251
|
Oishi A, Gbahou F, Jockers R. Melatonin receptors, brain functions, and therapies. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:345-356. [PMID: 34225974 DOI: 10.1016/b978-0-12-819975-6.00022-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In mammals, including humans, the neurohormone melatonin is mainly secreted from the pineal gland at night and acts on two high-affinity G protein-coupled receptors, the melatonin MT1 and MT2 receptors. Major functions of melatonin receptors in the brain are the regulation of circadian rhythms and sleep. Correspondingly, the main indications of the currently available drugs for these receptors indicate this as targets. Yet these drugs may not only improve circadian rhythm- and sleep-related disorders but may also be beneficial for complex diseases like major depression, Alzheimer's disease, autism, and attention-deficit/hyperactivity disorders. Here, we will focus on the hypothalamic functions of melatonin receptors by updating our knowledge on their hypothalamic expression pattern at normal, aged, and disease states, by discussing their capacity to regulate circadian rhythms and sleep and by presenting the clinical applications of the melatonin receptor-targeting drugs ramelteon, tasimelteon, and agomelatine or of prolonged-release melatonin formulations. Finally, we speculate about future trends in the field of melatonin receptor drugs.
Collapse
Affiliation(s)
- Atsuro Oishi
- Institut Cochin, Université de Paris, Paris, France
| | | | - Ralf Jockers
- Institut Cochin, Université de Paris, Paris, France.
| |
Collapse
|
252
|
Sun Y, Ma L, Jin M, Zheng Y, Wang D, Ni H. Effects of Melatonin on Neurobehavior and Cognition in a Cerebral Palsy Model of plppr5-/- Mice. Front Endocrinol (Lausanne) 2021; 12:598788. [PMID: 33692754 PMCID: PMC7937640 DOI: 10.3389/fendo.2021.598788] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Cerebral palsy (CP), a group of clinical syndromes caused by non-progressive brain damage in the developing fetus or infant, is one of the most common causes of lifelong physical disability in children in most countries. At present, many researchers believe that perinatal cerebral hypoxic ischemic injury or inflammatory injury are the main causes of cerebral palsy. Previous studies including our works confirmed that melatonin has a protective effect against convulsive brain damage during development and that it affects the expression of various molecules involved in processes such as metabolism, plasticity and signaling in the brain. Integral membrane protein plppr5 is a new member of the plasticity-related protein family, which is specifically expressed in brain and spinal cord, and induces filopodia formation as well as neurite growth. It is highly expressed in the brain, especially in areas of high plasticity, such as the hippocampus. The signals are slightly lower in the cortex, the cerebellum, and in striatum. Noteworthy, during development plppr5 mRNA is expressed in the spinal cord, i.e., in neuron rich regions such as in medial motor nuclei, suggesting that plppr5 plays an important role in the regulation of neurons. However, the existing literature only states that plppr5 is involved in the occurrence and stability of dendritic spines, and research on its possible involvement in neonatal ischemic hypoxic encephalopathy has not been previously reported. We used plppr5 knockout (plppr5-/-) mice and their wild-type littermates to establish a model of hypoxicischemic brain injury (HI) to further explore the effects of melatonin on brain injury and the role of plppr5 in this treatment in an HI model, which mainly focuses on cognition, exercise, learning, and memory. All the tests were performed at 3-4 weeks after HI. As for melatonin treatment, which was performed 5 min after HI injury and followed by every 24h. In these experiments, we found that there was a significant interaction between genotype and treatment in novel object recognition tests, surface righting reflex tests and forelimb suspension reflex tests, which represent learning and memory, motor function and coordination, and the forelimb grip of the mice, respectively. However, a significant main effect of genotype and treatment on performance in all behavioral tests were observed. Specifically, wild-type mice with HI injury performed better than plppr5-/- mice, regardless of treatment with melatonin or vehicle. Moreover, treatment with melatonin could improve behavior in the tests for wild-type mice with HI injury, but not for plppr5-/- mice. This study showed that plppr5 knockout aggravated HI damage and partially weakened the neuroprotection of melatonin in some aspects (such as novel object recognition test and partial nerve reflexes), which deserves further study.
Collapse
|
253
|
Chen Z, Tao S, Zhu R, Tian S, Sun Y, Wang H, Yan R, Shao J, Zhang Y, Zhang J, Yao Z, Lu Q. Aberrant functional connectivity between the suprachiasmatic nucleus and the superior temporal gyrus: Bridging RORA gene polymorphism with diurnal mood variation in major depressive disorder. J Psychiatr Res 2021; 132:123-130. [PMID: 33091686 DOI: 10.1016/j.jpsychires.2020.09.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/24/2020] [Accepted: 09/30/2020] [Indexed: 10/23/2022]
Abstract
Diurnal mood variation (DMV), a common symptom of major depressive disorder (MDD), is associated with circadian related genes and dysregulation of the suprachiasmatic nucleus (SCN). Previous research confirmed that the RORA gene is involved in the regulation of circadian rhythms. In this study, we hypothesized that polymorphisms of RORA may affect DMV symptoms of MDD through functional changes in the SCN. A total of 208 patients diagnosed with depression and 120 control subjects were enrolled and underwent a resting-state functional magnetic resonance imaging (rs-fMRI). Blood samples were collected and genotyping of 9 RORA gene SNPs were performed using next-generation sequencing technology. Patients were categorized as an AA genotype or C allele carriers based on RORA rs72752802 polymorphism. SCN-seed functional connectivity (FC) was compared between the two groups and correlation with severity of DMV was analyzed. Finally, a mediation analysis was performed to further determine FC intermediary effects. We observed that rs72752802 was significantly associated with patients' DMV symptoms. C allele carriers of rs72752802 showed significantly decreased FC between the right SCN and right superior temporal gyrus (rSTG). This was also correlated with DMV symptoms. In addition, the rs72752802 SNP influenced DMV symptoms through intermediary effects of SCN-rSTG connectivity. The study presented here provides a neurological and genetic basis for understanding depressed patients experiencing DMV.
Collapse
Affiliation(s)
- Zhilu Chen
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shiwan Tao
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Rongxin Zhu
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shui Tian
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, 210096, China
| | - Yurong Sun
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, 210096, China
| | - Huan Wang
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, 210096, China
| | - Rui Yan
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China; Nanjing Brain Hospital, Medical School of Nanjing University, Nanjing, 210093, China
| | - Junneng Shao
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, 210096, China
| | - Yujie Zhang
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, 210096, China
| | - Jie Zhang
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhijian Yao
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China; School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Nanjing Brain Hospital, Medical School of Nanjing University, Nanjing, 210093, China.
| | - Qing Lu
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, 210096, China.
| |
Collapse
|
254
|
Yoshimoto A, Yamashiro K, Suzuki T, Ikegaya Y, Matsumoto N. Ramelteon modulates gamma oscillations in the rat primary motor cortex during non-REM sleep. J Pharmacol Sci 2021; 145:97-104. [PMID: 33357785 DOI: 10.1016/j.jphs.2020.11.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
Sleep disorders adversely affect daily activities and cause physiological and psychiatric problems. The shortcomings of benzodiazepine hypnotics have led to the development of ramelteon, a melatonin MT1 and MT2 agonist. Although the sleep-promoting effects of ramelteon have been documented, few studies have precisely investigated the structure of sleep and neural oscillatory activities. In this study, we recorded electrocorticograms in the primary motor cortex, the primary somatosensory cortex and the olfactory bulb as well as electromyograms in unrestrained rats treated with either ramelteon or vehicle. A neural-oscillation-based algorithm was used to classify the behavior of the rats into three vigilance states (e.g., awake, rapid eye movement (REM) sleep, and non-REM (NREM) sleep). Moreover, we investigated the region-, frequency- and state-specific modulation of extracellular oscillations in the ramelteon-treated rats. We demonstrated that in contrast to benzodiazepine treatment, ramelteon treatment promoted NREM sleep and enhanced fast gamma power in the primary motor cortex during NREM sleep, while REM sleep was unaffected. Gamma oscillations locally coordinate neuronal firing, and thus, ramelteon modulates neural oscillations in sleep states in a unique manner and may contribute to off-line information processing during sleep.
Collapse
Affiliation(s)
- Airi Yoshimoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Kotaro Yamashiro
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Takeshi Suzuki
- Graduate School of Pharmaceutical Sciences, Keio University, Tokyo 105-8512, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka 565-0871, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
255
|
Yoshimoto A, Yamashiro K, Ikegaya Y, Matsumoto N. Acute Ramelteon Treatment Maintains the Cardiac Rhythms of Rats during Non-REM Sleep. Biol Pharm Bull 2021; 44:789-797. [PMID: 34078810 DOI: 10.1248/bpb.b20-00932] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sleep curtailment negatively affects cardiac activities and thus should be ameliorated by pharmacological methods. One of the therapeutic targets is melatonin receptors, which tune circadian rhythms. Ramelteon, a melatonin MT1/MT2 receptor agonist, has recently been developed to modulate sleep-wake rhythms. To date, the sleep-promoting effect of ramelteon has been widely delineated, but whether ramelteon treatment physiologically influences cardiac function is not well understood. To address this question, we recorded electrocardiograms, electromyograms, and electrocorticograms in the frontal cortex and the olfactory bulb of unrestrained rats treated with either ramelteon or vehicle. We detected vigilance states based on physiological measurements and analyzed cardiac and muscular activities. We found that during non-rapid eye movement (non-REM) sleep, heartrate variability was maintained by ramelteon treatment. Analysis of the electromyograms confirmed that neither microarousal during non-REM sleep nor the occupancy of phasic periods during REM sleep was altered by ramelteon. Our results indicate that ramelteon has a remedial effect on cardiac activity by keeping the heartrate variability and may reduce cardiac dysfunction during sleep.
Collapse
Affiliation(s)
- Airi Yoshimoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Kotaro Yamashiro
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
- Institute for AI and Beyond, The University of Tokyo
- Center for Information and Neural Networks, National Institute of Information and Communications Technology
| | | |
Collapse
|
256
|
Barbosa-Méndez S, Pérez-Sánchez G, Becerril-Villanueva E, Salazar-Juárez A. Melatonin decreases cocaine-induced locomotor sensitization and cocaine-conditioned place preference in rats. J Psychiatr Res 2021; 132:97-110. [PMID: 33080430 DOI: 10.1016/j.jpsychires.2020.09.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/11/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
Melatonin is a hormone that produces behavioral, pharmacological, and physiological effects through the activation of MT1 and MT2 melatonin receptors. Melatonin receptors participate in the modulation of the reinforcing effects of cocaine. Some studies report that dosing of melatonin decreases cocaine-induced locomotor activity and cocaine self-administration and that luzindole, an MT1, and MT2 melatonin receptor antagonist, blocks the melatonin-dependent decrease in cocaine-induced locomotor activity. The objective of this study was to evaluate the effect of acute or chronic dosing of melatonin on the induction and expression of cocaine-induced locomotor sensitization and cocaine-CPP in rats. Male Wistar rats received cocaine during the induction and expression of locomotor sensitization. Melatonin was administered 30 min before cocaine. After each treatment, locomotor activity was recorded for 30 min. Additionally, dopamine levels were determined in the ventral striatum, the prefrontal cortex (PFc), and the ventral tegmental area (VTA) by HPLC in animals treated with melatonin and cocaine. Melatonin decreased cocaine-induced locomotor sensitization and intracellular dopamine levels, as well as cocaine-CPP. Luzindole blocked the melatonin-induced decrease in the expression of locomotor sensitization in rats. These data suggest that melatonin may be a useful therapeutic agent to reduce cocaine abuse; additionally, they suggest that MT1 and MT2 receptors could be therapeutic targets, useful for the treatment of drug abuse disorder.
