1
|
Kargar A, Fetratjoo DH, Moattar R, Tarki A, Golsokhan A, Pouyan N, Amjadi-Goojgi Z, Mostafaei H, Kakeri F, Zendehbad AS, Lima BS, Esmaily H, Noroozian M. Efficacy of Add-On Agomelatine on Agitation, Aggression, and Neuroprotection in Alzheimer's Disease: A Randomized, Blinded, Controlled Trial. Am J Geriatr Psychiatry 2025:S1064-7481(25)00082-X. [PMID: 40221303 DOI: 10.1016/j.jagp.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Agitation and aggression are challenging behavioral and psychological symptoms of dementia (BPSD) in Alzheimer's disease (AD), which is diagnosed based on NINCDS-ADRDA criteria. Limited efficacy and safety of existing pharmacotherapies complicate treatment. Agomelatine, a melatonergic agonist and serotonergic antagonist, may provide a safer and more effective alternative. OBJECTIVES The primary outcome is to evaluate the efficacy of agomelatine as an add-on therapy to nonpharmacological treatment in reducing agitation and aggression in patients with AD, with secondary outcomes assessing its safety and potential neuroprotective effects. METHODS In a randomized, triple-blind, placebo-controlled trial, 56 patients with AD (aged ≥65) experiencing agitation and aggression received 12.5 mg agomelatine (n = 28) or placebo (n = 28) daily for 6 weeks. The primary outcome was the Cohen-Mansfield Agitation Inventory (CMAI) score change. Secondary outcomes included serum brain-derived neurotrophic factor (BDNF) levels and safety. RESULTS At 6 weeks, the agomelatine group showed a greater reduction in CMAI scores than placebo (mean difference: -12.39, 95% CI: -19.37 to -5.42; p = 0.001). No significant BDNF changes were detected (p = 0.848). Total adverse events (AEs) were more common in the agomelatine group (50.0% vs. 21.4%, p = 0.050), but no serious AEs or liver enzyme abnormalities were reported. CONCLUSION A daily dose of 12.5 mg agomelatine may effectively and safely reduce agitation and aggression in patients with AD when used as an add-on therapy. However, further studies with larger samples and extended durations are needed to definitively confirm agomelatine's role in managing BPSD in AD.
Collapse
Affiliation(s)
- Alireza Kargar
- Department of Clinical Pharmacy (AK, HE), School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cognitive Neurology (AK, DHF, RM, AT, ZAG, MN), Dementia, and Neuropsychiatry Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Delara Hazegh Fetratjoo
- Cognitive Neurology (AK, DHF, RM, AT, ZAG, MN), Dementia, and Neuropsychiatry Research Center, Tehran University of Medical Sciences, Tehran, Iran; School of Pharmacy (DHF), Tehran University of Medical Sciences, Tehran, Iran
| | - Reihaneh Moattar
- Cognitive Neurology (AK, DHF, RM, AT, ZAG, MN), Dementia, and Neuropsychiatry Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Anahita Tarki
- Cognitive Neurology (AK, DHF, RM, AT, ZAG, MN), Dementia, and Neuropsychiatry Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Health Psychology (AT), Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Aram Golsokhan
- Yaadmaan Institute for Brain (AG, HM, FK), Cognition, and Memory Studies, Tehran, Iran; Department of Psychology (AG), Faculty of Psychology and Educational Sciences, Islamic Azad University, Karaj Branch, Karaj, Iran
| | - Niloufar Pouyan
- Department of Anesthesiology (NP), University of Michigan Medical School, Ann Arbor, MI; Neuroscience Graduate Program (NP), University of Michigan, Ann Arbor, MI
| | - Zahra Amjadi-Goojgi
- Cognitive Neurology (AK, DHF, RM, AT, ZAG, MN), Dementia, and Neuropsychiatry Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamideh Mostafaei
- Yaadmaan Institute for Brain (AG, HM, FK), Cognition, and Memory Studies, Tehran, Iran
| | - Fariba Kakeri
- Yaadmaan Institute for Brain (AG, HM, FK), Cognition, and Memory Studies, Tehran, Iran
| | - Azadeh Sadat Zendehbad
- Department of Geriatric Health (ASZ), Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Behnam Safarpour Lima
- Department of Neurology (BSL), School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Esmaily
- Department of Clinical Pharmacy (AK, HE), School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maryam Noroozian
- Cognitive Neurology (AK, DHF, RM, AT, ZAG, MN), Dementia, and Neuropsychiatry Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Mahmood NMS, Mahmud AMR, Maulood IM. The interactions between melatonin and the renin-angiotensin system (RAS) in vascular attenuation in diabetic and non-diabetic conditions. Acta Diabetol 2025:10.1007/s00592-025-02479-2. [PMID: 40080199 DOI: 10.1007/s00592-025-02479-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 02/23/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND The hormone melatonin (MEL), primarily acknowledged for its role in regulating circadian rhythms, has demonstrated itself to be a complicated molecule with significant implications for vascular physiology. Melatonin exerts extensive physiological effects directly via the MEL receptor type 1 (MT1R) and the MEL receptor type 2 (MT2R), as well as indirectly through the improvement of antioxidant vascular tone. OBJECTIVE This review aims to analyse the intricate relationships between MEL and the renin-angiotensin system (RAS) in the vascular attenuation of non-diabetic (non-DM) and diabetic (DM) contexts. Alterations in the expression of RAS components and their dysregulation are prevalent in diabetes. Melatonin exhibits vasoprotective advantages in non-diabetic conditions. In the context of DM, vascular problrms such as vascular endothelial dysfunction (VED), hypertension, and atherosclerosis result from the dysregulation of MEL-RAS interactions. Comprehending the actions of MEL on RAS components in diabetes vasculature is essential for formulating tailored pharmaceutical therapy methods. CONCLUSION This review consolidates existing knowledge regarding the vascular effects of MEL in relation to RAS activation, emphasising its potential role as a modulating factor for angiotensin 1-8 (Ang 1-8), angiotensin-converting enzyme 2 (ACE2), and angiotensin 1-7 (Ang 1-7) in the management of vascular complications associated with DM.
