In this study, we used detection of thyroid-stimulating hormone (TSH) in a proof-of-concept assay. The limit of detection (LOD) with AuNS-based LFA was 0.16 µIU/mL, which is 26 times more sensitive than the conventional colorimetric LFA that utilizes AuNP as a label. The dynamic range of the calibration curve was 0.16−9.5 µIU/mL, making it possible to diagnose both hyperthyroidism (<0.5 µIU/mL) and hypothyroidism (>5 µIU/mL) using AuNS-based LFA. Thus, the developed device has a strong potential for early screening and diagnosis of diseases related to the thyroid hormone.
Collapse
Affiliation(s)
- Santosh Kumar Bikkarolla
- School of Engineering, Engineering Research Institute, University of Ulster, Newtownabbey BT37 0QB, UK;
| | - Sara E. McNamee
- School of Engineering, Engineering Research Institute, University of Ulster, Newtownabbey BT37 0QB, UK;
| | - Paul Vance
- Randox Laboratories Ltd., 55 Diamond Road, Crumlin, County Antrim BT29 4QY, UK;
| | - James McLaughlin
- School of Engineering, Engineering Research Institute, University of Ulster, Newtownabbey BT37 0QB, UK;
| |
Collapse
|
25
|
Cioffi F, Giacco A, Goglia F, Silvestri E. Bioenergetic Aspects of Mitochondrial Actions of Thyroid Hormones. Cells 2022; 11:cells11060997. [PMID: 35326451 PMCID: PMC8947633 DOI: 10.3390/cells11060997] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/04/2022] [Accepted: 03/13/2022] [Indexed: 02/07/2023] Open
Abstract
Much is known, but there is also much more to discover, about the actions that thyroid hormones (TH) exert on metabolism. Indeed, despite the fact that thyroid hormones are recognized as one of the most important regulators of metabolic rate, much remains to be clarified on which mechanisms control/regulate these actions. Given their actions on energy metabolism and that mitochondria are the main cellular site where metabolic transformations take place, these organelles have been the subject of extensive investigations. In relatively recent times, new knowledge concerning both thyroid hormones (such as the mechanisms of action, the existence of metabolically active TH derivatives) and the mechanisms of energy transduction such as (among others) dynamics, respiratory chain organization in supercomplexes and cristes organization, have opened new pathways of investigation in the field of the control of energy metabolism and of the mechanisms of action of TH at cellular level. In this review, we highlight the knowledge and approaches about the complex relationship between TH, including some of their derivatives, and the mitochondrial respiratory chain.
Collapse
|
26
|
Redox Properties of 3-Iodothyronamine (T1AM) and 3-Iodothyroacetic Acid (TA1). Int J Mol Sci 2022; 23:ijms23052718. [PMID: 35269859 PMCID: PMC8910694 DOI: 10.3390/ijms23052718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/24/2022] [Accepted: 02/27/2022] [Indexed: 01/07/2023] Open
Abstract
3-iodothyronamine (T1AM) and 3-iodothyroacetic acid (TA1) are thyroid-hormone-related compounds endowed with pharmacological activity through mechanisms that remain elusive. Some evidence suggests that they may have redox features. We assessed the chemical activity of T1AM and TA1 at pro-oxidant conditions. Further, in the cell model consisting of brown adipocytes (BAs) differentiated for 6 days in the absence (M cells) or in the presence of 20 nM T1AM (M + T1AM cells), characterized by pro-oxidant metabolism, or TA1 (M + TA1 cells), we investigated the expression/activity levels of pro- and anti-oxidant proteins, including UCP-1, sirtuin-1 (SIRT1), mitochondrial monoamine (MAO-A and MAO-B), semicarbazide-sensitive amine oxidase (SSAO), and reactive oxygen species (ROS)-dependent lipoperoxidation. T1AM and TA1 showed in-vitro antioxidant and superoxide scavenging properties, while only TA1 acted as a hydroxyl radical scavenger. M + T1AM cells showed higher lipoperoxidation levels and reduced SIRT1 expression and activity, similar MAO-A, but higher MAO-B activity in terms of M cells. Instead, the M + TA1 cells exhibited increased levels of SIRT1 protein and activity and significantly lower UCP-1, MAO-A, MAO-B, and SSAO in comparison with the M cells, and did not show signs of lipoperoxidation. Our results suggest that SIRT1 is the mediator of T1AM and TA1 pro-or anti-oxidant effects as a result of ROS intracellular levels, including the hydroxyl radical. Here, we provide evidence indicating that T1AM and TA1 administration impacts on the redox status of a biological system, a feature that indicates the novel mechanism of action of these two thyroid-hormone-related compounds.
Collapse
|
27
|
T1AM Attenuates the Hypoxia/Reoxygenation-Induced Necroptosis of H9C2 Cardiomyocytes via RIPK1/RIPK3 Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4833791. [PMID: 35265713 PMCID: PMC8901330 DOI: 10.1155/2022/4833791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/26/2022] [Indexed: 11/19/2022]
Abstract
Purpose To investigate the detailed mechanism of 3-iodothyronamine (T1AM) in cell apoptosis and programmed necrosis of hypoxia/reoxygenation- (H/R-) induced H9C2 injury. Materials and Methods Cardiomyocyte H9C2 cells were cultured in vitro for the establishment of cardiomyocyte H/R models. Cells were randomly divided into four groups: the control group, H/R group, T1AM pretreatment group, T1AM pretreatment and H/R (6 μm T1AM+H/R) group. The degree of myocardial injury was determined by the detection of the cardiomyocyte inhibition rate by CCK8 and the detection of lactic dehydrogenase (LDH) activity. Cell apoptosis was assessed through TUNEL assay and flow cytometry analysis. The protein level and mRNA level of RIPK1, RIPK3, and CAMKII were detected by western blotting and qRT-PCR. Results Compared with the control group, the cell inhibition rate was dramatically elevated in the H/R group. LDH release of cardiomyocytes was significantly increased. Protein and mRNA expressions of RIPK1, RIPK3, and CAMKII were significantly enhanced. Compared with the H/R group, the cell inhibition rate, LDH release, cardiomyocyte necroptosis rate, and protein and mRNA levels of RIPK1, RIPK3, and CAMKII of the T1AM+H/R group were significantly decreased. Conclusion Pretreatment with T1AM could alleviate cardiomyocytes' H/R injury and inhibit necroptosis of cardiomyocytes, which might exert a protective function upon activation of the RIPK1/RIPK3 pathway.
Collapse
|
28
|
Zhao M, Xie H, Shan H, Zheng Z, Li G, Li M, Hong L. Development of Thyroid Hormones and Synthetic Thyromimetics in Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:1102. [PMID: 35163026 PMCID: PMC8835192 DOI: 10.3390/ijms23031102] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the fastest-growing liver disease in the world. Despite targeted agents which are needed to provide permanent benefits for patients with NAFLD, no drugs have been approved to treat NASH. Thyroid hormone is an important signaling molecule to maintain normal metabolism, and in vivo and vitro studies have shown that regulation of the 3,5,3'-triiodothyronine (T3)/ thyroid hormone receptor (TR) axis is beneficial not only for metabolic symptoms but also for the improvement of NAFLD and even for the repair of liver injury. However, the non-selective regulation of T3 to TR subtypes (TRα/TRβ) could cause unacceptable side effects represented by cardiotoxicity. To avoid deleterious effects, TRβ-selective thyromimetics were developed for NASH studies in recent decades. Herein, we will review the development of thyroid hormones and synthetic thyromimetics based on TR selectivity for NAFLD, and analyze the role of TR-targeted drugs for the treatment of NAFLD in the future.