Collapse
Affiliation(s)
- Susana Barbosa-Méndez
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Ciudad de México, 14370, Mexico
| | - Gilberto Pérez-Sánchez
- Dirección de Neurociencias, Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría, Ciudad de México, 14370, Mexico
| | - Enrique Becerril-Villanueva
- Dirección de Neurociencias, Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría, Ciudad de México, 14370, Mexico
| | - Alberto Salazar-Juárez
- Subdirección de Investigaciones Clínicas, Laboratorio de Neurofarmacología Conductual, Microcirugía y Terapéutica Experimental, Ciudad de México, 14370, Mexico.
| |
Collapse
|
257
|
Cheng F, Chang H, Yan F, Yang A, Liu J, Liu Y. Agomelatine Attenuates Isoflurane-Induced Inflammation and Damage in Brain Endothelial Cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:5589-5598. [PMID: 33376303 PMCID: PMC7755371 DOI: 10.2147/dddt.s281582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/05/2020] [Indexed: 12/25/2022]
Abstract
Background and Purpose Neurotoxicity of anesthetics has been widely observed by clinicians. It is reported that inflammation and oxidative stress are involved in the pathological process. In the present study, we aimed to assess the therapeutic effects of agomelatine against isoflurane-induced inflammation and damage to brain endothelial cells. Materials and Methods MTT assay was used to detect cell viability in order to determine the optimized concentration of agomelatine. The bEnd.3 brain endothelial cells were treated with 2% isoflurane in the presence or absence of agomelatine (5, 10 μM) for 24 h. LDH release was evaluated and the ROS levels were checked using DHE staining assay. The expressions of IL-6, IL-8, TNF-α, VEGF, TF, VCAM-1, and ICAM-1 were evaluated using real-time PCR and ELISA. Real-time PCR and Western blot analysis were used to determine the expression level of Egr-1. Results The decreased cell viability promoted LDH release and elevated ROS levels induced by isoflurane were significantly reversed by the introduction of agomelatine in a dose-dependent manner. The expression levels of IL-6, IL-8, TNF-α, VEGF, TF, VCAM-1, and ICAM-1 were elevated by stimulation with isoflurane, which were significantly suppressed by the administration of agomelatine. The up-regulation of transcriptional factor Egr-1 induced by isoflurane was down-regulated by agomelatine. Conclusion Agomelatine might attenuate isoflurane-induced inflammation and damage via down-regulating Egr-1 in brain endothelial cells.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Anesthesiology and Pain Clinic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University
| | - Huanxian Chang
- Department of Neurology, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University
| | - Fengfeng Yan
- Department of Anesthesiology and Pain Clinic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University
| | - Aixing Yang
- Department of Anesthesiology and Pain Clinic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University
| | - Jing Liu
- Department of Anesthesiology and Pain Clinic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University
| | - Yuliang Liu
- Department of Neurosurgery, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| |
Collapse
|
258
|
Yin CD, Hou YL, Liu XR, He YS, Wang XP, Li CJ, Tan XH, Liu J. Development of an immune-related prognostic index associated with osteosarcoma. Bioengineered 2020; 12:172-182. [PMID: 33371790 PMCID: PMC8806312 DOI: 10.1080/21655979.2020.1864096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Tumor immunity is closely associated with the prognosis of tumors, including osteosarcoma (OS). The aim of the present study was to construct an immune-related prognostic index (PI) to predict the prognosis of OS. Herein, OS expression data were sourced from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database. We divided the OS patients into nonmetastatic and metastatic groups, allowing differentially immune-related genes (DIRGs) to be selected. After DIRGs were further investigated by enrichment analysis, four keys prognostic IRGs (CD79A, CSF3R, MTNR1B and NPPC) were identified using a Cox proportional hazards model. Then, an immune-related prognostic index was constructed. Finally, gene set enrichment analysis (GSEA) was employed to further explore the underlying mechanisms. The difference in tumor-infiltrating immune cell (TIIC) abundance was also discussed. In our study, eight upregulated genes and 30 downregulated genes were identified. Several Gene Ontology (GO) terms and the most significantly enriched KEGG pathways were immune-associated functions and pathways. Four genes, including CD79A, CSF3R, MTNR1B and NPPC, were used to establish a risk assessment model for evaluating OS prognosis. GSEA revealed that the risk score was related to cytokine receptor interaction and to the chemokine and B cell receptor signaling pathways. Furthermore, high risk markedly related to the infiltration of several immune cell types, including M2 macrophages, naïve CD4 T cells, and CD8 T cells. In sum, we developed a survival model for OS. The underlying molecular mechanisms of the high-risk group may affect immune-related biological processes and TIICs.Abbreviations TARGET: Therapeutically Applicable Research To Generate Effective Treatments; PI: Prognostic index; OS: Osteosarcoma; DIRGs: Differentially immune-related genes; GSEA: Gene set enrichment analysis; TIIC: Tumor-infiltrating immune cell.
Collapse
Affiliation(s)
- Chao-Dong Yin
- Department of Hand and Foot Surgery and Microsurgery, Affiliated to the First People's Hospital of Chenzhou , P.R. China
| | - Ying-Lan Hou
- Health Management Centre, Affiliated to the First People's Hospital of Chenzhou , P.R. China
| | - Xiao-Ren Liu
- Department of Hand and Foot Surgery and Microsurgery, Affiliated to the First People's Hospital of Chenzhou , P.R. China
| | - Yu-Sheng He
- Department of Hand and Foot Surgery and Microsurgery, Affiliated to the First People's Hospital of Chenzhou , P.R. China
| | - Xin-Ping Wang
- Department of Hand and Foot Surgery and Microsurgery, Affiliated to the First People's Hospital of Chenzhou , P.R. China
| | - Cheng-Jie Li
- Department of Hand and Foot Surgery and Microsurgery, Affiliated to the First People's Hospital of Chenzhou , P.R. China
| | - Xiao-Hong Tan
- Department of Hand and Foot Surgery and Microsurgery, Affiliated to the First People's Hospital of Chenzhou , P.R. China
| | - Jun Liu
- Department of Hand and Foot Surgery and Microsurgery, Affiliated to the First People's Hospital of Chenzhou , P.R. China
| |
Collapse
|
259
|
Pourhanifeh MH, Mehrzadi S, Hosseinzadeh A. Melatonin and regulation of miRNAs: novel targeted therapy for cancerous and noncancerous disease. Epigenomics 2020; 13:65-81. [PMID: 33350862 DOI: 10.2217/epi-2020-0241] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
miRNAs, small noncoding RNAs with crucial diagnostic and prognostic capabilities, play essential therapeutic roles in different human diseases. These biomarkers are involved in several biological mechanisms and are responsible for the regulation of multiple genes expressions in cells. miRNA-based therapy has shown a very bright future in the case of clinical interventions. Melatonin, the main product of the pineal gland, is a multifunctional neurohormone with numerous therapeutic potentials in human diseases. Melatonin is able to regulate miRNAs in different pathologies such as malignant and nonmalignant diseases, which can be considered as a novel kind of targeted therapy. Herein, this review discusses possible therapeutic utility of melatonin for the regulation of miRNAs in various pathological conditions.
Collapse
Affiliation(s)
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
260
|
Farré-Alins V, Narros-Fernández P, Palomino-Antolín A, Decouty-Pérez C, Lopez-Rodriguez AB, Parada E, Muñoz-Montero A, Gómez-Rangel V, López-Muñoz F, Ramos E, González-Rodríguez Á, Gandía L, Romero A, Egea J. Melatonin Reduces NLRP3 Inflammasome Activation by Increasing α7 nAChR-Mediated Autophagic Flux. Antioxidants (Basel) 2020; 9:antiox9121299. [PMID: 33353046 PMCID: PMC7767051 DOI: 10.3390/antiox9121299] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 12/16/2022] Open
Abstract
Microglia controls the immune system response in the brain. Specifically, the activation and dysregulation of the NLRP3 inflammasome is responsible for the initiation of the inflammatory process through IL-1β and IL-18 release. In this work, we have focused on studying the effect of melatonin on the regulation of the NLRP3 inflammasome through α7 nicotinic receptor (nAChR) and its relationship with autophagy. For this purpose, we have used pharmacological and genetic approaches in lipopolysaccharide (LPS)-induced inflammation models in both in vitro and in vivo models. In the BV2 cell line, LPS inhibited autophagy, which increased NLRP3 protein levels. However, melatonin promoted an increase in the autophagic flux. Treatment of glial cultures from wild-type (WT) mice with LPS followed by extracellular adenosine triphosphate (ATP) produced the release of IL-1β, which was reversed by melatonin pretreatment. In cultures from α7 nAChR knock-out (KO) mice, melatonin did not reduce IL-1β release. Furthermore, melatonin decreased the expression of inflammasome components and reactive oxygen species (ROS) induced by LPS; co-incubation of melatonin with α-bungarotoxin (α-bgt) or luzindole abolished the anti-inflammatory and antioxidant effects. In vivo, melatonin reverted LPS-induced cognitive decline, reduced NLRP3 levels and promoted autophagic flux in the hippocampi of WT mice, whereas in α7 nAChR KO mice melatonin effect was not observed. These results suggest that melatonin may modulate the complex interplay between α7 nAChR and autophagy signaling.
Collapse
Affiliation(s)
- Víctor Farré-Alins
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Paloma Narros-Fernández
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Alejandra Palomino-Antolín
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Céline Decouty-Pérez
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Ana Belen Lopez-Rodriguez
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Esther Parada
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Alicia Muñoz-Montero
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Vanessa Gómez-Rangel
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Francisco López-Muñoz
- Faculty of Health Sciences, University Camilo José Cela, Villanueva de la Cañada, 28692 Madrid, Spain;
- Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute (i+12), 28041 Madrid, Spain
- Portucalense Institute of Neuropsychology and Cognitive and Behavioural Neurosciences (INPP), Portucalense University, 4200-072 Porto, Portugal
- Thematic Network for Cooperative Health Research (RETICS), Addictive Disorders Network, Health Institute Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| | - Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (E.R.); (A.R.)
| | - Águeda González-Rodríguez
- Research Unit, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD, ISCIII), 28029 Madrid, Spain
| | - Luis Gandía
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (E.R.); (A.R.)
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, 28006 Madrid, Spain; (V.F.-A.); (P.N.-F.); (A.P.-A.); (C.D.-P.); (A.B.L.-R.); (E.P.); (V.G.-R.)
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, 28029 Madrid, Spain; (A.M.-M.); (L.G.)
- Correspondence: ; Tel.: +34-915574402
| |
Collapse
|
261
|
A Topical Formulation of Melatoninergic Compounds Exerts Strong Hypotensive and Neuroprotective Effects in a Rat Model of Hypertensive Glaucoma. Int J Mol Sci 2020; 21:ijms21239267. [PMID: 33291737 PMCID: PMC7730513 DOI: 10.3390/ijms21239267] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/23/2020] [Accepted: 12/02/2020] [Indexed: 02/08/2023] Open
Abstract
Melatonin is of great importance for regulating several eye processes, including pressure homeostasis. Melatonin in combination with agomelatine has been recently reported to reduce intraocular pressure (IOP) with higher efficacy than each compound alone. Here, we used the methylcellulose (MCE) rat model of hypertensive glaucoma, an optic neuropathy characterized by the apoptotic death of retinal ganglion cells (RGCs), to evaluate the hypotensive and neuroprotective efficacy of an eye drop nanomicellar formulation containing melatonin/agomelatine. Eye tissue distribution of melatonin/agomelatine in healthy rats was evaluated by HPLC/MS/MS. In the MCE model, we assessed by tonometry the hypotensive efficacy of melatonin/agomelatine. Neuroprotection was revealed by electroretinography; by levels of inflammatory and apoptotic markers; and by RGC density. The effects of melatonin/agomelatine were compared with those of timolol (a beta blocker with prevalent hypotensive activity) or brimonidine (an alpha 2 adrenergic agonist with potential neuroprotective efficacy), two drugs commonly used to treat glaucoma. Both melatonin and agomelatine penetrate the posterior segment of the eye. In the MCE model, IOP elevation was drastically reduced by melatonin/agomelatine with higher efficacy than that of timolol or brimonidine. Concomitantly, gliosis-related inflammation and the Bax-associated apoptosis were partially prevented, thus leading to RGC survival and recovered retinal dysfunction. We suggest that topical melatoninergic compounds might be beneficial for ocular health.