Collapse
Affiliation(s)
- Nazar M Shareef Mahmood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq.
| | - Almas M R Mahmud
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Ismail M Maulood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
3
|
Kilinc E, Torun IE, Baranoglu Kilinc Y, Töre F. Proposed receptor-mediated mechanisms of melatonin in nitroglycerin-induced migraine-like hyperalgesic conditions in rats. J Nutr Biochem 2025; 136:109800. [PMID: 39537039 DOI: 10.1016/j.jnutbio.2024.109800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Melatonin has a therapeutic effect on migraine, but the mechanisms underlying its antimigraine effect have not been elucidated. This study therefore investigated for the first time the receptor-mediated mechanisms of action of melatonin in nitroglycerin (NTG)- induced migraine-like hyperalgesic conditions in rats. Melatonin, nonselective MT1/MT2 antagonist luzindole, selective MT2 antagonist DH97 or potent MT3 antagonist prazosin, alone or in various combinations, were administered to NTG-induced migraine rats and ex-vivo meningeal preparations. Basal and drug-treated pain behaviors were assessed with the von-Frey test. CGRP levels in the trigeminal ganglia, trigeminal nucleus caudalis (TNC) and ex-vivo superfusate medium, as well as c-fos level in the TNC, were measured by ELISA. Meningeal mast cells were stained with toluidine-blue and examined histologically for their activation and count. Melatonin mitigated mechanical hyperalgesia, and c-fos and CGRP expression in the TNC, CGRP expression in trigeminal ganglia, CGRP release from meningeal afferents, all of which were induced by NTG, and also suppressed NTG-stimulated meningeal mast cell activation. The effects of melatonin were abolished in the presence of luzindole and DH97, respectively. However, prazosin did not reverse the effects of melatonin except for mechanical hyperalgesia. Luzindole and DH97 in combinations with prazosin also canceled the effects of melatonin, respectively, other than CGRP expression in the TNC. Melatonin exerts its anti-hyperalgesic effects through modulation of trigeminal expression and meningeal release of CGRP, and meningeal mast cell activation in experimental migraine-like conditions. The effects of melatonin are mainly mediated by MT2 receptors, without excluding a possible role for MT1.
Collapse
Affiliation(s)
- Erkan Kilinc
- Department of Physiology, Bolu Abant Izzet Baysal University, Medical Faculty, Bolu, Türkiye.
| | - Ibrahim Ethem Torun
- Department of Physiology, Bolu Abant Izzet Baysal University, Medical Faculty, Bolu, Türkiye
| | | | - Fatma Töre
- Department of Physiology, Atlas University, Medical Faculty, Istanbul, Türkiye
| |
Collapse
|
4
|
Demas GE, Han Y, Fink HF. Melatonin. Trends Endocrinol Metab 2025; 36:191-192. [PMID: 39084917 PMCID: PMC11779981 DOI: 10.1016/j.tem.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Affiliation(s)
- Gregory E Demas
- Department of Biology and Program in Animal Behavior, Indiana University, Bloomington, IN 47405, USA.