Collapse
Affiliation(s)
- Man Zhao
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.Z.); (H.X.); (H.S.); (Z.Z.)
| | - Huazhong Xie
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.Z.); (H.X.); (H.S.); (Z.Z.)
| | - Hao Shan
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.Z.); (H.X.); (H.S.); (Z.Z.)
| | - Zhihua Zheng
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.Z.); (H.X.); (H.S.); (Z.Z.)
| | - Guofeng Li
- Health Science Centre, School of Pharmaceutical Sciences, Shenzhen University, Shenzhen 518060, China;
| | - Min Li
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.Z.); (H.X.); (H.S.); (Z.Z.)
| | - Liang Hong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (M.Z.); (H.X.); (H.S.); (Z.Z.)
| |
Collapse
|
29
|
Nguyen YTK, Ha HTT, Nguyen TH, Nguyen LN. The role of SLC transporters for brain health and disease. Cell Mol Life Sci 2021; 79:20. [PMID: 34971415 PMCID: PMC11071821 DOI: 10.1007/s00018-021-04074-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/05/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022]
Abstract
The brain exchanges nutrients and small molecules with blood via the blood-brain barrier (BBB). Approximately 20% energy intake for the body is consumed by the brain. Glucose is known for its critical roles for energy production and provides substrates for biogenesis in neurons. The brain takes up glucose via glucose transporters GLUT1 and 3, which are expressed in several neural cell types. The brain is also equipped with various transport systems for acquiring amino acids, lactate, ketone bodies, lipids, and cofactors for neuronal functions. Unraveling the mechanisms by which the brain takes up and metabolizes these nutrients will be key in understanding the nutritional requirements in the brain. This could also offer opportunities for therapeutic interventions in several neurological disorders. For instance, emerging evidence suggests a critical role of lactate as an alternative energy source for neurons. Neuronal cells express monocarboxylic transporters to acquire lactate. As such, treatment of GLUT1-deficient patients with ketogenic diets to provide the brain with alternative sources of energy has been shown to improve the health of the patients. Many transporters are present in the brain, but only a small number has been characterized. In this review, we will discuss about the roles of solute carrier (SLC) transporters at the blood brain barrier (BBB) and neural cells, in transport of nutrients and metabolites in the brain.
Collapse
Affiliation(s)
- Yen T K Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Hoa T T Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Tra H Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
- SLING/Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- Immunology Translational and Cardiovascular Disease Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.
| |
Collapse
|
30
|
The 3-iodothyronamine (T1AM) and the 3-iodothyroacetic acid (TA1) indicate a novel connection with the histamine system for neuroprotection. Eur J Pharmacol 2021; 912:174606. [PMID: 34717926 DOI: 10.1016/j.ejphar.2021.174606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022]
Abstract
The 3-iodothyronamine (T1AM) and 3-iodothryoacetic acid (TA1), are endogenous occurring compounds structurally related with thyroid hormones (THs, the pro-hormone T4 and the active hormone T3) initially proposed as possible mediators of the rapid effects of T3. However, after years from their identification, the physio-pathological meaning of T1AM and TA1 tissue levels remains an unsolved issue while pharmacological evidence indicates both compounds promote in rodents central and peripheral effects with mechanisms which remain mostly elusive. Pharmacodynamics of T1AM includes the recognition of G-coupled receptors, ion channels but also biotransformation into an active metabolite, i.e. the TA1. Furthermore, long term T1AM treatment associates with post-translational modifications of cell proteins. Such array of signaling may represent an added value, rather than a limit, equipping T1AM to play different functions depending on local expression of targets and enzymes involved in its biotransformation. Up to date, no information regarding TA1 mechanistic is available. We here review some of the main findings describing effects of T1AM (and TA1) which suggest these compounds interplay with the histaminergic system. These data reveal T1AM and TA1 are part of a network of signals involved in neuronal plasticity including neuroprotection and suggest T1AM and TA1 as lead compounds for a novel class of atypical psychoactive drugs.
Collapse
|
31
|
Louzada RA, Padron AS, Marques-Neto SR, Maciel L, Werneck-de-Castro JP, Ferreira ACF, Nascimento JHM, Carvalho DP. 3,5-Diiodothyronine protects against cardiac ischaemia-reperfusion injury in male rats. Exp Physiol 2021; 106:2185-2197. [PMID: 34605090 DOI: 10.1113/ep089589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? 3,5-Diiodothyronine (3,5-T2) administration increases resting metabolic rate, prevents or treats liver steatosis in rodent models, and ameliorates insulin resistance: what are its effects on cardiac electrical and contractile properties and autonomic regulation? What is the main finding and its importance? Chronic 3,5-T2 administration has no adverse effects on cardiac function. Remarkably, 3,5-T2 improves the autonomous control of the rat heart and protects against ischaemia-reperfusion injury. ABSTRACT The use of 3,5,3'-triiodothyronine (T3) and thyroxine (T4) to treat metabolic diseases has been hindered by potential adverse effects on liver, lipid metabolism and cardiac electrical properties. It is recognized that 3,5-diiodothyronine (3,5-T2) administration increases resting metabolic rate, prevents or treats liver steatosis in rodent models and ameliorates insulin resistance, suggesting 3,5-T2 as a potential therapeutic tool. However, a comprehensive assessment of cardiac electrical and contractile properties has not been made so far. Three-month-old Wistar rats were daily administered vehicle, 3,5-T2 or 3,5-T2+T4 and no signs of atrial or ventricular arrhythmia were detected in non-anaesthetized rats during 90 days. Cardiac function was preserved as heart rate, left ventricle diameter and shortening fraction in 3,5-T2-treated rats compared to vehicle and 3,5-T2+T4 groups. Power spectral analysis indicated an amelioration of the heart rate variability only in 3,5-T2-treated rats. An increased baroreflex sensitivity at rest was observed in both 3,5-T2-treated groups. Finally, 3,5-T2 Langendorff-perfused hearts presented a significant recovery of left ventricular function and remarkably smaller infarction area after ischaemia-reperfusion injury. In conclusion, chronic 3,5-T2 administration ameliorates tonic cardiac autonomic control and confers cardioprotection against ischaemia-reperfusion injury in healthy male rats.