Collapse
|
262
|
Veschsanit N, Yang JL, Ngampramuan S, Viwatpinyo K, Pinyomahakul J, Lwin T, Chancharoen P, Rungruang S, Govitrapong P, Mukda S. Melatonin reverts methamphetamine-induced learning and memory impairments and hippocampal alterations in mice. Life Sci 2020; 265:118844. [PMID: 33278389 DOI: 10.1016/j.lfs.2020.118844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023]
Abstract
AIMS Methamphetamine (METH) has become a major public health problem because of its abuse and profound neurotoxic effects, causing alterations in brain structure and function, and impairing cognitive functions, including attention, decision making, emotional memory, and working memory. This study aimed to determine whether melatonin (MEL), the circadian-control hormone, which has roles beyond circadian rhythm regulation, could restore METH-induced cognitive and neuronal impairment. MAIN METHODS Mice were treated with either METH (1 mg/kg) or saline for 7 days, followed by MEL (10 mg/kg) or saline for another 14 days. The Morris water maze (MWM) test was performed one day after the last saline or MEL injection. The hippocampal neuronal density, synaptic density, and receptors involved in learning and memory, along with downstream signaling molecules (NMDA receptor subunits GluN2A, GluN2B, and CaMKII) were investigated by immunoblotting. KEY FINDINGS METH administration significantly extended escape latency in learning phase and reduced the number of target crossings in memory test-phase as well as decreased the expression of BDNF, NMDA receptors, TrkB receptors, CaMKII, βIII tubulin, and synaptophysin. MEL treatment significantly ameliorated METH-induced increased escape latency, decreased the number of target crossings and decreased expression of BDNF, NMDA receptors, TrkB receptors, CaMKII, βIII tubulin and synaptophysin. SIGNIFICANCE METH administration impairs learning and memory in mice, and MEL administration restores METH-induced neuronal impairments which is probably through the changes in BDNF, NMDA receptors, TrkB receptors, CaMKII, βIII tubulin and synaptophysin. Therefore, MEL is potentially an innovative and promising treatment for learning and memory impairment of humans.
Collapse
Affiliation(s)
- Nisarath Veschsanit
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Jenq-Lin Yang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Sukonthar Ngampramuan
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Kittikun Viwatpinyo
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Jitrapa Pinyomahakul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Thit Lwin
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand; Department of Anatomy, Defence Services Medical Academy, Mingalardon, Yangon 11021, Myanmar
| | - Pongrung Chancharoen
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand; Faculty of Allied Health Sciences, Burapha University, Seansuk, Chonburi 20131, Thailand
| | - Saowalak Rungruang
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Piyarat Govitrapong
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand; Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Sujira Mukda
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand.
| |
Collapse
|
263
|
Sleep Deprivation and Neurological Disorders. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5764017. [PMID: 33381558 PMCID: PMC7755475 DOI: 10.1155/2020/5764017] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022]
Abstract
Sleep plays an important role in maintaining neuronal circuitry, signalling and helps maintain overall health and wellbeing. Sleep deprivation (SD) disturbs the circadian physiology and exerts a negative impact on brain and behavioural functions. SD impairs the cellular clearance of misfolded neurotoxin proteins like α-synuclein, amyloid-β, and tau which are involved in major neurodegenerative diseases like Alzheimer's disease and Parkinson's disease. In addition, SD is also shown to affect the glymphatic system, a glial-dependent metabolic waste clearance pathway, causing accumulation of misfolded faulty proteins in synaptic compartments resulting in cognitive decline. Also, SD affects the immunological and redox system resulting in neuroinflammation and oxidative stress. Hence, it is important to understand the molecular and biochemical alterations that are the causative factors leading to these pathophysiological effects on the neuronal system. This review is an attempt in this direction. It provides up-to-date information on the alterations in the key processes, pathways, and proteins that are negatively affected by SD and become reasons for neurological disorders over a prolonged period of time, if left unattended.
Collapse
|
264
|
Insights into Potential Targets for Therapeutic Intervention in Epilepsy. Int J Mol Sci 2020; 21:ijms21228573. [PMID: 33202963 PMCID: PMC7697405 DOI: 10.3390/ijms21228573] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a chronic brain disease that affects approximately 65 million people worldwide. However, despite the continuous development of antiepileptic drugs, over 30% patients with epilepsy progress to drug-resistant epilepsy. For this reason, it is a high priority objective in preclinical research to find novel therapeutic targets and to develop effective drugs that prevent or reverse the molecular mechanisms underlying epilepsy progression. Among these potential therapeutic targets, we highlight currently available information involving signaling pathways (Wnt/β-catenin, Mammalian Target of Rapamycin (mTOR) signaling and zinc signaling), enzymes (carbonic anhydrase), proteins (erythropoietin, copine 6 and complement system), channels (Transient Receptor Potential Vanilloid Type 1 (TRPV1) channel) and receptors (galanin and melatonin receptors). All of them have demonstrated a certain degree of efficacy not only in controlling seizures but also in displaying neuroprotective activity and in modifying the progression of epilepsy. Although some research with these specific targets has been done in relation with epilepsy, they have not been fully explored as potential therapeutic targets that could help address the unsolved issue of drug-resistant epilepsy and develop new antiseizure therapies for the treatment of epilepsy.
Collapse
|
265
|
Melatonin regulates Aβ production/clearance balance and Aβ neurotoxicity: A potential therapeutic molecule for Alzheimer's disease. Biomed Pharmacother 2020; 132:110887. [PMID: 33254429 DOI: 10.1016/j.biopha.2020.110887] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease with multiple predisposing factors and complicated pathogenesis. Aβ peptide is one of the most important pathogenic factors in the etiology of AD. Accumulating evidence indicates that the imbalance of Aβ production and Aβ clearance in the brain of AD patients leads to Aβ deposition and neurotoxic Aβ oligomer formation. Melatonin shows a potent neuroprotective effect and can prevent or slow down the progression of AD, supporting the view that melatonin is a potential therapeutic molecule for AD. Melatonin modulates the regulatory network of secretase expression and affects the function of secretase, thereby inhibiting amyloidogenic APP processing and Aβ production. Additionally, melatonin ameliorates Aβ-induced neurotoxicity and probably promotes Aβ clearance through glymphatic-lymphatic drainage, BBB transportation and degradation pathways. In this review, we summarize and discuss the role of melatonin against Aβ-dependent AD pathogenesis. We explore the potential cellular and molecular mechanisms of melatonin on Aβ production and assembly, Aβ clearance, Aβ neurotoxicity and circadian cycle disruption. We summarize multiple clinical trials of melatonin treatment in AD patients, showing that melatonin has a promising effect on improving sleep quality and cognitive function. This review aims to stimulate further research on melatonin as a potential therapeutic agent for AD.
Collapse
|
266
|
Liang JH, Wang C, Huo XK, Tian XG, Zhao WY, Wang X, Sun CP, Ma XC. The genus Uncaria: A review on phytochemical metabolites and biological aspects. Fitoterapia 2020; 147:104772. [PMID: 33152463 DOI: 10.1016/j.fitote.2020.104772] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/03/2020] [Accepted: 10/25/2020] [Indexed: 12/14/2022]
Abstract
The genus Uncaira (Rubiaceae) comprises of 34 species, many of which are usually used as traditional Chinese medicines (TCMs) to treat hypertension, fever, headache, gastrointestinal illness, and fungal infection. Over the past twenty years, Uncaira species have been paid the considerable attentions in phytochemical and biological aspects, and about 100 new secondary metabolites, including alkaloids, triterpenes, and flavonoids, have been elucidated. This review aims to present a comprehensive and up-to date overview of the biological source, structures and their biosynthetic pathways, as well as the pharmacological of the compounds reported in the genus Uncaria for the past two decades. It would provide an insight into the emerging pharmacological applications of the genus Uncaria.
Collapse
Affiliation(s)
- Jia-Hao Liang
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Chao Wang
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Xiao-Kui Huo
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Xiang-Ge Tian
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Wen-Yu Zhao
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China
| | - Xun Wang
- Department of Neurosurgery, The Third People's Hospital of Dalian, Non-Directly Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Cheng-Peng Sun
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China.
| | - Xiao-Chi Ma
- College of Pharmacy, College of Integrative Medicine, Dalian Medical University, Dalian, China; Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, Dalian Medical University, Dalian, China.
| |
Collapse
|
267
|
Abstract
The scope of this article is to review the effects on sleep of prescription drugs that are commonly prescribed for chronic insomnia in adults. The following groups are discussed: benzodiazepines and its receptor agonists, the dual orexin receptor antagonist suvorexant, melatonin and its receptor agonists, sedating antidepressants, and antipsychotics. Together with the neurobiologic and pharmacologic properties of these drugs, clinical effects are described, including subjective and objective effects on sleep duration, continuity, and architecture. Medical prescription information is given when available. Recently published American and European guidelines for the treatment of insomnia serve as reference frame.
Collapse
Affiliation(s)
- Sylvie Dujardin
- Sleep Medicine Center Kempenhaeghe, PO Box 61, Heeze 5590 AB, The Netherlands
| | - Angelique Pijpers
- Sleep Medicine Center Kempenhaeghe, PO Box 61, Heeze 5590 AB, The Netherlands
| | - Dirk Pevernagie
- Sleep Medicine Center Kempenhaeghe, PO Box 61, Heeze 5590 AB, The Netherlands; Department of Internal Medicine and Paediatrics, Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium.
| |
Collapse
|
268
|
Bhatwadekar AD, Rameswara V. Circadian rhythms in diabetic retinopathy: an overview of pathogenesis and investigational drugs. Expert Opin Investig Drugs 2020; 29:1431-1442. [PMID: 33107770 DOI: 10.1080/13543784.2020.1842872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Circadian rhythm is a natural endogenous process occurring roughly every 24 hours. Circadian rhythm dysfunction is involved in diabetic retinopathy (DR) pathogenesis. Interestingly, there are investigational drugs that exhibit potential in the treatment of DR by targeting circadian rhythm dysfunction. AREAS COVERED We performed a literature search in June 2020 using PubMed's Medical Subject Heading (MeSH) terms 'circadian clock,' 'circadian rhythms,' and 'diabetic retinopathy.' This article offers an overview of the physiology of the biological clock and clock regulatory genes and presents an examination of the retinal clock. It discusses the pathogenic mechanisms of DR and emphasizes how circadian rhythm dysfunction at structural, physiological, metabolic and cellular levels, plays a critical role in the development of DR. The latter part of the paper sheds light on those investigational drugs (such as melatonin, tasimelteon and metformin) which exhibit potential in the treatment of DR by the targeting of circadian rhythm dysfunction. EXPERT OPINION An enhanced understanding of circadian rhythm and its role in DR could offer therapeutic potential by targeting of circadian rhythm dysfunction.