| | - Yuqi Han
- Department of Biology and Program in Animal Behavior, Indiana University, Bloomington, IN 47405, USA
| | - Hannah F Fink
- Department of Biology and Program in Animal Behavior, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
5
|
Tian Z, Zhang Q, Wang L, Li M, Li T, Wang Y, Cao Z, Jiang X, Luo P. Progress in the mechanisms of pain associated with neurodegenerative diseases. Ageing Res Rev 2024; 102:102579. [PMID: 39542176 DOI: 10.1016/j.arr.2024.102579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Neurodegenerative diseases (NDDs) represent a class of neurological disorders characterized by the progressive degeneration or loss of neurons, impacting millions of individuals globally. In addition to the typical manifestations, pain is a prevalent symptom associated with NDDs, seriously impacting the quality of life for patients. The pathogenesis of pain associated with NDDs is intricate and multifaceted. Currently, the clinical management of NDDs-related pain symptoms predominantly relies on conventional pharmacological agents or physical therapy. However, these approaches often fail to produce satisfactory outcomes. This article summarizes the underlying mechanisms of major NDDs-associated pain: Neuroinflammation, Brain and spinal cord dysfunctions, Mitochondrial dysfunction, Risk gene and pathological protein, as well as Receptor, channel, and neurotransmitter. While numerous studies have investigated the downstream pathological processes associated with these mechanisms, there remains a significant gap in identifying the key initiating factors. Specifically, there is insufficient evidence for the upstream elements that activate microglia and astrocytes in neuroinflammation leading to pain in NDDs. Likewise, there is an absence of upstream factors elucidating how dysfunctions in the brain and spinal cord, as well as mitochondrial impairments, contribute to the development of pain. Furthermore, the specific mechanisms through which hallmark pathological proteins related to NDDs contribute to these pathological processes remain inadequately understood. The objective of this article is to synthesize the existing mechanisms underlying pain associated with NDDs, including Alzheimer's disease, Parkinson's disease, Huntington's disease, Schizophrenia, Amyotrophic lateral sclerosis, and Multiple sclerosis, while also identifying gaps and deficiencies in these mechanisms. This paper offers insights for future research trajectories. Given the intricate pathogenesis of NDDs-related pain, it emphasizes that a promising short-term strategy is combination therapy-intervening concurrently in multiple pathological processes-akin to the cocktail approach utilized in treating acquired immunodeficiency syndrome (AIDS). For long-term advancements, achieving breakthroughs in the treatment of the NDDs themselves will remain essential for alleviating accompanying pain symptoms.
Collapse
Affiliation(s)
- Zhicheng Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Qi Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; The Fifth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ling Wang
- Xi'an Children's Hospital, Xi'an 710002, China
| | - Mengxiang Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; The Fifth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Tianjing Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yujie Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Zixuan Cao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; The Sixth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
6
|
Muñoz-Jurado A, Escribano BM. Presence of melatonin in foods of daily consumption: The benefit of this hormone for health. Food Chem 2024; 458:140172. [PMID: 38943958 DOI: 10.1016/j.foodchem.2024.140172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024]
Abstract
Melatonin (MLT) is a hormone that exists in all living organisms, including bacteria, yeast, fungi, animals, and plants, many of which are ingested daily in the diet. However, the exact concentrations of melatonin in each of the foods and the effect on health of the intake of foods rich in MLT are not known. Therefore, the aim of this review was to gather the available information on the melatonin content of different foods and to evaluate the effect that this hormone has on different pathologies. The amount of MLT may vary depending on the variety, origin, heat treatment, processing, and analysis technique, among other factors. Dietary interventions with foods rich in MLT report health benefits, but there is no evidence that hormone is partially responsible for the clinical improvement. Therefore, it is necessary to evaluate the MLT content in more foods, as well as the effect that cooking/processing has on the amount of MLT, to estimate its total intake in a typical diet and better explore its potential impact on the health.
Collapse
Affiliation(s)
- Ana Muñoz-Jurado
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Spain.; Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBIC), Cordoba, Spain..
| | - Begoña M Escribano
- Department of Cell Biology, Physiology and Immunology, Faculty of Veterinary Medicine, University of Cordoba, Spain.; Maimonides Institute for Research in Biomedicine of Cordoba, (IMIBIC), Cordoba, Spain..
| |
Collapse
|
7
|
Kuzmenko NV, Tsyrlin VA, Pliss MG. [Meta-analysis of experimental studies of the effect of melatonin monotherapy on the levels of thyroid hormones and glucocorticoids in rats kept under standard condition]. PROBLEMY ENDOKRINOLOGII 2024; 70:91-105. [PMID: 39509641 DOI: 10.14341/probl13396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 11/15/2024]
Abstract
BACKGROUND Melatonin is known to modulate circadian and seasonal rhythms in metabolism, reproduction, and behavior. However, the effect of exogenous melatonin supplementation on the functioning of the thyroid and adrenal glands in species without a clear seasonality in reproduction is still unclear. AIM Using a meta-analysis of publications, to investigate the effect of melatonin monotherapy on the concentrations of pituitary thyroid-stimulating hormone, thyroid hormones (TG), pituitary adrenocorticotropic hormone and corticosterone (CS) in rats kept under standard laboratory conditions. MATERIALS AND METHODS In our work, using the Review Manager 5.3 program, we conducted a meta-analysis of publications examining the effect of melatonin monotherapy on the functioning of the thyroid gland (22 papers) and adrenal glands (20 papers) in rats kept under standard conditions. RESULTS According to the results of our meta-analysis, the effects of melatonin on the levels of TG and CS depend on the dose and duration of therapy. A decrease in TG and CS was associated with therapy lasting no more than 4-5 weeks and with high doses of melatonin. An increase in CS and a trend toward increased TG levels were observed with longer therapy. However, a few studies have observed a decrease in TG with very long-term melatonin therapy (≥32 weeks). Among all TGs, total thyroxine (T4) showed maximum sensitivity to exogenous melatonin, which indicates the influence of melatonin on the secretory function of the thyroid gland. In addition, melatonin increased the relative weight of the adrenal glands. There was no convincing evidence that the effects of melatonin were influenced by the route and timing of administration, or the timing of blood sampling. CONCLUSION As a result, exogenous melatonin can modulate TG and CS levels, even in species without a clear seasonality in reproductive function.