Collapse
Affiliation(s)
- Ruy Andrade Louzada
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Biologia do Exercício, Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alvaro Souto Padron
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Silvio Rodrigues Marques-Neto
- Laboratório de Biologia do Exercício, Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Ciências da Atividade Física, Niterói, RJ, Brazil.,Universidade Estácio de Sá (UNESA), Laboratório de Fisiologia do Exercício (LAFIEX), Curso de Educação Física, Rio de Janeiro, Brazil
| | - Leonardo Maciel
- Laboratório de Biologia do Exercício, Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,NUMPEX, Campus Duque de Caxias, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João Pedro Werneck-de-Castro
- Laboratório de Biologia do Exercício, Escola de Educação Física e Desportos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Division of Endocrinology, Diabetes and Metabolism, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Andrea Claudia Freitas Ferreira
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,NUMPEX, Campus Duque de Caxias, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose Hamilton Matheus Nascimento
- Laboratório de Eletrofisiologia Cardíaca Antonio Paes de Carvalho, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise Pires Carvalho
- Laboratório de Fisiologia Endócrina Doris Rosenthal, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Trace Amine-Associated Receptor 1 Contributes to Diverse Functional Actions of O-Phenyl-Iodotyramine in Mice but Not to the Effects of Monoamine-Based Antidepressants. Int J Mol Sci 2021; 22:ijms22168907. [PMID: 34445611 PMCID: PMC8396211 DOI: 10.3390/ijms22168907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 12/29/2022] Open
Abstract
Trace Amine-Associated Receptor 1 (TAAR1) is a potential target for the treatment of depression and other CNS disorders. However, the precise functional roles of TAAR1 to the actions of clinically used antidepressants remains unclear. Herein, we addressed these issues employing the TAAR1 agonist, o-phenyl-iodotyramine (o-PIT), together with TAAR1-knockout (KO) mice. Irrespective of genotype, systemic administration of o-PIT led to a similar increase in mouse brain concentrations. Consistent with the observation of a high density of TAAR1 in the medial preoptic area, o-PIT-induced hypothermia was significantly reduced in TAAR1-KO mice. Furthermore, the inhibition of a prepulse inhibition response by o-PIT, as well as its induction of striatal tyrosine hydroxylase phosphorylation and elevation of extracellular DA in prefrontal cortex, were all reduced in TAAR1-KO compared to wildtype mice. O-PIT was active in both forced-swim and marble-burying tests, and its effects were significantly blunted in TAAR1-KO mice. Conversely, the actions on behaviour and prefrontal cortex dialysis of a broad suite of clinically used antidepressants were unaffected in TAAR1-KO mice. In conclusion, o-PIT is a useful tool for exploring the hypothermic and other functional antidepressant roles of TAAR1. By contrast, clinically used antidepressants do not require TAAR1 for expression of their antidepressant properties.
Collapse
|
33
|
Vaganova AN, Murtazina RZ, Shemyakova TS, Prjibelski AD, Katolikova NV, Gainetdinov RR. Pattern of TAAR5 Expression in the Human Brain Based on Transcriptome Datasets Analysis. Int J Mol Sci 2021; 22:ijms22168802. [PMID: 34445502 PMCID: PMC8395715 DOI: 10.3390/ijms22168802] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022] Open
Abstract
Trace amine-associated receptors (TAAR) recognize organic compounds, including primary, secondary, and tertiary amines. The TAAR5 receptor is known to be involved in the olfactory sensing of innate socially relevant odors encoded by volatile amines. However, emerging data point to the involvement of TAAR5 in brain functions, particularly in the emotional behaviors mediated by the limbic system which suggests its potential contribution to the pathogenesis of neuropsychiatric diseases. TAAR5 expression was explored in datasets available in the Gene Expression Omnibus, Allen Brain Atlas, and Human Protein Atlas databases. Transcriptomic data demonstrate ubiquitous low TAAR5 expression in the cortical and limbic brain areas, the amygdala and the hippocampus, the nucleus accumbens, the thalamus, the hypothalamus, the basal ganglia, the cerebellum, the substantia nigra, and the white matter. Altered TAAR5 expression is identified in Down syndrome, major depressive disorder, or HIV-associated encephalitis. Taken together, these data indicate that TAAR5 in humans is expressed not only in the olfactory system but also in certain brain structures, including the limbic regions receiving olfactory input and involved in critical brain functions. Thus, TAAR5 can potentially be involved in the pathogenesis of brain disorders and represents a valuable novel target for neuropsychopharmacology.
Collapse
Affiliation(s)
- Anastasia N. Vaganova
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.); (R.Z.M.); (T.S.S.); (A.D.P.); (N.V.K.)
| | - Ramilya Z. Murtazina
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.); (R.Z.M.); (T.S.S.); (A.D.P.); (N.V.K.)
| | - Taisiia S. Shemyakova
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.); (R.Z.M.); (T.S.S.); (A.D.P.); (N.V.K.)
| | - Andrey D. Prjibelski
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.); (R.Z.M.); (T.S.S.); (A.D.P.); (N.V.K.)
| | - Nataliia V. Katolikova
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.); (R.Z.M.); (T.S.S.); (A.D.P.); (N.V.K.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia; (A.N.V.); (R.Z.M.); (T.S.S.); (A.D.P.); (N.V.K.)
- St. Petersburg University Hospital, St. Petersburg State University, Universitetskaya nab. 7/9, 199034 St. Petersburg, Russia
- Correspondence:
| |
Collapse
|
34
|
Vogelsang TLR, Vattai A, Schmoeckel E, Kaltofen T, Chelariu-Raicu A, Zheng M, Mahner S, Mayr D, Jeschke U, Trillsch F. Trace Amine-Associated Receptor 1 (TAAR1) Is a Positive Prognosticator for Epithelial Ovarian Cancer. Int J Mol Sci 2021; 22:ijms22168479. [PMID: 34445181 PMCID: PMC8395182 DOI: 10.3390/ijms22168479] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022] Open
Abstract
Trace amine-associated receptor 1 (TAAR1) is a Gαs- protein coupled receptor that plays an important role in the regulation of the immune system and neurotransmission in the CNS. In ovarian cancer cell lines, stimulation of TAAR1 via 3-iodothyronamine (T1AM) reduces cell viability and induces cell death and DNA damage. Aim of this study was to evaluate the prognostic value of TAAR1 on overall survival of ovarian carcinoma patients and the correlation of TAAR1 expression with clinical parameters. Ovarian cancer tissue of n = 156 patients who were diagnosed with epithelial ovarian cancer (serous, n = 110 (high-grade, n = 80; low-grade, n = 24; unknown, n = 6); clear cell, n = 12; endometrioid, n = 21; mucinous, n = 13), and who underwent surgery at the Department of Obstetrics and Gynecology, University Hospital of the Ludwig-Maximilians University Munich, Germany between 1990 and 2002, were analyzed. The tissue was stained immunohistochemically with anti-TAAR1 and evaluated with the semiquantitative immunoreactive score (IRS). TAAR1 expression was correlated with grading, FIGO and TNM-classification, and analyzed via the Spearman’s rank correlation coefficient. Further statistical analysis was obtained using nonparametric Kruskal-Wallis rank-sum test and Mann-Whitney-U-test. This study shows that high TAAR1 expression is a positive prognosticator for overall survival in ovarian cancer patients and is significantly enhanced in low-grade serous carcinomas compared to high-grade serous carcinomas. The influence of TAAR1 as a positive prognosticator on overall survival indicates a potential prognostic relevance of signal transduction of thyroid hormone derivatives in epithelial ovarian cancer. Further studies are required to evaluate TAAR1 and its role in the development of ovarian cancer.