Collapse
Affiliation(s)
- Ashay D Bhatwadekar
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute , Indianapolis, IN, USA
| | - Varun Rameswara
- Indiana University School of Medicine. Indiana University , Indianapolis, IN, USA
| |
Collapse
|
269
|
Genario R, Cipolla-Neto J, Bueno AA, Santos HO. Melatonin supplementation in the management of obesity and obesity-associated disorders: A review of physiological mechanisms and clinical applications. Pharmacol Res 2020; 163:105254. [PMID: 33080320 DOI: 10.1016/j.phrs.2020.105254] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/11/2020] [Accepted: 10/11/2020] [Indexed: 02/08/2023]
Abstract
Despite the evolving advances in clinical approaches to obesity and its inherent comorbidities, the therapeutic challenge persists. Among several pharmacological tools already investigated, recent studies suggest that melatonin supplementation could be an efficient therapeutic approach in the context of obesity. In the present review, we have amalgamated the evidence so far available on physiological effects of melatonin supplementation in obesity therapies, addressing its effects upon neuroendocrine systems, cardiometabolic biomarkers and body composition. Most studies herein appraised employed melatonin supplementation at dosages ranging from 1 to 20 mg/day, and most studies followed up participants for periods from 3 weeks to 12 months. Overall, it was observed that melatonin plays an important role in glycaemic homeostasis, in addition to modulation of white adipose tissue activity and lipid metabolism, and mitochondrial activity. Additionally, melatonin increases brown adipose tissue volume and activity, and its antioxidant and anti-inflammatory properties have also been demonstrated. There appears to be a role for melatonin in adiposity reduction; however, several questions remain unanswered, for example melatonin baseline levels in obesity, and whether any seeming hypomelatonaemia or melatonin irresponsiveness could be clarifying factors. Supplementation dosage studies and more thorough clinical trials are needed to ascertain not only the relevance of such findings but also the efficacy of melatonin supplementation.
Collapse
Affiliation(s)
- Rafael Genario
- School of Medicine, University of Sao Paulo (USP), São Paulo, Brazil.
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Worcester, United Kingdom
| | - Heitor O Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil.
| |
Collapse
|
270
|
Niu G, Yousefi B, Qujeq D, Marjani A, Asadi J, Wang Z, Mir SM. Melatonin and doxorubicin co-delivered via a functionalized graphene-dendrimeric system enhances apoptosis of osteosarcoma cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111554. [PMID: 33321618 DOI: 10.1016/j.msec.2020.111554] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 12/18/2022]
Abstract
A functionalized graphene-dendrimeric system was designed via Fe3O4 nanoparticle (NP) as a magnetic nanocarrier for co-delivery of doxorubicin (DOX) and melatonin (MLT). Accordingly, β-Cyclodextrin (β-CD) was modified by creating amine functional groups. The modified β-CD was grafted with Graphene oxide (GO), and the resulting platform gain many functional groups, including the hydroxyl (-OH), carboxylic acid (-COOH), and amine functional groups (-NH2). Finally, magnetic NPs were synthesized on the prepared platform to efficiently controlling and targeting drugs to tumor sites. The human osteosarcoma cell lines including Saos-2 and MG-63, as well as Human Bone Marrow Mesenchymal Stem Cells (hBM-MSC) line, were used to determine the in vitro biological effects of the functionalized graphene-dendrimeric system. The magnetic nanocarrier has encapsulation efficiency (EE) values of 99.92% for DOX and 21.5% for MLT. The biocompatibility tests of the nanocarrier revealed that the magnetic nanocarrier was appropriate as a drug carrier. Co-delivery of DOX and MLT with an efficiently anticancer performance was also was confirmed by cellular uptake, 4',6-diamidino-2-phenylindole (DAPI) staining, and apoptosis analysis in comparison with free DOX and MLT. Moreover, there was a synergy in the antitumor effect when MLT was combined with DOX, especially in the nano-formulation form, which may be due to the down-regulation of X-linked Inhibitor of Apoptosis (XIAP), survivin, and human telomerase catalytic subunit (hTERT) (p < 0.0001). Overall, the result of our study suggests that the designed carrier is a promising nanocarrier for targeted co-delivery of DOX and MLT with improved anticancer efficacy in cancer cells and thus reduced toxicity in normal cells.
Collapse
Affiliation(s)
- Guangfeng Niu
- Department of Orthopaedics, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324 Jingwu Weiqi Road, Jinan, Shandong 250021, China; Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Weiqi Road, Jinan, Shandong 250021, China
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdoljalal Marjani
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine Golestan University of Medical Sciences, Gorgan, Iran
| | - Jahanbakhsh Asadi
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine Golestan University of Medical Sciences, Gorgan, Iran
| | - Zhan Wang
- Department of Orthopaedics, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China.
| | - Seyed Mostafa Mir
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
271
|
Singhanat K, Apaijai N, Jaiwongkam T, Kerdphoo S, Chattipakorn SC, Chattipakorn N. Melatonin as a therapy in cardiac ischemia-reperfusion injury: Potential mechanisms by which MT2 activation mediates cardioprotection. J Adv Res 2020; 29:33-44. [PMID: 33842003 PMCID: PMC8020169 DOI: 10.1016/j.jare.2020.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/13/2020] [Accepted: 09/22/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction Previous studies reported the beneficial effects of pretreatment with melatonin on the heart during cardiac ischemia/reperfusion (I/R) injury. However, the effects of melatonin given after cardiac ischemia, as well as its comparative temporal effects are unknown. These include pretreatment, during ischemia, and at the onset of reperfusion. Also, the association between melatonin receptors and cardiac arrhythmias, mitochondrial function and dynamics, autophagy, and mitophagy during cardiac I/R have not been investigated. Objectives We tested two major hypotheses in this study. Firstly, the temporal effect of melatonin administration exerts different cardioprotective efficacy during cardiac I/R. Secondly, melatonin provides cardioprotective effects via MT2 activation, leading to improvement in cardiac mitochondrial function and dynamics, reduced excessive mitophagy and autophagy, and decreased cardiac arrhythmias, resulting in improved LV function. Methods Male rats were subjected to cardiac I/R, and divided into 4 intervention groups: vehicle, pretreatment with melatonin, melatonin given during ischemia, and melatonin given at the onset of reperfusion. In addition, either a non-specific melatonin receptor (MT) blocker or specific MT2 blocker was given to rats. Results Treatment with melatonin at all time points alleviated cardiac I/R injury to a similar extent, quantified by reduction in infarct size, arrhythmia score, LV dysfunction, cardiac mitochondrial dysfunction, imbalance of mitochondrial dynamics, excessive mitophagy, and a decreased Bax/Bcl2 ratio. In H9C2 cells, melatonin increased %cell viability by reducing mitochondrial dynamic imbalance and a decrease in Bax protein expression. The cardioprotective effects of melatonin were dependent on MT2 activation. Conclusion Melatonin given before or after ischemia exerted equal levels of cardioprotection on the heart with I/R injury, and its beneficial effects on cardiac arrhythmias, cardiac mitochondrial function and dynamics were dependent upon the activation of MT2.
Collapse
Affiliation(s)
- Kodchanan Singhanat
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thidarat Jaiwongkam
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
272
|
Abstract
Melatonin (Mel) promotes sleep through G protein-coupled receptors. However, the downstream molecular target(s) is unknown. We identified the Caenorhabditis elegans BK channel SLO-1 as a molecular target of the Mel receptor PCDR-1-. Knockout of pcdr-1, slo-1, or homt-1 (a gene required for Mel synthesis) causes substantially increased neurotransmitter release and shortened sleep duration, and these effects are nonadditive in double knockouts. Exogenous Mel inhibits neurotransmitter release and promotes sleep in wild-type (WT) but not pcdr-1 and slo-1 mutants. In a heterologous expression system, Mel activates the human BK channel (hSlo1) in a membrane-delimited manner in the presence of the Mel receptor MT1 but not MT2 A peptide acting to release free Gβγ also activates hSlo1 in a MT1-dependent and membrane-delimited manner, whereas a Gβλ inhibitor abolishes the stimulating effect of Mel. Our results suggest that Mel promotes sleep by activating the BK channel through a specific Mel receptor and Gβλ.
Collapse
|
273
|
Chiral Recognition of Flexible Melatonin Receptor Ligands Induced by Conformational Equilibria. Molecules 2020; 25:molecules25184057. [PMID: 32899888 PMCID: PMC7570888 DOI: 10.3390/molecules25184057] [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: 07/28/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 01/17/2023] Open
Abstract
N-anilinoethylamides are a class of melatoninergic agents with the aniline portion mimicking the indole ring of the natural ligand and the ethylamide chain reproducing that of melatonin. The simplest compound in this class, N-{2-[(3-methoxyphenyl)methylamino]ethyl}acetamide (UCM793), has nanomolar binding affinity for MT1 and MT2 membrane receptors. To explore the effect of chain conformation on receptor binding, a methyl group was inserted on the methylene alpha or beta to the amide nitrogen and conformational equilibria were investigated by NMR spectroscopy and molecular dynamics simulations. Receptor affinity was conserved only for the beta-methyl derivative, which also showed significant stereoselectivity, with the (S) enantiomer being the eutomer. Molecular dynamics simulations, validated by NMR spectroscopy, showed that the beta-methyl group affects the conformational preferences of the ethylamide chain. Docking into the receptor crystal structure provides a rationale for the observed chiral recognition, suggesting that the (S)-beta-methyl group favors the conformation that better fits the receptor binding site.
Collapse
|
274
|
Bae H, Yang C, Lee JY, Park S, Bazer FW, Song G, Lim W. Melatonin improves uterine-conceptus interaction via regulation of SIRT1 during early pregnancy. J Pineal Res 2020; 69:e12670. [PMID: 32421880 DOI: 10.1111/jpi.12670] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Melatonin has been shown to improve in vitro fertilization and offspring survival after bacterial infection, but its role in regulating maternal-fetal communication during early pregnancy has not been investigated. Results of this study demonstrated expression of abundant melatonin receptors in conceptus and endometrium during early pregnancy. In gilts, expression of melatonin receptor 1A (MTNR1A or MT1) and melatonin receptor 1B (MTNR1B or MT2) increased in trophectoderm (Tr) and uterine luminal epithelium (LE) with advancing days during early pregnancy in a different manner. Melatonin increased proliferation and migration of porcine trophectoderm (pTr) cell, the percent pTr cells in the G2 phase of the cell cycle, and the expression of implantation-related genes by pTr cells and endometrial luminal epithelium (pLE). Melatonin also attenuated the production of LPS-induced pro-inflammatory cytokines and tunicamycin-induced endoplasmic reticulum (ER) stress-sensing proteins. The expression of sirtuin 1 (SIRT1) as a potential target of melatonin increased between Days 9 and 14 of gestation. Co-treatment with SIRT1 inhibitor EX527 and melatonin restored cell-cell interactions through PI3K and MAPK signaling. Knockdown of SIRT1 decreased the expression of implantation-related genes, as well as migration of pTr and pLE cells. The expression of microRNAs regulated by SIRT1 was suppressed in response to melatonin. Furthermore, melatonin significantly increased lipopolysaccharide (LPS)-reduced fertilization and embryogenesis in zebrafish model. These results suggest that melatonin may improve the uterine-conceptus interactions via the regulation of SIRT1 during early pregnancy.