Collapse
Affiliation(s)
- N V Kuzmenko
- Almazov National Medical Research Centre; First Pavlov State Medical University of St. Petersburg
| | | | - M G Pliss
- Almazov National Medical Research Centre
| |
Collapse
|
8
|
Loonen AJM. The putative role of the habenula in animal migration. Physiol Behav 2024; 286:114668. [PMID: 39151652 DOI: 10.1016/j.physbeh.2024.114668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/26/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND When an addicted animal seeks a specific substance, it is based on the perception of internal and external cues that strongly motivate to pursue the acquisition of that compound. In essence, a similar process acts out when an animal leaves its present area to begin its circannual migration. This review article examines the existence of scientific evidence for possible relatedness of migration and addiction by influencing Dorsal Diencephalic Conduction System (DDCS) including the habenula. METHODS For this review especially the databases of Pubmed and Embase were frequently and non-systematically searched. RESULTS The mechanisms of bird migration have been thoroughly investigated. Especially the mechanism of the circannual biorhythm and its associated endocrine regulation has been well elucidated. A typical behavior called "Zugunruhe" marks the moment of leaving in migratory birds. The role of magnetoreception in navigation has also been clarified in recent years. However, how bird migration is regulated at the neuronal level in the forebrain is not well understood. Among mammals, marine mammals are most similar to birds. They use terrestrial magnetic field when navigating and often bridge long distances between breeding and foraging areas. Population migration is further often seen among the large hoofed mammals in different parts of the world. Importantly, learning processes and social interactions with conspecifics play a major role in these ungulates. Considering the evolutionary development of the forebrain in vertebrates, it can be postulated that the DDCS plays a central role in regulating the readiness and intensity of essential (emotional) behaviors. There is manifold evidence that this DDCS plays an important role in relapse to abuse after prolonged periods of abstinence from addictive behavior. It is also possible that the DDCS plays a role in navigation. CONCLUSIONS The role of the DDCS in the neurobiological regulation of bird migration has hardly been investigated. The involvement of this system in relapse to addiction in mammals might suggest to change this. It is recommended that particularly during "Zugunruhe" the role of neuronal regulation via the DDCS will be further investigated.
Collapse
Affiliation(s)
- Anton J M Loonen
- Pharmacotherapy, Epidemiology & Economics, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands.
| |
Collapse
|
9
|
Fujimoto M, Sekiyama H, Nakamoto H, Takata J, Sawamura S. Changes of melatonin secretion in the neuropathic pain induced sleep disorder model rat. Sleep Biol Rhythms 2024; 22:463-470. [PMID: 39300980 PMCID: PMC11408445 DOI: 10.1007/s41105-024-00529-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/01/2024] [Indexed: 09/22/2024]
Abstract
Chronic pain due to peripheral neuropathy can lead to sleep disorders that significantly worsen the patient's quality of life. Previously, we conducted brain wave measurements in a rat model of neuropathic pain and identified its potential as a model for sleep disorders associated with chronic pain (reported). In this study, we quantified melatonin secretion and assessed its circadian rhythm in a rat model of pain-induced sleep disorder. To create a model of chronic constriction injury (CCI), rats were loosely tied around the sciatic nerve, with approximately 1 mm spacing, 14 days before the experiment. Rats with no ties around the sciatic nerve were used as controls. Electroencephalograms and electromyograms were recorded for 3 days, and the episodes of waking, REM sleep, and non-REM sleep were compared between the groups. The samples for microanalysis were collected every 30 min and used for melatonin analysis. Compared to the control group, the CCI model group exhibited an increase in wake episodes and a decrease in non-REM sleep episodes. Analysis of the area under the curve of melatonin secretion revealed a significant increase in melatonin secretion and a loss of circadian rhythm in the CCI model group. Melatonin secretion markedly increased accompanied by loss of circadian rhythm in a rat model of CCI. Further studies investigating the causal relationship between neuropathic pain and melatonin secretion are warranted.
Collapse
Affiliation(s)
- Moe Fujimoto
- Department of Anesthesiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605 Japan
| | - Hiroshi Sekiyama
- Department of Anesthesiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605 Japan
| | - Hirofumi Nakamoto
- Department of Anesthesiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605 Japan
| | - Junko Takata
- Department of Anesthesiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605 Japan
| | - Shigehito Sawamura
- Department of Anesthesiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-Ku, Tokyo, 173-8605 Japan
| |
Collapse
|
10
|
Cardenas-Padilla AJ, Jimenez-Trejo F, Cerbon M, Chavez-Garcia A, Cruz-Cano NB, Martinez-Torres M, Alcantar-Rodriguez A, Medrano A. Sperm melatonin receptors, seminal plasma melatonin and semen freezability in goats. Theriogenology 2024; 225:98-106. [PMID: 38801791 DOI: 10.1016/j.theriogenology.2024.05.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Goat bucks are seasonal breeders that show variation in sperm quality, endogenous melatonin (MLT), and presumably in the expression of MLT receptors on the sperm throughout the year, which may modify sperm freezability. The aim of this study was to determine whether sperm freezability is associated with (i) endogenous melatonin levels in seminal plasma and (ii) the expression of sperm plasma membrane melatonin receptors (MT1, MT2). To evaluate this, spermatozoa from seven Saanen goat bucks were cryopreserved throughout the year in Mexico using a standard freezing protocol. Seminal plasma MLT concentrations were determined by ELISA and the expression and localization of MT1 and MT2 were detected by immunocytochemistry and confirmed by western blotting. The recovery rate of progressive motility after thawing was higher in spring than autumn and winter; in contrast, the F pattern (CTC assay) was higher in winter than in the other seasons. A proportional increase in the AR pattern (CTC assay) was smaller in winter than in the other seasons and the proportion of sperm showing high plasma membrane fluidity was higher in spring than in summer and autumn. The seminal plasma MLT concentrations showed no significant interseasonal differences. The MT1 receptor was immunolocalised at the apical region of the sperm head, while MT2 was mainly localised in the neck. The relative expression of MLT receptors showed significant differences between summer and winter for all bands, except at 75 kDa of MT2. In conclusion, there was an association between the relative expression of MT1 and MT2 receptors throughout the year and sperm freezability in goat bucks in México. Post-thaw sperm quality is enhanced in semen samples collected during breeding season.