Collapse
Affiliation(s)
- Tilman L. R. Vogelsang
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, 80337 Munich, Germany; (T.L.R.V.); (A.V.); (T.K.); (A.C.-R.); (M.Z.); (S.M.); (F.T.)
| | - Aurelia Vattai
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, 80337 Munich, Germany; (T.L.R.V.); (A.V.); (T.K.); (A.C.-R.); (M.Z.); (S.M.); (F.T.)
| | - Elisa Schmoeckel
- Faculty of Medicine, Institute of Pathology, LMU Munich, 80337 Munich, Germany; (E.S.); (D.M.)
| | - Till Kaltofen
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, 80337 Munich, Germany; (T.L.R.V.); (A.V.); (T.K.); (A.C.-R.); (M.Z.); (S.M.); (F.T.)
| | - Anca Chelariu-Raicu
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, 80337 Munich, Germany; (T.L.R.V.); (A.V.); (T.K.); (A.C.-R.); (M.Z.); (S.M.); (F.T.)
| | - Mingjun Zheng
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, 80337 Munich, Germany; (T.L.R.V.); (A.V.); (T.K.); (A.C.-R.); (M.Z.); (S.M.); (F.T.)
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, 80337 Munich, Germany; (T.L.R.V.); (A.V.); (T.K.); (A.C.-R.); (M.Z.); (S.M.); (F.T.)
| | - Doris Mayr
- Faculty of Medicine, Institute of Pathology, LMU Munich, 80337 Munich, Germany; (E.S.); (D.M.)
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, 80337 Munich, Germany; (T.L.R.V.); (A.V.); (T.K.); (A.C.-R.); (M.Z.); (S.M.); (F.T.)
- Department of Obstetrics and Gynecology, University Hospital Augsburg, 86156 Augsburg, Germany
- Correspondence: ; Tel.: +49-89-4400-74775
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, 80337 Munich, Germany; (T.L.R.V.); (A.V.); (T.K.); (A.C.-R.); (M.Z.); (S.M.); (F.T.)
| |
Collapse
|
35
|
Role of trace amine‑associated receptor 1 in the medial prefrontal cortex in chronic social stress-induced cognitive deficits in mice. Pharmacol Res 2021; 167:105571. [PMID: 33753244 DOI: 10.1016/j.phrs.2021.105571] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022]
Abstract
Emerging evidence supports an essential role of trace amine-associated receptor 1 (TAAR1) in neuropsychiatric disorders such as depression and schizophrenia. Stressful events are critical contributors to various neuropsychiatric disorders. This study examined the role of TAAR1 in mediating the negative outcomes of stressful events. In mice that experienced chronic social defeat stress but not acute stress, a significant reduction in the TAAR1 mRNA level was found in the medial prefrontal cortex (mPFC), a brain region that is known to be vulnerable to stress experience. Conditional TAAR1 knockout in the mPFC mimicked the cognitive deficits induced by chronic stress. In addition, chronic treatment with the selective TAAR1 partial agonist RO5263397 ameliorated chronic stress-induced changes in cognitive function, dendritic arborization, and the synapse number of pyramidal neurons in the mPFC but did not affect chronic stress-induced anxiety-like behaviors. Biochemically, chronic stress reduced the ratio of vesicular transporters of glutamate-1 (VGluT1) / vesicular GABA transporter (VGAT) in the mPFC,most prominently in the prelimbic cortex, and RO5263397 restored the excitatory-inhibitory (E/I) imbalance. Together, the results of this study reveal an essential role of TAAR1 in mediating chronic stress-induced cognitive impairments and suggest that TAAR1 agonists may be uniquely useful to treat MDD-related cognitive impairments.
Collapse
|
36
|
di Leo N, Moscato S, Borso’ M, Sestito S, Polini B, Bandini L, Grillone A, Battaglini M, Saba A, Mattii L, Ciofani G, Chiellini G. Delivery of Thyronamines (TAMs) to the Brain: A Preliminary Study. Molecules 2021; 26:molecules26061616. [PMID: 33799468 PMCID: PMC7999687 DOI: 10.3390/molecules26061616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 12/21/2022] Open
Abstract
Recent reports highlighted the significant neuroprotective effects of thyronamines (TAMs), a class of endogenous thyroid hormone derivatives. In particular, 3-iodothyronamine (T1AM) has been shown to play a pleiotropic role in neurodegeneration by modulating energy metabolism and neurological functions in mice. However, the pharmacological response to T1AM might be influenced by tissue metabolism, which is known to convert T1AM into its catabolite 3-iodothyroacetic acid (TA1). Currently, several research groups are investigating the pharmacological effects of T1AM systemic administration in the search of novel therapeutic approaches for the treatment of interlinked pathologies, such as metabolic and neurodegenerative diseases (NDDs). A critical aspect in the development of new drugs for NDDs is to know their distribution in the brain, which is fundamentally related to their ability to cross the blood–brain barrier (BBB). To this end, in the present study we used the immortalized mouse brain endothelial cell line bEnd.3 to develop an in vitro model of BBB and evaluate T1AM and TA1 permeability. Both drugs, administered at 1 µM dose, were assayed by high-performance liquid chromatography coupled to mass spectrometry. Our results indicate that T1AM is able to efficiently cross the BBB, whereas TA1 is almost completely devoid of this property.
Collapse
Affiliation(s)
- Nicoletta di Leo
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy or (N.d.L.); (S.M.); (A.G.); (M.B.); (G.C.)
- The Biorobotics Institute, Scuola Superiore Sant’Anna, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy
| | - Stefania Moscato
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy or (N.d.L.); (S.M.); (A.G.); (M.B.); (G.C.)
- Department of Clinical & Experimental Medicine, University of Pisa, Via Savi 10, 56126 Pisa, Italy;
| | - Marco Borso’
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (M.B.); or (S.S.); (B.P.); (L.B.) (A.S.)
| | - Simona Sestito
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (M.B.); or (S.S.); (B.P.); (L.B.) (A.S.)
- Department of Chemistry and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Beatrice Polini
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (M.B.); or (S.S.); (B.P.); (L.B.) (A.S.)
| | - Lavinia Bandini
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (M.B.); or (S.S.); (B.P.); (L.B.) (A.S.)
| | - Agostina Grillone
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy or (N.d.L.); (S.M.); (A.G.); (M.B.); (G.C.)
| | - Matteo Battaglini
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy or (N.d.L.); (S.M.); (A.G.); (M.B.); (G.C.)
| | - Alessandro Saba
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (M.B.); or (S.S.); (B.P.); (L.B.) (A.S.)
| | - Letizia Mattii
- Department of Clinical & Experimental Medicine, University of Pisa, Via Savi 10, 56126 Pisa, Italy;
| | - Gianni Ciofani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy or (N.d.L.); (S.M.); (A.G.); (M.B.); (G.C.)
| | - Grazia Chiellini
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (M.B.); or (S.S.); (B.P.); (L.B.) (A.S.)