Collapse
Affiliation(s)
- Hyocheol Bae
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Changwon Yang
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Jin-Young Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sunwoo Park
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Fuller W Bazer
- Department of Animal Science, Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, TX, USA
| | - Gwonhwa Song
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Whasun Lim
- Department of Food and Nutrition, College of Science and Technology, Kookmin University, Seoul, Korea
| |
Collapse
|
275
|
Reiter RJ, Rosales-Corral S, Sharma R. Circadian disruption, melatonin rhythm perturbations and their contributions to chaotic physiology. Adv Med Sci 2020; 65:394-402. [PMID: 32763813 DOI: 10.1016/j.advms.2020.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 06/15/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
The aim of this report is to summarize the data documenting the vital nature of well-regulated cellular and organismal circadian rhythms, which are also reflected in a stable melatonin cycle, in supporting optimal health. Cellular fluctuations in physiology exist in most cells of multicellular organisms with their stability relying on the prevailing light:dark cycle, since it regulates, via specialized intrinsically-photoreceptive retinal ganglion cells (ipRGC) and the retinohypothalamic tract, the master circadian oscillator, i.e., the suprachiasmatic nuclei (SCN). The output message of the SCN, as determined by the light:dark cycle, is transferred to peripheral oscillators, so-called slave cellular oscillators, directly via the autonomic nervous system with its limited distribution. and indirectly via the pineal-derived circulating melatonin rhythm, which contacts every cell. Via its regulatory effects on the neuroendocrine system, particularly the hypothalamo-pituitary-adrenal axis, the SCN also has a major influence on the adrenal glucocorticoid rhythm which impacts neurological diseases and psychological behaviors. Moreover, the SCN regulates the circadian production and secretion of melatonin. When the central circadian oscillator is disturbed, such as by light at night, it passes misinformation to all organs in the body. When this occurs the physiology of cells becomes altered and normal cellular functions are compromised. This physiological upheaval is a precursor to pathologies. The deterioration of the SCN/pineal network is often a normal consequence of aging and its related diseases, but in today's societies where manufactured light is becoming progressively more common worldwide, the associated pathologies may also be occurring at an earlier age.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX, USA.
| | - Sergio Rosales-Corral
- Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX, USA
| |
Collapse
|
276
|
Poza J, Pujol M, Ortega-Albás J, Romero O. Melatonin in sleep disorders. NEUROLOGÍA (ENGLISH EDITION) 2020; 37:575-585. [DOI: 10.1016/j.nrleng.2018.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/18/2018] [Indexed: 12/18/2022] Open
|
277
|
Chitimus DM, Popescu MR, Voiculescu SE, Panaitescu AM, Pavel B, Zagrean L, Zagrean AM. Melatonin's Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease. Biomolecules 2020; 10:biom10091211. [PMID: 32825327 PMCID: PMC7563541 DOI: 10.3390/biom10091211] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/30/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
There is a growing consensus that the antioxidant and anti-inflammatory properties of melatonin are of great importance in preserving the body functions and homeostasis, with great impact in the peripartum period and adult life. Melatonin promotes adaptation through allostasis and stands out as an endogenous, dietary, and therapeutic molecule with important health benefits. The anti-inflammatory and antioxidant effects of melatonin are intertwined and are exerted throughout pregnancy and later during development and aging. Melatonin supplementation during pregnancy can reduce ischemia-induced oxidative damage in the fetal brain, increase offspring survival in inflammatory states, and reduce blood pressure in the adult offspring. In adulthood, disturbances in melatonin production negatively impact the progression of cardiovascular risk factors and promote cardiovascular and neurodegenerative diseases. The most studied cardiovascular effects of melatonin are linked to hypertension and myocardial ischemia/reperfusion injury, while the most promising ones are linked to regaining control of metabolic syndrome components. In addition, there might be an emerging role for melatonin as an adjuvant in treating coronavirus disease 2019 (COVID 19). The present review summarizes and comments on important data regarding the roles exerted by melatonin in homeostasis and oxidative stress and inflammation related pathologies.
Collapse
Affiliation(s)
- Diana Maria Chitimus
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Mihaela Roxana Popescu
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, Elias University Hospital, 010164 Bucharest, Romania;
| | - Suzana Elena Voiculescu
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Anca Maria Panaitescu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, Filantropia Clinical Hospital, 010164 Bucharest, Romania;
| | - Bogdan Pavel
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Leon Zagrean
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Ana-Maria Zagrean
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
- Correspondence:
| |
Collapse
|
278
|
Berbets AM, Davydenko IS, Barbe AM, Konkov DH, Albota OM, Yuzko OM. Melatonin 1A and 1B Receptors' Expression Decreases in the Placenta of Women with Fetal Growth Restriction. Reprod Sci 2020; 28:197-206. [PMID: 32804352 DOI: 10.1007/s43032-020-00285-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/02/2020] [Indexed: 12/30/2022]
Abstract
Melatonin and its metabolites prevent oxidative stress and apoptosis, and it is actively produced by the placenta during pregnancy. Melatonin 1A and 1B receptors are present in human villous trophoblastic cells. We aimed to investigate the expression of melatonin 1A and 1B receptors in human placental tissue in the case of placental insufficiency manifested as the intrauterine growth restriction syndrome of the fetus (IUGR). Thirty-two pregnant women aged 18-36 with placental insufficiency manifested at the term 36 weeks of gestation as the IUGR syndrome (the estimated fetal weight less than the 3rd percentile) were included in the experimental group; all their babies had the diagnosis confirmed at birth, which occurred after 37 weeks of gestation. The control group consisted of 30 women with uncomplicated pregnancy of the same term. Pieces of the placental tissue were obtained after deliveries, and melatonin 1A and 1B receptors were immunoassayed; the richness of melatonin receptors in the placental tissue was estimated on the basis of immunohistochemical (IHC) staining of receptors, calculated in the IHC image score. The optical density of melatonin 1A receptors in the placentas obtained from women whose pregnancies were complicated with IUGR was significantly lower than that in the placentas from uncomplicated pregnancies: generally in the trophoblast, it was 0.095 ± 0.0009 IHC image score (in the control group, 0.194 ± 0.0015, p < 0.0001); in the apical parts of the syncytiotrophoblast, 0.108 ± 0.0016 IHC image score (in the control group, 0.221 ± 0.0013, p < 0.0001); and in the stromal cells of placental villi, 0.112 ± 0.0013 IHC image score (in the control group, 0.156 ± 0.0011, p < 0.0001). The optical density of melatonin 1B receptors in placentas obtained from women whose pregnancies were complicated with IUGR was also lower than that in the placentas from uncomplicated pregnancies: generally in the trophoblast, it was 0.165 ± 0.0019 IHC image score (in the control group, 0.231 ± 0.0013, p < 0.0001), and in the apical parts of the syncytiotrophoblast, 0.188 ± 0.0028 IHC image score (in the control group, 0.252 ± 0.0009, p < 0.0001). There was no difference found in the optical density of melatonin 1B receptors in the stromal cells of placental villi between the two groups: in the experimental group, 0.109 ± 0.006 IHC image score, and in the control group, 0.114 ± 0.0011 (p = 0.65). Melatonin receptors 1A and 1B are significantly less expressed in the placental tissue in the case that pregnancy is complicated with placental insufficiency, manifested as the intrauterine growth restriction syndrome of the fetus.
Collapse
Affiliation(s)
- Andrii M Berbets
- Higher State Educational Establishment of Ukraine "Bukovinian State Medical University", Chernivtsi, Ukraine.
| | - Igor S Davydenko
- Higher State Educational Establishment of Ukraine "Bukovinian State Medical University", Chernivtsi, Ukraine
| | - Adrian M Barbe
- Higher State Educational Establishment of Ukraine "Bukovinian State Medical University", Chernivtsi, Ukraine
| | - Dmytro H Konkov
- National Pirogov Memorial Medical University, Vinnytsia, Ukraine
| | - Olena M Albota
- Higher State Educational Establishment of Ukraine "Bukovinian State Medical University", Chernivtsi, Ukraine
| | - Oleksandr M Yuzko
- Higher State Educational Establishment of Ukraine "Bukovinian State Medical University", Chernivtsi, Ukraine
| |
Collapse
|
279
|
Nuszkiewicz J, Woźniak A, Szewczyk-Golec K. Ionizing Radiation as a Source of Oxidative Stress-The Protective Role of Melatonin and Vitamin D. Int J Mol Sci 2020; 21:E5804. [PMID: 32823530 PMCID: PMC7460937 DOI: 10.3390/ijms21165804] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/31/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023] Open
Abstract
Ionizing radiation (IR) has found widespread application in modern medicine, including medical imaging and radiotherapy. As a result, both patients and healthcare professionals are exposed to various IR doses. To minimize the negative side effects of radiation associated with oxidative imbalance, antioxidant therapy has been considered. In this review, studies on the effects of melatonin and vitamin D on radiation-induced oxidative stress are discussed. According to the research data, both substances meet the conditions for use as agents that protect humans against IR-induced tissue damage. Numerous studies have confirmed that melatonin, a hydro- and lipophilic hormone with strong antioxidant properties, can potentially be used as a radioprotectant in humans. Less is known about the radioprotective effects of vitamin D, but the results to date have been promising. Deficiencies in melatonin and vitamin D are common in modern societies and may contribute to the severity of adverse side effects of medical IR exposure. Hence, supporting supplementation with both substances seems to be of first importance. Interestingly, both melatonin and vitamin D have been found to selectively radiosensitise cancer cells, which makes them promising adjuvants in radiotherapy. More research is needed in this area, especially in humans.
Collapse
Affiliation(s)
- Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St, 85-092 Bydgoszcz, Poland;
| | | | - Karolina Szewczyk-Golec
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St, 85-092 Bydgoszcz, Poland;
| |
Collapse
|
280
|
Rosenthal SJ, Josephs T, Kovtun O, McCarty R. Seasonal effects on bipolar disorder: A closer look. Neurosci Biobehav Rev 2020; 115:199-219. [DOI: 10.1016/j.neubiorev.2020.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 11/15/2022]
|
281
|
Yang B, Zhang LY, Chen Y, Bai YP, Jia J, Feng JG, Liu KX, Zhou J. Melatonin alleviates intestinal injury, neuroinflammation and cognitive dysfunction caused by intestinal ischemia/reperfusion. Int Immunopharmacol 2020; 85:106596. [PMID: 32442902 DOI: 10.1016/j.intimp.2020.106596] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/29/2020] [Accepted: 05/10/2020] [Indexed: 12/31/2022]
Abstract
Intestinal ischemia/reperfusion (I/R) can cause multiple organ damage with extremely high morbidity and mortality. Melatonin has anti-inflammatory, anti-oxidative and anti-apoptotic effects against various diseases. This study aimed to explore whether melatonin had a protective effect against intestinal I/R-induced neuroinflammation and cognitive dysfunction, and investigate its potential mechanisms. In this study, melatonin was administered to the rats with intestinal I/R, then histological changes in intestine and brain (frontal cortex and hippocampal CA1 area) tissues and cognitive function were detected, respectively. The encephaledema and blood-brain barrier (BBB) permeability were observed. Moreover, the alterations of proinflammatory factors (tumor necrosis factor-α, interleukin-6 and interleukin-1β), oxidative response (malondialdehyde, superoxide dismutase, and reactive oxygen species), apoptosis and proteins associated with inflammation,including Toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (Myd88) and phosphorylated nuclear factor kappa beta (NF-κB), and apoptosis (cleaved caspase-3) in brain tissues were examined. Furthermore, the expressions of TLR4, Myd88, and microglial activity were observed by multiple immunofluorescence staining. The results showed that intestinal I/R-induced abnormal neurobehavior and cerebral damage were ameliorated after melatonin treatment, which were demonstrated by improved cognitive dysfunction and aggravated histology. Furthermore, melatonin decreased the levels of proinflammatory factors and oxidative stress in plasma, intestine and brain tissues, attenuated apoptotic cell, and inhibited the expressions of related proteins and the immunoreactivity of TLR4 or Myd88 in microglia in brain tissues. These findings showed that melatonin might relieve neuroinflammation and cognitive dysfunction caused by intestinal I/R, which could be, at least partially, related to the inhibition of the TLR4/Myd88 signaling in microglia.