Collapse
Affiliation(s)
- Alberto J Cardenas-Padilla
- Laboratorio de Reproducción Animal, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, 54714, Estado de México, Mexico.
| | | | - Marco Cerbon
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico.
| | - Araceli Chavez-Garcia
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico.
| | - Norma B Cruz-Cano
- Laboratorio de Biología de la Reproducción, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, 54090, Estado de México, Mexico.
| | - Martin Martinez-Torres
- Laboratorio de Biología de la Reproducción, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, 54090, Estado de México, Mexico.
| | - Alicia Alcantar-Rodriguez
- Laboratorio de Reproducción Animal, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, 54714, Estado de México, Mexico.
| | - Alfredo Medrano
- Laboratorio de Reproducción Animal, Unidad de Investigación Multidisciplinaria, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli, 54714, Estado de México, Mexico.
| |
Collapse
|
11
|
Brandt N, Köper F, Hausmann J, Bräuer AU. Spotlight on plasticity-related genes: Current insights in health and disease. Pharmacol Ther 2024; 260:108687. [PMID: 38969308 DOI: 10.1016/j.pharmthera.2024.108687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/07/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
The development of the central nervous system is highly complex, involving numerous developmental processes that must take place with high spatial and temporal precision. This requires a series of complex and well-coordinated molecular processes that are tighly controlled and regulated by, for example, a variety of proteins and lipids. Deregulations in these processes, including genetic mutations, can lead to the most severe maldevelopments. The present review provides an overview of the protein family Plasticity-related genes (PRG1-5), including their role during neuronal differentiation, their molecular interactions, and their participation in various diseases. As these proteins can modulate the function of bioactive lipids, they are able to influence various cellular processes. Furthermore, they are dynamically regulated during development, thus playing an important role in the development and function of synapses. First studies, conducted not only in mouse experiments but also in humans, revealed that mutations or dysregulations of these proteins lead to changes in lipid metabolism, resulting in severe neurological deficits. In recent years, as more and more studies have shown their involvement in a broad range of diseases, the complexity and broad spectrum of known and as yet unknown interactions between PRGs, lipids, and proteins make them a promising and interesting group of potential novel therapeutic targets.
Collapse
Affiliation(s)
- Nicola Brandt
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Franziska Köper
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Jens Hausmann
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Anja U Bräuer
- Research Group Anatomy, Department of Human Medicine, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany; Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
| |
Collapse
|
12
|
Megha KB, Arathi A, Shikha S, Alka R, Ramya P, Mohanan PV. Significance of Melatonin in the Regulation of Circadian Rhythms and Disease Management. Mol Neurobiol 2024; 61:5541-5571. [PMID: 38206471 DOI: 10.1007/s12035-024-03915-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
Melatonin, the 'hormone of darkness' is a neuronal hormone secreted by the pineal gland and other extra pineal sites. Responsible for the circadian rhythm and seasonal behaviour of vertebrates and mammals, melatonin is responsible for regulating various physiological conditions and the maintenance of sleep, body weight and the neuronal activities of the ocular sites. With its unique amphiphilic structure, melatonin can cross the cellular barriers and elucidate its activities in the subcellular components, including mitochondria. Melatonin is a potential scavenger of oxygen and nitrogen-reactive species and can directly obliterate the ROS and RNS by a receptor-independent mechanism. It can also regulate the pro- and anti-inflammatory cytokines in various pathological conditions and exhibit therapeutic activities against neurodegenerative, psychiatric disorders and cancer. Melatonin is also found to show its effects on major organs, particularly the brain, liver and heart, and also imparts a role in the modulation of the immune system. Thus, melatonin is a multifaceted candidate with immense therapeutic potential and is still considered an effective supplement on various therapies. This is primarily due to rectification of aberrant circadian rhythm by improvement of sleep quality associated with risk development of neurodegenerative, cognitive, cardiovascular and other metabolic disorders, thereby enhancing the quality of life.