- Correspondence:
| |
Collapse
|
37
|
Li ZM, Miller M, Gachkar S, Mittag J, Schriever SC, Pfluger PT, Schramm KW, De Angelis M. Determination of 3-iodothyronamine (3-T 1AM) in mouse liver using liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1165:122553. [PMID: 33503577 DOI: 10.1016/j.jchromb.2021.122553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/09/2020] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
Abstract
3-iodothyronamine (3-T1AM) has been suggested as a novel chemical messenger and potent trace amine-associated receptor 1 ligand in the CNS that occurs naturally as endogenous metabolite of the thyroid hormones. Discrepancies and variations in 3-T1AM plasma and tissue concentrations have nonetheless caused controversy regarding the existence and biological role of 3-T1AM. These discussions are at least partially based on potential analytical artefacts caused by differential decay kinetics of 3-T1AM and the widely used deuterated quantification standard D4-T1AM. Here, we report a novel LC-MS/MS method for the quantification of 3-T1AM in biological specimens using stable isotope dilution with 13C6-T1AM, a new internal standard that showed pharmacodynamic properties comparable to endogenous 3-T1AM. The method detection limit (MDL) and method quantification limit (MQL) of 3-T1AM were 0.04 and 0.09 ng/g, respectively. The spike-recoveries of 3-T1AM were between 85.4% and 94.3%, with a coefficient of variation of 3.7-5.8%. The intra-day and inter-day variations of 3-T1AM were 8.45-11.2% and 3.58-5.73%, respectively. Endogenous 3-T1AM liver values in C57BL/6J mice were 2.20 ± 0.49 pmol/g with a detection frequency of 50%. Higher liver 3-T1AM values were found when C57BL/6J mice were treated with N-acetyl-3-iodothyronamine or O-acetyl-3-iodothyronamine. Overall, our new stable isotope dilution LC-MS/MS method improves both the sensitivity and selectivity compared with existing methods. The concomitant possibility to quantify additional thyroid hormones such as thyroxine, 3,5,3'-triiodo-L-thyronine, 3,3',5'-triiodo-L-thyronine, 3,3'-diiodo-L-thyronine, and 3,5-diiodo-L-thyronine further adds to the value of our novel method in exploring the natural occurrence and fate of 3-T1AM in biological tissues and fluids.
Collapse
Affiliation(s)
- Zhong-Min Li
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Molecular EXposomics, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; School of Life Sciences Weihenstephan (Nutrition), Technical University Munich, 85354 Freising, Germany.
| | - Manuel Miller
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Research Unit Comparative Medicine (AVM), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Sogol Gachkar
- Institute for Endocrinology & Diabetes, Molecular Endocrinology, Center of Brain, Behavior & Metabolism, University of Lübeck, Lübeck, Germany
| | - Jens Mittag
- Institute for Endocrinology & Diabetes, Molecular Endocrinology, Center of Brain, Behavior & Metabolism, University of Lübeck, Lübeck, Germany
| | - Sonja C Schriever
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Germany; Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, Neuherberg, Germany
| | - Paul T Pfluger
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Germany; Research Unit Neurobiology of Diabetes, Helmholtz Zentrum München, Neuherberg, Germany; TUM School of Medicine, Neurobiology of Diabetes, Technical University Munich, Germany
| | - Karl-Werner Schramm
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Molecular EXposomics, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Department für Biowissenschaftliche Grundlagen, Technische Universität München, Weihenstephaner Steig 23, 85350 Freising, Germany
| | - Meri De Angelis
- Helmholtz Zentrum München-German Research Center for Environmental Health (GmbH), Molecular EXposomics, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| |
Collapse
|
38
|
Tozzi F, Rutigliano G, Borsò M, Falcicchia C, Zucchi R, Origlia N. T 1AM-TAAR1 signalling protects against OGD-induced synaptic dysfunction in the entorhinal cortex. Neurobiol Dis 2021; 151:105271. [PMID: 33482355 DOI: 10.1016/j.nbd.2021.105271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/04/2020] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
Abnormalities in thyroid hormones (TH) availability and/or metabolism have been hypothesized to contribute to Alzheimer's disease (AD) and to be a risk factor for stroke. Recently, 3-iodothyronamine (T1AM), an endogenous amine putatively derived from TH metabolism, gained interest for its ability to promote learning and memory in the mouse. Moreover, T1AM has been demonstrated to rescue the β-Amyloid dependent LTP impairment in the entorhinal cortex (EC), a brain area crucially involved in learning and memory and early affected during AD. In the present work, we have investigated the effect of T1AM on ischemia-induced EC synaptic dysfunction. In EC brain slices exposed to oxygen-glucose deprivation (OGD), we demonstrated that the acute perfusion of T1AM (5 μM) was capable of preventing ischemia-induced synaptic depression and that this protective effect was mediated by the trace amine-associated receptor 1 (TAAR1). Moreover, we demonstrated that activation of the BDNF-TrkB signalling is required for T1AM action during ischemia. The protective effect of T1AM was more evident when using EC slices from transgenic mutant human APP (mhAPP mice) that are more vulnerable to the effect of OGD. Our results confirm that the TH derivative T1AM can rescue synaptic function after transient ischemia, an effect that was also observed in a Aβ-enriched environment.
Collapse
Affiliation(s)
- Francesca Tozzi
- Bio@SNS laboratory, Scuola Normale Superiore, 56124 Pisa, Italy
| | | | - Marco Borsò
- Department of Pathology, University of Pisa, 56100 Pisa, Italy
| | - Chiara Falcicchia
- Institute of Neuroscience of the Italian National Research Council (CNR), Pisa, Italy
| | - Riccardo Zucchi
- Department of Pathology, University of Pisa, 56100 Pisa, Italy
| | - Nicola Origlia
- Institute of Neuroscience of the Italian National Research Council (CNR), Pisa, Italy.
| |
Collapse
|
39
|
Frare C, Williams CT, Drew KL. Thermoregulation in hibernating mammals: The role of the "thyroid hormones system". Mol Cell Endocrinol 2021; 519:111054. [PMID: 33035626 PMCID: PMC8091518 DOI: 10.1016/j.mce.2020.111054] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 07/15/2020] [Accepted: 10/04/2020] [Indexed: 12/19/2022]
Abstract
Hibernation is a unique evolutionary adaptation to conserve energy. During the pre-hibernation (i.e. fall) season, a progressive decline in core body temperature and further decrease in metabolism underlie a seasonal modulation in thermoregulation. The onset of hibernation requires marked changes in thermoregulatory attributes including adjustment in body temperature and tissue specific increases in thermogenic capacity. The hibernation season is characterized by a regulated suppression in thermogenesis allowing the onset of torpor interrupted by periodic activation of thermogenesis to sustain interbout arousals. Thyroid hormones are known to regulate both body temperature and metabolism, and for this reason, the hypothalamic-pituitary-thyroid axis and thyroid hormones have been investigated as modulators of thermogenesis in the phenomenon of hibernation, but the mechanisms remain poorly understood. In this review, we present an overview of what is known about the thermogenic roles of thyroid hormones in hibernating species across seasons and within the hibernating season (torpor-interbout arousal cycle). Overall, the hypothalamic-pituitary-thyroid axis and thyroid hormones play a role in the pre-hibernation season to enhance thermogenic capacity. During hibernation, thermogenesis is attenuated at the level of sympathetic premotor neurons within the raphe pallidus and by deiodinase expression in the hypothalamus. Further, as recent work highlights the direct effect of thyroid hormones within the central nervous system in activating thermogenesis, we speculate how similar mechanisms may occur in hibernating species to modulate thermogenesis across seasons and to sustain interbout arousals. However, further experiments are needed to elucidate the role of thyroid hormones in hibernation, moving towards the understanding that thyroid hormones metabolism, transport and availability within tissues may be the most telling indicator of thyroid status.