Collapse
Affiliation(s)
- Bo Yang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Li-Yin Zhang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Yi-Ping Bai
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Jian-Guo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China.
| |
Collapse
|
282
|
Afsar B, Elsurer Afsar R, Sag AA, Kanbay A, Korkmaz H, Cipolla-Neto J, Covic A, Ortiz A, Kanbay M. Sweet dreams: therapeutic insights, targeting imaging and physiologic evidence linking sleep, melatonin and diabetic nephropathy. Clin Kidney J 2020; 13:522-530. [PMID: 32905249 PMCID: PMC7467577 DOI: 10.1093/ckj/sfz198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/16/2019] [Indexed: 12/25/2022] Open
Abstract
Melatonin is the main biochronologic molecular mediator of circadian rhythm and sleep. It is also a powerful antioxidant and has roles in other physiologic pathways. Melatonin deficiency is associated with metabolic derangements including glucose and cholesterol dysregulation, hypertension, disordered sleep and even cancer, likely due to altered immunity. Diabetic nephropathy (DN) is a key microvascular complication of both type 1 and 2 diabetes. DN is the end result of a complex combination of metabolic, haemodynamic, oxidative and inflammatory factors. Interestingly, these same factors have been linked to melatonin deficiency. This report will collate in a clinician-oriented fashion the mechanistic link between melatonin deficiency and factors contributing to DN.
Collapse
Affiliation(s)
- Baris Afsar
- Division of Nephrology, Department of Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Rengin Elsurer Afsar
- Division of Nephrology, Department of Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Alan A Sag
- Division of Vascular and Interventional Radiology, Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - Asiye Kanbay
- Department of Pulmonary Medicine, Istanbul Medeniyet University School of Medicine, Istanbul, Turkey
| | - Hakan Korkmaz
- Division of Endocrinology, Department of Medicine, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - José Cipolla-Neto
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Adrian Covic
- Dialysis Unit, School of Medicine, IIS-Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Nephrology Clinic, Dialysis and Renal Transplant Center, ‘C.I. PARHON’ University Hospital and ‘Grigore T. Popa’ University of Medicine, Iasi, Romania
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koç University School of Medicine, Istanbul, Turkey
| |
Collapse
|
283
|
Xu Y, Cui K, Li J, Tang X, Lin J, Lu X, Huang R, Yang B, Shi Y, Ye D, Huang J, Yu S, Liang X. Melatonin attenuates choroidal neovascularization by regulating macrophage/microglia polarization via inhibition of RhoA/ROCK signaling pathway. J Pineal Res 2020; 69:e12660. [PMID: 32323368 DOI: 10.1111/jpi.12660] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/30/2020] [Accepted: 04/18/2020] [Indexed: 12/16/2022]
Abstract
Choroidal neovascularization (CNV) is an important characteristic of advanced wet age-related macular degeneration (AMD) and leads to severe visual impairment among elderly patients. Previous studies have demonstrated that melatonin induces several biological effects related to antioxidation, anti-inflammation, and anti-angiogenesis. However, the role of melatonin in CNV, and its underlying mechanisms, has not been investigated thus far. In this study, we found that melatonin administration significantly reduced the scale and volume of CNV lesions, suppressed vascular leakage, and inhibited the capacity of vascular proliferation in the laser-induced mouse CNV model. Additionally, the results also show that the melatonin-treated retinal microglia in the laser-induced mice exhibited enhanced expression of M1-type markers, such as iNOS, CCL-3, CCL-5, and TNF-α, as well as decreased production of M2-type markers, such as Arg-1, Fizz-1, IL-10, YM-1, and CD206, indicating that melatonin switched the macrophage/microglia polarization from pro-angiogenic M2 phenotype to anti-angiogenic M1 phenotype. Furthermore, the RhoA/ROCK signaling pathway was activated during CNV formation, yet was suppressed after an intraperitoneal injection of melatonin. In conclusion, melatonin attenuated CNV, reduced vascular leakage, and inhibited vascular proliferation by switching the macrophage/microglia polarization from M2 phenotype to M1 phenotype via inhibition of RhoA/ROCK signaling pathway in CNV. This suggests that melatonin could be a novel agent for the treatment of AMD.
Collapse
Affiliation(s)
- Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jia Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jianqiang Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Rong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuxun Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Dan Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
284
|
Royant-Parola S, Kovess V, Brion A, Dagneaux S, Hartley S. Do hypnotics increase the risk of driving accidents or near miss accidents due to hypovigilance? The effects of sex, chronic sleepiness, sleep habits and sleep pathology. PLoS One 2020; 15:e0236404. [PMID: 32716956 PMCID: PMC7384619 DOI: 10.1371/journal.pone.0236404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 07/06/2020] [Indexed: 01/02/2023] Open
Abstract
Driving accidents due to hypovigilance are common but the role of hypnotics is unclear in patients suffering from sleep disorders. Our study examined factors influencing accidents and near miss accidents attributed to sleepiness at the wheel (ANMAS). Using data from an online questionnaire aimed at patients with sleep disorders, we analysed the associations between ANMAS, sociodemographic data, symptoms of sleep disorders, severity of insomnia (Insomnia Severity Index (ISI)) symptoms of anxiety and depression (Hospital Anxiety and Depression scale with depression (HADD) and anxiety (HADA) subscales), chronic sleepiness (Epworth sleepiness scale ESS), hypnotic use and information about sleep habits. Hypnotics were hierarchically grouped into Z-drugs, sedative medication, melatonin and over the counter (OTC) alternative treatments. Of 10802 participants; 9.1% reported ANMAS (Men 11.1% women 8.3%) and 24.4% took hypnotics (Z-drugs 8.5%, sedative medication 8%, melatonin 5.6% and alternative treatments 2.5%). Logistic regression analysis identified the following risk factors for ANMAS: moderate (OR 2.4; CI: 2.10-2.79) and severe sleepiness (ESS OR 5.66; CI: 4.74-6.77), depression (HADD OR 1.2; CI: 1.03-1.47), anxiety (HADA OR 1.2;CI: 1.01-1.47), and insufficient sleep (OR1.4; CI: 1.2-1.7). Hypnotics were not associated with an increased risk of ANMAS in patients suffering from insomnia. Risk factors varied according to sex: in females, sex (OR 0.; CI: 0.55-0.74), mild insomnia (OR 0.5; CI: 0.3-0.8) and use of alternative treatments (OR 0.455, CI:0.23-0.89) were protective factors and risk was increased by sleepiness, sleep debt, social jetlag, caffeine use, anxiety and depression. In men no protective factors were identified: sleepiness, sleep debt, and severe insomnia were associated with an increased risk of ANMAS. In clinical practice, all patients with daytime sleepiness and men with severe insomnia should be counselled concerning driving risk and encouraged to avoid sleep debt.
Collapse
Affiliation(s)
| | | | | | | | - Sarah Hartley
- Réseau Morphée, Garches, France
- EA 4047, APHP Hôpital Raymond Poincaré, Sleep Center, Université de Versailles Saint-Quentin en Yvelines, Garches, France
| |
Collapse
|
285
|
Reiter RJ, Sharma R, Ma Q, Rorsales-Corral S, de Almeida Chuffa LG. Melatonin inhibits Warburg-dependent cancer by redirecting glucose oxidation to the mitochondria: a mechanistic hypothesis. Cell Mol Life Sci 2020; 77:2527-2542. [PMID: 31970423 PMCID: PMC11104865 DOI: 10.1007/s00018-019-03438-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022]
Abstract
Melatonin has the ability to intervene in the initiation, progression and metastasis of some experimental cancers. A large variety of potential mechanisms have been advanced to describe the metabolic and molecular events associated with melatonin's interactions with cancer cells. There is one metabolic perturbation that is common to a large number of solid tumors and accounts for the ability of cancer cells to actively proliferate, avoid apoptosis, and readily metastasize, i.e., they use cytosolic aerobic glycolysis (the Warburg effect) to rapidly generate the necessary ATP required for the high metabolic demands of the cancer cells. There are several drugs, referred to as glycolytic agents, that cause cancer cells to abandon aerobic glycolysis and shift to the more conventional mitochondrial oxidative phosphorylation for ATP synthesis as in normal cells. In doing so, glycolytic agents also inhibit cancer growth. Herein, we hypothesize that melatonin also functions as an inhibitor of cytosolic glycolysis in cancer cells using mechanisms, i.e., downregulation of the enzyme (pyruvate dehydrogenase kinase) that interferes with the conversion of pyruvate to acetyl CoA in the mitochondria, as do other glycolytic drugs. In doing so, melatonin halts the proliferative activity of cancer cells, reduces their metastatic potential and causes them to more readily undergo apoptosis. This hypothesis is discussed in relation to the previously published reports. Whereas melatonin is synthesized in the mitochondria of normal cells, we hypothesize that this synthetic capability is not present in cancer cell mitochondria because of the depressed acetyl CoA; acetyl CoA is necessary for the rate limiting enzyme in melatonin synthesis, arylalkylamine-N-acetyltransferase. Finally, the ability of melatonin to switch glucose oxidation from the cytosol to the mitochondria also explains how tumors that become resistant to conventional chemotherapies are re-sensitized to the same treatment when melatonin is applied.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA.
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Qiang Ma
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Sergio Rorsales-Corral
- Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Mexico
| | | |
Collapse
|
286
|
Samanta S. Melatonin: an endogenous miraculous indolamine, fights against cancer progression. J Cancer Res Clin Oncol 2020; 146:1893-1922. [PMID: 32583237 DOI: 10.1007/s00432-020-03292-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Melatonin is an amphipathic indolamine molecule ubiquitously present in all organisms ranging from cyanobacteria to humans. The pineal gland is the site of melatonin synthesis and secretion under the influence of the retinohypothalamic tract. Some extrapineal tissues (skin, lens, gastrointestinal tract, testis, ovary, lymphocytes, and astrocytes) also enable to produce melatonin. Physiologically, melatonin regulates various functions like circadian rhythm, sleep-wake cycle, gonadal activity, redox homeostasis, neuroprotection, immune-modulation, and anticancer effects in the body. Inappropriate melatonin secretion advances the aging process, tumorigenesis, visceral adiposity, etc. METHODS: For the preparation of this review, I had reviewed the literature on the multidimensional activities of melatonin from the NCBI website database PubMed, Springer Nature, Science Direct (Elsevier), Wiley Online ResearchGate, and Google Scholar databases to search relevant articles. Specifically, I focused on the roles and mechanisms of action of melatonin in cancer prevention. RESULTS The actions of melatonin are primarily mediated by G-protein coupled MT1 and MT2 receptors; however, several intracellular protein and nuclear receptors can modulate the activity. Normal levels of the melatonin protect the cells from adverse effects including carcinogenesis. Therapeutically, melatonin has chronomedicinal value; it also shows a remarkable anticancer property. The oncostatic action of melatonin is multidimensional, associated with the advancement of apoptosis, the arrest of the cell cycle, inhibition of metastasis, and antioxidant activity. CONCLUSION The present review has emphasized the mechanism of the anti-neoplastic activity of melatonin that increases the possibilities of the new approaches in cancer therapy.