Collapse
Affiliation(s)
- K B Megha
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, 695 012, Kerala, India
| | - A Arathi
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, 695 012, Kerala, India
| | - Saini Shikha
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
| | - Rao Alka
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Prabhu Ramya
- P.G. Department of Biotechnology, Government Arts College, Trivandrum, 695 014, India
| | - P V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, 695 012, Kerala, India.
| |
Collapse
|
13
|
Smit TH. On growth and scoliosis. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2024; 33:2439-2450. [PMID: 38705903 DOI: 10.1007/s00586-024-08276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/15/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE To describe the physiology of spinal growth in patients with adolescent idiopathic scoliosis (AIS). METHODS Narrative review of the literature with a focus on mechanisms of growth. RESULTS In his landmark publication On Growth and Form, D'Arcy Thompson wrote that the anatomy of an organism reflects the forces it is subjected to. This means that mechanical forces underlie the shape of tissues, organs and organisms, whether healthy or diseased. AIS is called idiopathic because the underlying cause of the deformation is unknown, although many factors are associated. Eventually, however, any deformity is due to mechanical forces. It has long been shown that the typical curvature and rotation of the scoliotic spine could result from vertebrae and intervertebral discs growing faster than the ligaments attached to them. This raises the question why in AIS the ligaments do not keep up with the speed of spinal growth. The spine of an AIS patient deviates from healthy spines in various ways. Growth is later but faster, resulting in higher vertebrae and intervertebral discs. Vertebral bone density is lower, which suggests less spinal compression. This also preserves the notochordal cells and the swelling pressure in the nucleus pulposus. Less spinal compression is due to limited muscular activity, and low muscle mass indeed underlies the lower body mass index (BMI) in AIS patients. Thus, AIS spines grow faster because there is less spinal compression that counteracts the force of growth (Hueter-Volkmann Law). Ligaments consist of collagen fibres that grow by tension, fibrillar sliding and the remodelling of cross-links. Growth and remodelling are enhanced by dynamic loading and by hormones like estrogen. However, they are opposed by static loading. CONCLUSION Increased spinal elongation and reduced ligamental growth result in differential strain and a vicious circle of scoliotic deformation. Recognising the physical and biological cues that contribute to differential growth allows earlier diagnosis of AIS and prevention in children at risk.
Collapse
Affiliation(s)
- Theodoor H Smit
- Department of Orthopaedic Surgery and Sports Medicine, Amsterdam University Medical Centres, Amsterdam Movement Sciences, Amsterdam, The Netherlands.
- Department of Medical Biology, Amsterdam University Medical Centres, Meibergdreef 9, Room K2-140, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Yehia A, Abulseoud OA. Melatonin: a ferroptosis inhibitor with potential therapeutic efficacy for the post-COVID-19 trajectory of accelerated brain aging and neurodegeneration. Mol Neurodegener 2024; 19:36. [PMID: 38641847 PMCID: PMC11031980 DOI: 10.1186/s13024-024-00728-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024] Open
Abstract
The unprecedented pandemic of COVID-19 swept millions of lives in a short period, yet its menace continues among its survivors in the form of post-COVID syndrome. An exponentially growing number of COVID-19 survivors suffer from cognitive impairment, with compelling evidence of a trajectory of accelerated aging and neurodegeneration. The novel and enigmatic nature of this yet-to-unfold pathology demands extensive research seeking answers for both the molecular underpinnings and potential therapeutic targets. Ferroptosis, an iron-dependent cell death, is a strongly proposed underlying mechanism in post-COVID-19 aging and neurodegeneration discourse. COVID-19 incites neuroinflammation, iron dysregulation, reactive oxygen species (ROS) accumulation, antioxidant system repression, renin-angiotensin system (RAS) disruption, and clock gene alteration. These events pave the way for ferroptosis, which shows its signature in COVID-19, premature aging, and neurodegenerative disorders. In the search for a treatment, melatonin shines as a promising ferroptosis inhibitor with its repeatedly reported safety and tolerability. According to various studies, melatonin has proven efficacy in attenuating the severity of certain COVID-19 manifestations, validating its reputation as an anti-viral compound. Melatonin has well-documented anti-aging properties and combating neurodegenerative-related pathologies. Melatonin can block the leading events of ferroptosis since it is an efficient anti-inflammatory, iron chelator, antioxidant, angiotensin II antagonist, and clock gene regulator. Therefore, we propose ferroptosis as the culprit behind the post-COVID-19 trajectory of aging and neurodegeneration and melatonin, a well-fitting ferroptosis inhibitor, as a potential treatment.
Collapse
Affiliation(s)
- Asmaa Yehia
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ, 58054, USA
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Osama A Abulseoud
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ, 58054, USA.