Collapse
Affiliation(s)
- C Frare
- Department of Chemistry and Biochemistry University of Alaska Fairbanks, Fairbanks, AK, 99775, USA; Institute of Arctic Biology, Center for Transformative Research in Metabolism, University of Alaska Fairbanks 2140 Koyukuk Drive, Fairbanks, AK, 99775, USA
| | - Cory T Williams
- Institute of Arctic Biology, Center for Transformative Research in Metabolism, University of Alaska Fairbanks 2140 Koyukuk Drive, Fairbanks, AK, 99775, USA; Department of Biology and Wildlife, University of Alaska Fairbanks, Fairbanks, AK, 99775, USA
| | - Kelly L Drew
- Department of Chemistry and Biochemistry University of Alaska Fairbanks, Fairbanks, AK, 99775, USA; Institute of Arctic Biology, Center for Transformative Research in Metabolism, University of Alaska Fairbanks 2140 Koyukuk Drive, Fairbanks, AK, 99775, USA.
| |
Collapse
|
40
|
Shi Z, Qin M, Huang L, Xu T, Chen Y, Hu Q, Peng S, Peng Z, Qu LN, Chen SG, Tuo QH, Liao DF, Wang XP, Wu RR, Yuan TF, Li YH, Liu XM. Human torpor: translating insights from nature into manned deep space expedition. Biol Rev Camb Philos Soc 2020; 96:642-672. [PMID: 33314677 DOI: 10.1111/brv.12671] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/09/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
During a long-duration manned spaceflight mission, such as flying to Mars and beyond, all crew members will spend a long period in an independent spacecraft with closed-loop bioregenerative life-support systems. Saving resources and reducing medical risks, particularly in mental heath, are key technology gaps hampering human expedition into deep space. In the 1960s, several scientists proposed that an induced state of suppressed metabolism in humans, which mimics 'hibernation', could be an ideal solution to cope with many issues during spaceflight. In recent years, with the introduction of specific methods, it is becoming more feasible to induce an artificial hibernation-like state (synthetic torpor) in non-hibernating species. Natural torpor is a fascinating, yet enigmatic, physiological process in which metabolic rate (MR), body core temperature (Tb ) and behavioural activity are reduced to save energy during harsh seasonal conditions. It employs a complex central neural network to orchestrate a homeostatic state of hypometabolism, hypothermia and hypoactivity in response to environmental challenges. The anatomical and functional connections within the central nervous system (CNS) lie at the heart of controlling synthetic torpor. Although progress has been made, the precise mechanisms underlying the active regulation of the torpor-arousal transition, and their profound influence on neural function and behaviour, which are critical concerns for safe and reversible human torpor, remain poorly understood. In this review, we place particular emphasis on elaborating the central nervous mechanism orchestrating the torpor-arousal transition in both non-flying hibernating mammals and non-hibernating species, and aim to provide translational insights into long-duration manned spaceflight. In addition, identifying difficulties and challenges ahead will underscore important concerns in engineering synthetic torpor in humans. We believe that synthetic torpor may not be the only option for manned long-duration spaceflight, but it is the most achievable solution in the foreseeable future. Translating the available knowledge from natural torpor research will not only benefit manned spaceflight, but also many clinical settings attempting to manipulate energy metabolism and neurobehavioural functions.
Collapse
Affiliation(s)
- Zhe Shi
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.,Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Meng Qin
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lu Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
| | - Tao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qin Hu
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, Beijing, 100024, China
| | - Sha Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhuang Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Li-Na Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Shan-Guang Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Qin-Hui Tuo
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Duan-Fang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xiao-Ping Wang
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ren-Rong Wu
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China
| | - Ying-Hui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xin-Min Liu
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| |
Collapse
|
41
|
Novel 1-Amidino-4-Phenylpiperazines as Potent Agonists at Human TAAR1 Receptor: Rational Design, Synthesis, Biological Evaluation and Molecular Docking Studies. Pharmaceuticals (Basel) 2020; 13:ph13110391. [PMID: 33202687 PMCID: PMC7697893 DOI: 10.3390/ph13110391] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/19/2022] Open
Abstract
Targeting trace amine-associated receptor 1 (TAAR1) receptor continues to offer an intriguing opportunity to develop innovative therapies in different pharmacological settings. Pursuing our endeavors in the search for effective and safe human TAAR1 (hTAAR1) ligands, we synthesized a new series of 1-amidino-4-phenylpiperazine derivatives (1–16) based on the application of a combined pharmacophore model/scaffold simplification strategy for an in-house series of biguanide-based TAAR1 agonists. Most of the novel compounds proved to be more effective than their prototypes, showing nanomolar EC50 values in functional activity at hTAAR1 and low general cytotoxicity (CC50 > 80 µM) when tested on the Vero-76 cell line. In this new series, the main determinant for TAAR1 agonism ability appears to result from the appropriate combination between the steric size and position of the substituents on the phenyl ring rather than from their different electronic nature, since both electron-withdrawing and electron donor groups are permitted. In particular, the ortho-substitution seems to impose a more appropriate spatial geometry to the molecule that entails an enhanced TAAR1 potency profile, as experienced, in the following order, by compounds 15 (2,3-diCl, EC50 = 20 nM), 2 (2-CH3, EC50 = 30 nM), 6 (2-OCH3, EC50 = 93 nM) and 3 (2-Cl, EC50 = 160 nM). Apart from the interest in them as valuable leads for the development of promising hTAAR1 agonists, these simple small molecules have further allowed us to identify the minimal structural requirements for producing an efficient hTAAR1 targeting ability.