Collapse
Affiliation(s)
- Saptadip Samanta
- Department Physiology, Midnapore College, Paschim Medinipur, Midnapore, West Bengal, 721101, India.
| |
Collapse
|
287
|
Infusion of Melatonin Into the Paraventricular Nucleus Ameliorates Myocardial Ischemia-Reperfusion Injury by Regulating Oxidative Stress and Inflammatory Cytokines. J Cardiovasc Pharmacol 2020; 74:336-347. [PMID: 31356536 PMCID: PMC6791501 DOI: 10.1097/fjc.0000000000000711] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Melatonin, the receptors for which are abundant in the hypothalamic paraventricular nucleus (PVN), can protect the heart from myocardial ischemia–reperfusion (MI/R) injury. The aim of this study was to determine whether the infusion of melatonin into the PVN protects the heart from MI/R injury by suppressing oxidative stress or regulating the balance between proinflammatory cytokines and anti-inflammatory cytokines in MI/R rats. Male Sprague–Dawley rats were treated with a bilateral PVN infusion of melatonin. MI/R operation was performed 1 week after infusion. At the end of the third week after the infusion, all the rats were euthanized. This was followed by immunohistochemistry and immunofluorescence studies of the rats. MI/R rats showed larger infarct size, increased left ventricular (LV) end-diastolic volume, and decreased LV ejection fraction and LV fractional shortening. Moreover, MI/R rats had a higher level of norepinephrine in the plasma, heart, and PVN; higher PVN levels of reactive oxygen species, NOX2, NOX4, IL-1β, and NF-κB activity; and lower PVN levels of copper/zinc superoxide dismutase (Cu/Zn-SOD) and IL-10 compared with the sham group. Melatonin infusion in PVN reduced LV end-diastolic volume, norepinephrine, reactive oxygen species, NOX2, NOX4, IL-1β, and NF-κB activity, and increased LV ejection fraction, LV fractional shortening, Cu/Zn-SOD, and IL-10. Overall, these results suggest that the infusion of melatonin ameliorates sympathetic nerve activity and MI/R injury by attenuating oxidative stress and inflammatory cytokines in the PVN of MI/R rats.
Collapse
|
288
|
Gurunathan S, Kang MH, Kim JH. Role and Therapeutic Potential of Melatonin in the Central Nervous System and Cancers. Cancers (Basel) 2020; 12:cancers12061567. [PMID: 32545820 PMCID: PMC7352348 DOI: 10.3390/cancers12061567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
Melatonin (MLT) is a powerful chronobiotic hormone that controls a multitude of circadian rhythms at several levels and, in recent times, has garnered considerable attention both from academia and industry. In several studies, MLT has been discussed as a potent neuroprotectant, anti-apoptotic, anti-inflammatory, and antioxidative agent with no serious undesired side effects. These characteristics raise hopes that it could be used in humans for central nervous system (CNS)-related disorders. MLT is mainly secreted in the mammalian pineal gland during the dark phase, and it is associated with circadian rhythms. However, the production of MLT is not only restricted to the pineal gland; it also occurs in the retina, Harderian glands, gut, ovary, testes, bone marrow, and lens. Although most studies are limited to investigating the role of MLT in the CNS and related disorders, we explored a considerable amount of the existing literature. The objectives of this comprehensive review were to evaluate the impact of MLT on the CNS from the published literature, specifically to address the biological functions and potential mechanism of action of MLT in the CNS. We document the effectiveness of MLT in various animal models of brain injury and its curative effects in humans. Furthermore, this review discusses the synthesis, biology, function, and role of MLT in brain damage, and as a neuroprotective, antioxidative, anti-inflammatory, and anticancer agent through a collection of experimental evidence. Finally, it focuses on the effect of MLT on several neurological diseases, particularly CNS-related injuries.
Collapse
|
289
|
Liu L, Jockers R. Structure-Based Virtual Screening Accelerates GPCR Drug Discovery. Trends Pharmacol Sci 2020; 41:382-384. [DOI: 10.1016/j.tips.2020.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 04/12/2020] [Indexed: 12/18/2022]
|
290
|
Song YJ, Zhong CB, Wu W. Cardioprotective effects of melatonin: Focusing on its roles against diabetic cardiomyopathy. Biomed Pharmacother 2020; 128:110260. [PMID: 32447213 DOI: 10.1016/j.biopha.2020.110260] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/01/2020] [Accepted: 05/10/2020] [Indexed: 02/06/2023] Open
Abstract
Melatonin is a pineal-produced indole known for its anti-aging, antiapoptotic and antioxidant properties. In past decades, the protective potentials of melatonin for cardiovascular diseases, such as atherosclerosis and myocardial infarction, have been widely revealed, triggering more investigations focused on other cardioprotective effects of melatonin. Recently, the roles of melatonin in diabetic cardiomyopathy (DCM) have attracted increased attention. In this regard, researchers found that melatonin attenuated cardiac fibrosis and hypertrophy, thus interrupting the development of DCM. Retinoid-related orphan receptor α is a key melatonin receptor that contributed to the cardioprotective effect of melatonin in hearts with DCM. For the downstream mechanisms, the inhibition of mammalian STE20-like kinase 1 plays a pivotal role, which exerts antiapoptotic and proautophagic effects, thus enhancing cardiac tolerance in high-glucose conditions. In addition, other signalling mechanisms, such as sirtuin-1/peroxisome proliferator-activated receptor gamma-coactivator alpha and endoplasmic reticulum-related signalling, are also involved in the protective effects of melatonin on cardiomyocytes under diabetic conditions. This review will focus on the protective signalling mechanisms regulated by melatonin and provide a better understanding of the therapeutic applications of melatonin signalling in DCM.
Collapse
Affiliation(s)
- Yan-Jun Song
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, 1 Shuai Fu Yuan, Beijing, 100730, PR China.
| | - Chong-Bin Zhong
- Department of Cardiology, Heart Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, PR China.
| | - Wei Wu
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, 1 Shuai Fu Yuan, Beijing, 100730, PR China.
| |
Collapse
|
291
|
5-Azacytidine upregulates melatonin MT1 receptor expression in rat C6 glioma cells: oncostatic implications. Mol Biol Rep 2020; 47:4867-4873. [DOI: 10.1007/s11033-020-05482-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
|
292
|
Chan KH, Tse LH, Huang X, Wong YH. Molecular basis defining the selectivity of substituted isoquinolinones for the melatonin MT 2 receptor. Biochem Pharmacol 2020; 177:114020. [PMID: 32389636 DOI: 10.1016/j.bcp.2020.114020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/04/2020] [Indexed: 10/24/2022]
Abstract
Melatonin MT1 and MT2 receptors represent attractive drug targets for the treatment of various disorders. However, the high conservation of the melatonin binding pocket has hindered the development of subtype-selective compounds. By leveraging on the recently resolved crystal structures of MT1 and MT2 receptors, this study aims to elucidate the structural basis of MT2-selectivity of a panel of isoquinolinone derivatives. Molecular modelling and ligand docking approaches were employed to predict residues involved in forming interactions with the MT2-selective isoquinolinones. Seven conserved residues (Asn175, His208, Trp264, Asn268, Gly271, Tyr294 and Tyr298) were selected as targets for site-directed mutagenesis. Ca2+ mobilization, cAMP inhibition, phosphorylation of extracellular signal-regulated kinase, and ligand binding assays were performed to functionally characterize the receptor mutants in transfected CHO cells. Unlike melatonin, isoquinolinones bearing a 3-methoxybenzyloxyl substituent were unaffected by alanine substitution at His208 of MT2. Although alanine substitutions at Tyr294 or Tyr298 reduced the potency of melatonin and some isoquinolinones on MT2, similar mutations on MT1 allowed five hitherto ineffective isoquinolinones to act as agonists. An isoquinolinone antagonist bearing a 4-methoxybenzyloxyl moiety turned into an agonist at MT2 mutants with alanine substitutions at His208, Tyr294 or Tyr298. A subset of residues is apparently involved in forming a hydrophobic binding cavity to confer selectivity upon the aromatic substituent of isoquinolinone compounds. Two conserved tyrosine residues on transmembrane helix 7 may confer ligand selectivity at MT1 and MT2 receptors, while a conserved histidine on transmembrane helix 5 is apparently involved in receptor activation.
Collapse
Affiliation(s)
- King H Chan
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Lap H Tse
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xuhui Huang
- Department of Chemistry, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, and the Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yung H Wong
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; State Key Laboratory of Molecular Neuroscience, and the Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
293
|
Chen M, Cecon E, Karamitri A, Gao W, Gerbier R, Ahmad R, Jockers R. Melatonin MT 1 and MT 2 receptor ERK signaling is differentially dependent on G i/o and G q/11 proteins. J Pineal Res 2020; 68:e12641. [PMID: 32080899 DOI: 10.1111/jpi.12641] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/15/2020] [Accepted: 02/17/2020] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) transmit extracellular signals into cells by activating G protein- and β-arrestin-dependent pathways. Extracellular signal-regulated kinases (ERKs) play a central role in integrating these different linear inputs coming from a variety of GPCRs to regulate cellular functions. Here, we investigated human melatonin MT1 and MT2 receptors signaling through the ERK1/2 cascade by employing different biochemical techniques together with pharmacological inhibitors and siRNA molecules. We show that ERK1/2 activation by both receptors is exclusively G protein-dependent, without any participation of β-arrestin1/2 in HEK293 cells. ERK1/2 activation by MT1 is only mediated though Gi/o proteins, while MT2 is dependent on the cooperative activation of Gi/o and Gq/11 proteins. In the absence of Gq/11 proteins, however, MT2 -induced ERK1/2 activation switches to a β-arrestin1/2-dependent mode. The signaling cascade downstream of G proteins is the same for both receptors and involves activation of the PI3K/PKCζ/c-Raf/MEK/ERK cascade. The differential G protein dependency of MT1 - and MT2 -mediated ERK activation was confirmed at the level of EGR1 and FOS gene expression, two ERK1/2 target genes. Gi/o /Gq/11 cooperativity was also observed in Neuroscreen-1 cells expressing endogenous MT2 , whereas in the mouse retina, where MT2 is engaged into MT1 /MT2 heterodimers, ERK1/2 signaling is exclusively Gi/o -dependent. Collectively, our data reveal differential signaling modes of MT1 and MT2 in terms of ERK1/2 activation, with an unexpected Gi/o /Gq/11 cooperativity exclusively for MT2 . The plasticity of ERK activation by MT2 is highlighted by the switch to a β-arrestin1/2-dependent mode in the absence of Gq/11 proteins and by the switch to a Gi/o mode when engaged into MT1 /MT2 heterodimers, revealing a new mechanism underlying tissue-specific responses to melatonin.
Collapse
Affiliation(s)
- Min Chen
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | - Erika Cecon
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | | | - Wenwen Gao
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | - Romain Gerbier
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | - Raise Ahmad
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| | - Ralf Jockers
- Institut Cochin, CNRS, INSERM, Université de Paris, Paris, France
| |
Collapse
|
294
|
Fu Z, Jiao Y, Wang J, Zhang Y, Shen M, Reiter RJ, Xi Q, Chen Y. Cardioprotective Role of Melatonin in Acute Myocardial Infarction. Front Physiol 2020; 11:366. [PMID: 32411013 PMCID: PMC7201093 DOI: 10.3389/fphys.2020.00366] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
Melatonin is a pleiotropic, indole secreted, and synthesized by the human pineal gland. Melatonin has biological effects including anti-apoptosis, protecting mitochondria, anti-oxidation, anti-inflammation, and stimulating target cells to secrete cytokines. Its protective effect on cardiomyocytes in acute myocardial infarction (AMI) has caused widespread interest in the actions of this molecule. The effects of melatonin against oxidative stress, promoting autophagic repair of cells, regulating immune and inflammatory responses, enhancing mitochondrial function, and relieving endoplasmic reticulum stress, play crucial roles in protecting cardiomyocytes from infarction. Mitochondrial apoptosis and dysfunction are common occurrence in cardiomyocyte injury after myocardial infarction. This review focuses on the targets of melatonin in protecting cardiomyocytes in AMI, the main molecular signaling pathways that melatonin influences in its endogenous protective role in myocardial infarction, and the developmental prospect of melatonin in myocardial infarction treatment.