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, 5777 E Mayo Blvd, Phoenix, AZ, 85054, USA.
| |
Collapse
|
15
|
Qu S, Wang J, Guan X, Song C, Wang Y. Sleep disturbance in Angelman syndrome patients. Orphanet J Rare Dis 2024; 19:146. [PMID: 38580983 PMCID: PMC10996173 DOI: 10.1186/s13023-024-03154-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder caused by abnormal expression of the maternal ubiquitin protein ligase E3A gene (UBE3A). As one of the most challenging symptoms and important focuses of new treatment, sleep disturbance is reported to occur in 70-80% of patients with AS and has a serious impact on the lives of patients and their families. Although clinical studies and animal model studies have provided some clues, recent research into sleep disorders in the context of AS is still very limited. It is generally accepted that there is an interaction between neurodevelopment and sleep; however, there is no recognized mechanism for sleep disorders in AS patients. Accordingly, there are no aetiologically specific clinical treatments for AS-related sleep disorders. The most common approaches involve ameliorating symptoms through methods such as behavioural therapy and symptomatic pharmacotherapy. In recent years, preclinical and clinical studies on the targeted treatment of AS have emerged. Although precision therapy for restoring the UBE3A level and the function of its signalling pathways is inevitably hindered by many remaining obstacles, this approach has the potential to address AS-related sleep disturbance.
Collapse
Affiliation(s)
- Song Qu
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junyi Wang
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xingying Guan
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, China
| | - Cui Song
- Department of Endocrinology and Genetic Metabolism Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| | - Yanyan Wang
- Department of Medical Genetics, College of Basic Medical Science, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
16
|
Rong Y, Ma R, Zhang Y, Guo Z. Melatonin's effect on hair follicles in a goat ( Capra hircus) animal model. Front Endocrinol (Lausanne) 2024; 15:1361100. [PMID: 38628581 PMCID: PMC11018883 DOI: 10.3389/fendo.2024.1361100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction Melatonin can treat androgenetic alopecia in males. Goats can be used as animal models to study melatonin treatment for human alopecia. In this study, a meta-analysis of melatonin's effects on goat hair follicles was pursued. Methods Literature from the last 20 years was searched in Scopus, Science Direct, Web of Science and PubMed. Melatonin's effect on goat hair follicles and litter size were performed through a traditional meta-analysis and trial sequential analysis. A network meta-analysis used data from oocyte development to blastocyst. The hair follicle genes regulated by melatonin performed KEGG and PPI. We hypothesized that there are differences in melatonin receptors between different goats, and therefore completed melatonin receptor 1A homology modelling and molecular docking. Results The results showed that melatonin did not affect goat primary follicle or litter size. However, there was a positive correlation with secondary follicle growth. The goat melatonin receptor 1A SNPs influence melatonin's functioning. The wild type gene defect MR1 is a very valuable animal model. Discussion Future studies should focus on the relationship between goat SNPs and the effect of embedded melatonin. This study will provide theoretical guidance for the cashmere industry and will be informative for human alopecia research.
Collapse
Affiliation(s)
- Youjun Rong
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Rong Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yanjun Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
- Northern Agriculture and Livestock Husbandry Technical Innovation Center, Chinese Academy of Agricultural Sciences, Hohhot, China
| | - Zhenhua Guo
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
17
|
Liu Z, Zhu J, Shen Z, Ling Y, Zeng Y, Yang Y, Jiang G. Melatonin as an add-on treatment for epilepsy: A systematic review and meta-analysis. Seizure 2024; 117:133-141. [PMID: 38417212 DOI: 10.1016/j.seizure.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/17/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024] Open
Abstract
PURPOSE Epilepsy, one severe prevalent brain disorder, primarily relies on drug treatment. However, approximately one-third of patients with epilepsy do not achieve effective control with current medications, underscoring the need for more innovative treatment approaches. Notably, melatonin has gained attention for its anti-seizure properties and favourable safety profile. This systematic review aimed to evaluate the efficacy and safety of melatonin as an add-on treatment for epilepsy. METHODS We searched for articles published before June 2023 in Web of Science, Cochrane Library, and PubMed. We used RevMan 5.4 software to compute relative risks (RRs) and 95 % confidence intervals (CIs). Key outcomes included total sleep time, wakefulness after sleep onset, sleep latency, seizure frequency, seizure severity, and safety. The quality of randomised controlled studies (RCTs) was assessed using the Cochrane Risk of Bias tool. RESULTS Of the 264 publications retrieved, 10 RCTs were included in the meta-analysis. Add-on melatonin treatment improved sleep latency (RR: 0.56; 95 %CI: 0.10-1.02; P = 0.02) and seizure severity (RR: 0.33; 95 %CI: 0.04-0.62; P = 0.03) compared with placebo treatment. Adverse events (increased headache severity in children with a history of migraines, bronchitis, ear infections, agitation, and urinary frequency) were reported in only one trial. CONCLUSION This systematic review found that add-on melatonin therapy improved sleep latency and seizure severity in patients with epilepsy. However, several of the included studies did not systematically assess sleep quality, seizures, and safety and lacked long-term follow-up data. Further RCTs with extended follow-up periods are required to definitively determine the efficacy and safety of melatonin.