Collapse
|
42
|
Biebermann H, Brix K, Führer D. Seven Years of Active Thyroid Hormone Research in Germany: Thyroid
Hormone Action beyond Classical Concepts. Exp Clin Endocrinol Diabetes 2020; 128:355-357. [DOI: 10.1055/a-1163-7355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Heike Biebermann
- Charité – Universitätsmedizin Berlin, corporate
member of Freie Universität Berlin, Humboldt-Universität zu
Berlin, and Berlin Institute of Health; Institut für Experimentelle
Pädiatrische Endokrinologie, Berlin, Germany
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Jacobs University Bremen,
Bremen, Germany
| | - Dagmar Führer
- Universität Duisburg-Essen, Universitätsklinikum Essen,
Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Zentrallabor -
Bereich Forschung und Lehre, Essen, Germany
| |
Collapse
|
43
|
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
|
44
|
Gellrich L, Heitel P, Heering J, Kilu W, Pollinger J, Goebel T, Kahnt A, Arifi S, Pogoda W, Paulke A, Steinhilber D, Proschak E, Wurglics M, Schubert-Zsilavecz M, Chaikuad A, Knapp S, Bischoff I, Fürst R, Merk D. l-Thyroxin and the Nonclassical Thyroid Hormone TETRAC Are Potent Activators of PPARγ. J Med Chem 2020; 63:6727-6740. [DOI: 10.1021/acs.jmedchem.9b02150] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Leonie Gellrich
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Pascal Heitel
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Jan Heering
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Whitney Kilu
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Julius Pollinger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Tamara Goebel
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Astrid Kahnt
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Silvia Arifi
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Werner Pogoda
- Department of Forensic Toxicology, Institute of Forensic Medicine, Goethe University Frankfurt, Kennedyallee 104, D-60596 Frankfurt, Germany
| | - Alexander Paulke
- Department of Forensic Toxicology, Institute of Forensic Medicine, Goethe University Frankfurt, Kennedyallee 104, D-60596 Frankfurt, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, D-60596 Frankfurt, Germany
| | - Mario Wurglics
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Manfred Schubert-Zsilavecz
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Apirat Chaikuad
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, D-60438 Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
- Structural Genomics Consortium, Buchmann Institute for Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, D-60438 Frankfurt, Germany
| | - Iris Bischoff
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany
| |
Collapse
|
45
|
3-Iodothyronamine and Derivatives: New Allies Against Metabolic Syndrome? Int J Mol Sci 2020; 21:ijms21062005. [PMID: 32183490 PMCID: PMC7139928 DOI: 10.3390/ijms21062005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/02/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022] Open
Abstract
In the two decades since its discovery, a large body of evidence has amassed to highlight the potential of 3-iodothyronamine (T1AM) as an antiobesity drug, whose pleiotropic signaling actions profoundly impact energy metabolism. In the present review, we recapitulate the most relevant properties of T1AM, including its structural and functional relationship to thyroid hormone, its endogenous levels, molecular targets, as well as its genomic and non-genomic effects on metabolism elicited in experimental models after exogenous administration. The physiological and pathophysiological relevance of T1AM in the regulation of energy homeostasis and metabolism is also discussed, along with its potential therapeutic applications in metabolic disturbances. Finally, we examine a number of T1AM analogs that have been recently developed with the aim of designing novel pharmacological agents for the treatment of interlinked diseases, such as metabolic and neurodegenerative disorders, as well as additional synthetic tools that can be exploited to further explore T1AM-dependent mechanisms and the physiological roles of trace amine-associated receptor 1 (TAAR1)-mediated effects.
Collapse
|
46
|
Sacripanti G, Lorenzini L, Bandini L, Frascarelli S, Zucchi R, Ghelardoni S. 3-Iodothyronamine and 3,5,3'-triiodo-L-thyronine reduce SIRT1 protein expression in the HepG2 cell line. Horm Mol Biol Clin Investig 2020; 41:/j/hmbci.ahead-of-print/hmbci-2019-0045/hmbci-2019-0045.xml. [PMID: 32114521 DOI: 10.1515/hmbci-2019-0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/13/2019] [Indexed: 11/15/2022]
Abstract
Background 3-Iodothyronamine (T1AM) is an endogenous messenger chemically related to thyroid hormone. Recent results indicate significant transcriptional effects of chronic T1AM administration involving the protein family of sirtuins, which regulate important metabolic pathways and tumor progression. Therefore, the aim of this work was to compare the effect of exogenous T1AM and 3,5,3'-triiodo-L-thyronine (T3) chronic treatment on mammalian sirtuin expression in hepatocellular carcinoma cells (HepG2) and in primary rat hepatocytes at micromolar concentrations. Materials and methods Sirtuin (SIRT) activity and expression were determined using a colorimetric assay and Western blot analysis, respectively, in cells treated for 24 h with 1-20 μM T1AM or T3. In addition, cell viability was evaluated by the MTTtest upon 24 h of treatment with 0.1-20 μM T1AM or T3. Results In HepG2, T1AM significantly reduced SIRT 1 (20 μM) and SIRT4 (10-20 μM) protein expression, while T3 strongly decreased the expression of SIRT1 (20 μM) and SIRT2 (any tested concentration). In primary rat hepatocytes, T3 decreased SIRT2 expression and cellular nicotinamide adenine dinucleotide (NAD) concentration, while on sirtuin activity it showed opposite effects, depending on the evaluated cell fraction. The extent of MTT staining was moderately but significantly reduced by T1AM, particularly in HepG2 cells, whereas T3 reduced cell viability only in the tumor cell line. Conclusions T1AM and T3 downregulated the expression of sirtuins, mainly SIRT1, in hepatocytes, albeit in different ways. Differences in mechanisms are only observational, and further investigations are required to highlight the potential role of T1AM and T3 in modulating sirtuin expression and, therefore, in regulating cell cycle or tumorigenesis.
Collapse
Affiliation(s)
- Ginevra Sacripanti
- Department of Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Leonardo Lorenzini
- Department of Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Lavinia Bandini
- Department of Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Sabina Frascarelli
- Department of Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Riccardo Zucchi
- Department of Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Sandra Ghelardoni
- Department of Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| |
Collapse
|
47
|
Abstract
Trace amine-associated receptors (TAARs) are a family of G protein-coupled receptors (GPCRs) that are evolutionarily conserved in vertebrates. The first discovered TAAR1 is mainly expressed in the brain, and is able to detect low abundant trace amines. TAAR1 is also activated by several synthetic compounds and psychostimulant drugs like amphetamine. Activation of TAAR1 by specific agonists can regulate the classical monoaminergic systems in the brain. Further studies have revealed that other TAAR family members are highly expressed in the olfactory system which are termed olfactory TAARs. In vertebrates, olfactory TAARs can specifically recognize volatile or water-soluble amines. Some of these TAAR agonists are produced by decarboxylation of amino acids. In addition, some TAAR agonists are ethological odors that mediate animal innate behaviors. In this study, we provide a comprehensive review of TAAR agonists, including their structures, biosynthesis pathways, and functions.