Collapse
Affiliation(s)
- Zhenhong Fu
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yang Jiao
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jihang Wang
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ying Zhang
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mingzhi Shen
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, San Antonio, TX, United States
- San Antonio Cellular Therapeutics Institute, Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, TX, United States
| | - Qing Xi
- The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
295
|
Mirza-Aghazadeh-Attari M, Reiter RJ, Rikhtegar R, Jalili J, Hajalioghli P, Mihanfar A, Majidinia M, Yousefi B. Melatonin: An atypical hormone with major functions in the regulation of angiogenesis. IUBMB Life 2020; 72:1560-1584. [PMID: 32329956 DOI: 10.1002/iub.2287] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/10/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023]
Abstract
Melatonin (N-acetyl-5-methoxytryptamine), a pleotropic molecule with a wide distribution, has received considerable attention in recent years, mostly because of its various major effects on tissues or cells since it has both receptor-dependent and receptor-independent actions over a wide range of concentrations. These biological and physiological functions of melatonin include regulation of circadian rhythms by modulating the expression of core oscillator genes, scavenging the reactive oxygen species and reactive nitrogen species, modulating the immune system and inflammatory response, and exerting cytoprotective and antiapoptotic effects. Given the multiple critical roles of melatonin, dysregulation of its production or any disruption in signaling through its receptors may have contributed in the development of a wide range of disorders including type 2 diabetes, aging, immune-mediated diseases, hypertension, and cancer. Herein, we focus on the modulatory effects of melatonin on angiogenesis and its implications as a therapeutic strategy in cancer and related diseases.
Collapse
Affiliation(s)
- Mohammad Mirza-Aghazadeh-Attari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Reza Rikhtegar
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Jalili
- Radiology Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Hajalioghli
- Radiology Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ainaz Mihanfar
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
296
|
The effects of melatonin prophylaxis on sensory recovery and postoperative pain following orthognathic surgery: a triple-blind randomized controlled trial and biochemical analysis. Int J Oral Maxillofac Surg 2020; 49:446-453. [DOI: 10.1016/j.ijom.2019.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/21/2019] [Accepted: 07/04/2019] [Indexed: 01/30/2023]
|
297
|
How Sure Can We Be about ML Methods-Based Evaluation of Compound Activity: Incorporation of Information about Prediction Uncertainty Using Deep Learning Techniques. Molecules 2020; 25:molecules25061452. [PMID: 32210186 PMCID: PMC7144469 DOI: 10.3390/molecules25061452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/28/2020] [Accepted: 03/22/2020] [Indexed: 11/17/2022] Open
Abstract
A great variety of computational approaches support drug design processes, helping in selection of new potentially active compounds, and optimization of their physicochemical and ADMET properties. Machine learning is a group of methods that are able to evaluate in relatively short time enormous amounts of data. However, the quality of machine-learning-based prediction depends on the data supplied for model training. In this study, we used deep neural networks for the task of compound activity prediction and developed dropout-based approaches for estimating prediction uncertainty. Several types of analyses were performed: the relationships between the prediction error, similarity to the training set, prediction uncertainty, number and standard deviation of activity values were examined. It was tested whether incorporation of information about prediction uncertainty influences compounds ranking based on predicted activity and prediction uncertainty was used to search for the potential errors in the ChEMBL database. The obtained outcome indicates that incorporation of information about uncertainty of compound activity prediction can be of great help during virtual screening experiments.
Collapse
|
298
|
Wu GC, Peng CK, Liao WI, Pao HP, Huang KL, Chu SJ. Melatonin receptor agonist protects against acute lung injury induced by ventilator through up-regulation of IL-10 production. Respir Res 2020; 21:65. [PMID: 32143642 PMCID: PMC7059294 DOI: 10.1186/s12931-020-1325-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
Background It is well known that ventilation with high volume or pressure may damage healthy lungs or worsen injured lungs. Melatonin has been reported to be effective in animal models of acute lung injury. Melatonin exerts its beneficial effects by acting as a direct antioxidant and via melatonin receptor activation. However, it is not clear whether melatonin receptor agonist has a protective effect in ventilator-induced lung injury (VILI). Therefore, in this study, we determined whether ramelteon (a melatonin receptor agonist) can attenuate VILI and explore the possible mechanism for protection. Methods VILI was induced by high tidal volume ventilation in a rat model. The rats were randomly allotted into the following groups: control, control+melatonin, control+ramelteon, control+luzindole, VILI, VILI+luzindole, VILI + melatonin, VILI + melatonin + luzindole (melatonin receptor antagonist), VILI + ramelteon, and VILI + ramelteon + luzindole (n = 6 per group). The role of interleukin-10 (IL-10) in the melatonin- or ramelteon-mediated protection against VILI was also investigated. Results Ramelteon treatment markedly reduced lung edema, serum malondialdehyde levels, the concentration of inflammatory cytokines in bronchoalveolar lavage fluid (BALF), NF-κB activation, iNOS levels, and apoptosis in the lung tissue. Additionally, ramelteon treatment significantly increased heat shock protein 70 expression in the lung tissue and IL-10 levels in BALF. The protective effect of ramelteon was mitigated by the administration of luzindole or an anti-IL-10 antibody. Conclusions Our results suggest that a melatonin receptor agonist has a protective effect against VILI, and its protective mechanism is based on the upregulation of IL-10 production.
Collapse
Affiliation(s)
- Geng-Chin Wu
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Chung-Kan Peng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan. .,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, No. 325, Section 2, Chenggong Road, Neihu, Taipei, 114, Taiwan.
| |
Collapse
|
299
|
Patel N, Huang XP, Grandner JM, Johansson LC, Stauch B, McCorvy JD, Liu Y, Roth B, Katritch V. Structure-based discovery of potent and selective melatonin receptor agonists. eLife 2020; 9:e53779. [PMID: 32118583 PMCID: PMC7080406 DOI: 10.7554/elife.53779] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Melatonin receptors MT1 and MT2 are involved in synchronizing circadian rhythms and are important targets for treating sleep and mood disorders, type-2 diabetes and cancer. Here, we performed large scale structure-based virtual screening for new ligand chemotypes using recently solved high-resolution 3D crystal structures of agonist-bound MT receptors. Experimental testing of 62 screening candidates yielded the discovery of 10 new agonist chemotypes with sub-micromolar potency at MT receptors, with compound 21 reaching EC50 of 0.36 nM. Six of these molecules displayed selectivity for MT2 over MT1. Moreover, two most potent agonists, including 21 and a close derivative of melatonin, 28, had dramatically reduced arrestin recruitment at MT2, while compound 37 was devoid of Gi signaling at MT1, implying biased signaling. This study validates the suitability of the agonist-bound orthosteric pocket in the MT receptor structures for the structure-based discovery of selective agonists.
Collapse
Affiliation(s)
- Nilkanth Patel
- Department of Biological Sciences and Department of Chemistry, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern CaliforniaLos AngelesUnited States
| | - Xi Ping Huang
- Department of Pharmacology, University of North Carolina Chapel Hill Medical SchoolChapel HillUnited States
- National Institute of Mental Health Psychoactive Drug Screening Program, Department of Pharmacology, University of North Carolina Chapel Hill Medical SchoolChapel HillUnited States
| | - Jessica M Grandner
- Department of Biological Sciences and Department of Chemistry, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern CaliforniaLos AngelesUnited States
| | - Linda C Johansson
- Department of Biological Sciences and Department of Chemistry, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern CaliforniaLos AngelesUnited States
| | - Benjamin Stauch
- Department of Biological Sciences and Department of Chemistry, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern CaliforniaLos AngelesUnited States
| | - John D McCorvy
- Department of Pharmacology, University of North Carolina Chapel Hill Medical SchoolChapel HillUnited States
- National Institute of Mental Health Psychoactive Drug Screening Program, Department of Pharmacology, University of North Carolina Chapel Hill Medical SchoolChapel HillUnited States
| | - Yongfeng Liu
- Department of Pharmacology, University of North Carolina Chapel Hill Medical SchoolChapel HillUnited States
- National Institute of Mental Health Psychoactive Drug Screening Program, Department of Pharmacology, University of North Carolina Chapel Hill Medical SchoolChapel HillUnited States
| | - Bryan Roth
- Department of Pharmacology, University of North Carolina Chapel Hill Medical SchoolChapel HillUnited States
- National Institute of Mental Health Psychoactive Drug Screening Program, Department of Pharmacology, University of North Carolina Chapel Hill Medical SchoolChapel HillUnited States
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina Chapel Hill Medical SchoolChapel HillUnited States
| | - Vsevolod Katritch
- Department of Biological Sciences and Department of Chemistry, Bridge Institute, USC Michelson Center for Convergent Biosciences, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
300
|
Morton AJ, Middleton B, Rudiger S, Bawden CS, Kuchel TR, Skene DJ. Increased plasma melatonin in presymptomatic Huntington disease sheep (Ovis aries): Compensatory neuroprotection in a neurodegenerative disease? J Pineal Res 2020; 68:e12624. [PMID: 31742766 DOI: 10.1111/jpi.12624] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/05/2019] [Accepted: 11/14/2019] [Indexed: 01/09/2023]
Abstract
Melatonin is a pleiotrophic hormone, synthesised primarily by the pineal gland under the control of the suprachiasmatic nuclei (SCN). It not only provides a hormonal signal of darkness but also has neuroprotective properties. Huntington's disease (HD) is a progressive neurodegenerative disorder characterised by abnormal motor, cognitive and psychiatric symptoms. There is growing evidence, particularly from animal models, that circadian rhythms may also be disturbed in HD. We measured two circadian-regulated hormones, melatonin and cortisol, in plasma samples collected around-the-clock from normal and presymptomatic transgenic HD sheep (Ovis aries) at 5 and 7 years of age, to assess SCN-driven rhythms and the effect of genotype, sex and age. Melatonin-related precursors and metabolites (tryptophan, serotonin, kynurenine) were also measured by liquid chromatography (LC)-mass spectrometry (MS). At 5 years of age in both rams and ewes, plasma melatonin levels were significantly elevated in HD sheep. In ewes measured 2 years later, there was still a significant elevation of nocturnal melatonin. Furthermore, the daytime baseline levels of melatonin were significantly higher in HD sheep. Since increased melatonin could have global beneficial effects on brain function, we suggest that the increased melatonin measured in presymptomatic HD sheep is part of an autoprotective response to mutant huntingtin toxicity that may account, at least in part, for the late onset of disease that characterises HD.
Collapse
Affiliation(s)
- A Jennifer Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Benita Middleton
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Skye Rudiger
- South Australian Research and Development Institute, Roseworthy, South Australia, Australia
| | - C Simon Bawden
- South Australian Research and Development Institute, Roseworthy, South Australia, Australia
| | - Timothy R Kuchel
- Preclinical, Imaging and Research Laboratories (PIRL), SAHMRI, Adelaide, South Australia, Australia
| | - Debra J Skene
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|