Collapse
Affiliation(s)
- Zhengrong Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- Research Institute of Otolaryngology, Nanjing, 210008, China
| | - Qian Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, 201210, China.
| |
Collapse
|
48
|
Bellusci L, Runfola M, Carnicelli V, Sestito S, Fulceri F, Santucci F, Lenzi P, Fornai F, Rapposelli S, Origlia N, Zucchi R, Chiellini G. Endogenous 3-Iodothyronamine (T1AM) and Synthetic Thyronamine-like Analog SG-2 Act as Novel Pleiotropic Neuroprotective Agents Through the Modulation of SIRT6. Molecules 2020; 25:molecules25051054. [PMID: 32110992 PMCID: PMC7179148 DOI: 10.3390/molecules25051054] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/12/2020] [Accepted: 02/25/2020] [Indexed: 12/15/2022] Open
Abstract
3-iodothyronamine (T1AM) and the recently developed analog SG-2 are rapidly emerging as promising multi-target neuroprotective ligands able to reprogram lipid metabolism and to produce memory enhancement in mice. To elucidate the molecular mechanisms underlying the multi-target effects of these novel drug candidates, here we investigated whether the modulation of SIRT6, known to play a key role in reprogramming energy metabolism, might also drive the activation of clearing pathways, such as autophagy and ubiquitine-proteasome (UP), as further mechanisms against neurodegeneration. We show that both T1AM and SG-2 increase autophagy in U87MG cells by inducing the expression of SIRT6, which suppresses Akt activity thus leading to mTOR inhibition. This effect was concomitant with down-regulation of autophagy-related genes, including Hif1α, p53 and mTOR. Remarkably, when mTOR was inhibited a concomitant activation of autophagy and UP took place in U87MG cells. Since both compounds activate autophagy, which is known to sustain long term potentiation (LTP) in the entorhinal cortex (EC) and counteracting AD pathology, further electrophysiological studies were carried out in a transgenic mouse model of AD. We found that SG-2 was able to rescue LTP with an efficacy comparable to T1AM, further underlying its potential as a novel pleiotropic agent for neurodegenerative disorders treatment.
Collapse
Affiliation(s)
- Lorenza Bellusci
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (L.B.); (V.C.); (S.S.); (R.Z.)
| | - Massimiliano Runfola
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.R.); (S.R.)
| | - Vittoria Carnicelli
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (L.B.); (V.C.); (S.S.); (R.Z.)
| | - Simona Sestito
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (L.B.); (V.C.); (S.S.); (R.Z.)
| | - Federica Fulceri
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy;
| | | | - Paola Lenzi
- Unit of Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.L.); (F.F.)
| | - Francesco Fornai
- Unit of Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (P.L.); (F.F.)
- IRCCS Neuromed, 86077 Pozzilli (IS), Italy
| | - Simona Rapposelli
- Laboratory of Medicinal Chemistry, Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.R.); (S.R.)
- Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, 56126 Pisa, Italy
| | - Nicola Origlia
- National Research Council (CNR), Institute of Neuroscience, 56124 Pisa, Italy;
| | - Riccardo Zucchi
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (L.B.); (V.C.); (S.S.); (R.Z.)
| | - Grazia Chiellini
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, 56100 Pisa, Italy; (L.B.); (V.C.); (S.S.); (R.Z.)
- Correspondence: ; Tel.: +39-050-221-86-62
| |
Collapse
|
49
|
Brix K, Szumska J, Weber J, Qatato M, Venugopalan V, Al-Hashimi A, Rehders M. Auto-Regulation of the Thyroid Gland Beyond Classical Pathways. Exp Clin Endocrinol Diabetes 2020; 128:437-445. [PMID: 32074633 DOI: 10.1055/a-1080-2969] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This mini-review asks how self-regulation of the thyroid gland is realized at the cellular and molecular levels by canonical and non-canonical means. Canonical pathways of thyroid regulation comprise thyroid stimulating hormone-triggered receptor signaling. As part of non-canonical regulation, we hypothesized an interplay between protease-mediated thyroglobulin processing and thyroid hormone release into the circulation by means of thyroid hormone transporters like Mct8. We proposed a sensing mechanism by different thyroid hormone transporters, present in specific subcellular locations of thyroid epithelial cells, selectively monitoring individual steps of thyroglobulin processing, and thus, the cellular thyroid hormone status. Indeed, we found that proteases and thyroid hormone transporters are functionally inter-connected, however, in a counter-intuitive manner fostering self-thyrotoxicity in particular in Mct8- and/or Mct10-deficient mice. Furthermore, the possible role of the G protein-coupled receptor Taar1 is discussed, because we detected Taar1 at cilia of the apical plasma membrane of thyrocytes in vitro and in situ. Eventually, through pheno-typing Taar1-deficient mice, we identified a co-regulatory role of Taar1 and the thyroid stimulating hormone receptors. Recently, we showed that inhibition of thyroglobulin-processing enzymes results in disappearance of cilia from the apical pole of thyrocytes, while Taar1 is re-located to the endoplasmic reticulum. This pathway features a connection between thyrotropin-stimulated secretion of proteases into the thyroid follicle lumen and substrate-mediated self-assisted control of initially peri-cellular thyroglobulin processing, before its reinternalization by endocytosis, followed by extensive endo-lysosomal liberation of thyroid hormones, which are then released from thyroid follicles by means of thyroid hormone transporters.
Collapse
Affiliation(s)
- Klaudia Brix
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Joanna Szumska
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany.,Present address of JS is Department of Internal Medicine III, Cardiology, Angiology and Respiratory Medicine, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Jonas Weber
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Maria Qatato
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Vaishnavi Venugopalan
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Alaa Al-Hashimi
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| | - Maren Rehders
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Bremen, Germany
| |
Collapse
|
50
|
Gachkar S, Oelkrug R, Herrmann B, Scanlan TS, Sun Q, Biebermann H, Hoefig CS, Schomburg L, Mittag J. N- and O-Acetylated 3-Iodothyronamines Have No Metabolic or Thermogenic Effects in Male Mice. Eur Thyroid J 2020; 9:57-66. [PMID: 32257954 PMCID: PMC7109410 DOI: 10.1159/000504887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Injection of 3-iodothyronamine into experimental animals profoundly affects their metabolism and body temperature. As 3-iodothyronamine is rapidly acetylated in vivo after injection, it was hypothesized that the metabolites N- or O-acetyl-3-iodothyronamines could constitute the active hormones. METHODS Adult male mice were injected once daily with one of the metabolites (5 mg/kg body weight intraperitoneally dissolved in 60% DMSO in PBS) or solvent. Metabolism was monitored by indirect calorimetry, body temperature by infrared thermography, and body composition by nuclear magnetic resonance analysis. Signaling activities in brown fat or liver were assessed by studying target gene transcription by qPCR including uncoupling protein 1 or deiodinase type 1 or 2, and Western blot. RESULTS The markers of metabolism, body composition, or temperature tested were similar in the mice injected with solvent and those injected with one of the acetylated 3-iodothyronamines. CONCLUSIONS In our experimental setup, N- and O-acetyl-3-iodothyronamine do not constitute compounds contributing to the metabolic or temperature effects described for 3-iodothyronamine. The acetylation of 3-iodothyronamine observed in vivo may thus rather serve degradation and elimination purposes.
Collapse
Affiliation(s)
- Sogol Gachkar
- Molecular Endocrinology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Rebecca Oelkrug
- Molecular Endocrinology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Beate Herrmann
- Molecular Endocrinology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Thomas S. Scanlan
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon, USA
| | - Qian Sun
- Institute for Experimental Endocrinology, Charité − Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Heike Biebermann
- Institute of Experimental Pediatric Endocrinology, Charité − Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carolin S. Hoefig
- Institute for Experimental Endocrinology, Charité − Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité − Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jens Mittag
- Molecular Endocrinology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
- *Prof. Dr. Jens Mittag, Center of Brain, Behavior and Metabolism, Ratzeburger Allee 160, Haus 66, DE–23562 Lübeck (Germany), E-Mail
| |
Collapse
|