Collapse
Affiliation(s)
- Zhifan Liu
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Institute of neurological diseases, North Sichuan Medical College, Nanchong 637000, Sichuan, PR China
| | - Jie Zhu
- Chongqing Emergency Medical Center and Chongqing University Central Hospital, Chongqing 400042, PR China
| | - Ziyi Shen
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Institute of neurological diseases, North Sichuan Medical College, Nanchong 637000, Sichuan, PR China
| | - Yuanyuan Ling
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Institute of neurological diseases, North Sichuan Medical College, Nanchong 637000, Sichuan, PR China
| | - Yumei Zeng
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Institute of neurological diseases, North Sichuan Medical College, Nanchong 637000, Sichuan, PR China
| | - Yang Yang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Institute of neurological diseases, North Sichuan Medical College, Nanchong 637000, Sichuan, PR China
| | - Guohui Jiang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Institute of neurological diseases, North Sichuan Medical College, Nanchong 637000, Sichuan, PR China.
| |
Collapse
|
18
|
Klosen P. Thirty-seven years of MT1 and MT2 melatonin receptor localization in the brain: Past and future challenges. J Pineal Res 2024; 76:e12955. [PMID: 38606787 DOI: 10.1111/jpi.12955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/21/2024] [Accepted: 03/31/2024] [Indexed: 04/13/2024]
Abstract
Identifying the target cells of a hormone is a key step in understanding its function. Once the molecular nature of the receptors for a hormone has been established, researchers can use several techniques to detect these receptors. Here I will review the different tools used over the years to localize melatonin receptors and the problems associated with each of these techniques. The radioligand 2-[125I] iodomelatonin was the first tool to allow localization of melatonin receptors on tissue sections. Once the MT1 and MT2 receptors were cloned, in situ hybridization could be used to detect the messenger RNA for these receptors. The deduced amino acid sequences for MT1 and MT2 receptors allowed the production of peptide immunogens to generate antibodies against the MT1 and MT2 receptors. Finally, transgenic reporters driven by the promoter elements of the MT1 and MT2 genes have been used to map the expression of MT1 and MT2 in the brain and the retina. Several issues have complicated the localization of melatonin receptors and the characterization of melatonin target cells over the last three decades. Melatonin receptors are expressed at low levels, leading to sensitivity issues for their detection. The second problem are specificity issues with antibodies directed against the MT1 and MT2 melatonin receptors. These receptors are G protein-coupled receptors and many antibodies directed against such receptors have been shown to present similar problems concerning their specificity. Despite these specificity problems which start to be seriously addressed by recent studies, antibodies will be important tools in the future to identify and phenotype melatonin target cells. However, we will have to be more stringent than previously when establishing their specificity. The results obtained by these antibodies will have to be confronted and be coherent with results obtained by other techniques.
Collapse
Affiliation(s)
- Paul Klosen
- Regulation and Disruption of Neuroendocrine Rhythms, Institute of Cellular and Integrative Neurosciences, INCI CNRS UPR-3212, University of Strasbourg, Strasbourg, France
| |
Collapse
|
19
|
Bódizs R, Schneider B, Ujma PP, Horváth CG, Dresler M, Rosenblum Y. Fundamentals of sleep regulation: Model and benchmark values for fractal and oscillatory neurodynamics. Prog Neurobiol 2024; 234:102589. [PMID: 38458483 DOI: 10.1016/j.pneurobio.2024.102589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Homeostatic, circadian and ultradian mechanisms play crucial roles in the regulation of sleep. Evidence suggests that ratios of low-to-high frequency power in the electroencephalogram (EEG) spectrum indicate the instantaneous level of sleep pressure, influenced by factors such as individual sleep-wake history, current sleep stage, age-related differences and brain topography characteristics. These effects are well captured and reflected in the spectral exponent, a composite measure of the constant low-to-high frequency ratio in the periodogram, which is scale-free and exhibits lower interindividual variability compared to slow wave activity, potentially serving as a suitable standardization and reference measure. Here we propose an index of sleep homeostasis based on the spectral exponent, reflecting the level of membrane hyperpolarization and/or network bistability in the central nervous system in humans. In addition, we advance the idea that the U-shaped overnight deceleration of oscillatory slow and fast sleep spindle frequencies marks the biological night, providing somnologists with an EEG-index of circadian sleep regulation. Evidence supporting this assertion comes from studies based on sleep replacement, forced desynchrony protocols and high-resolution analyses of sleep spindles. Finally, ultradian sleep regulatory mechanisms are indicated by the recurrent, abrupt shifts in dominant oscillatory frequencies, with spindle ranges signifying non-rapid eye movement and non-spindle oscillations - rapid eye movement phases of the sleep cycles. Reconsidering the indicators of fundamental sleep regulatory processes in the framework of the new Fractal and Oscillatory Adjustment Model (FOAM) offers an appealing opportunity to bridge the gap between the two-process model of sleep regulation and clinical somnology.
Collapse
Affiliation(s)
- Róbert Bódizs
- Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary.
| | - Bence Schneider
- Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary
| | - Péter P Ujma
- Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary
| | - Csenge G Horváth
- Institute of Behavioural Sciences, Semmelweis University, Budapest, Hungary
| | - Martin Dresler
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands
| | - Yevgenia Rosenblum
- Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands
| |
Collapse
|
20
|
Zhang C, Ma Y, Zhao Y, Guo N, Han C, Wu Q, Mu C, Zhang Y, Tan S, Zhang J, Liu X. Systematic review of melatonin in cerebral ischemia-reperfusion injury: critical role and therapeutic opportunities. Front Pharmacol 2024; 15:1356112. [PMID: 38375039 PMCID: PMC10875093 DOI: 10.3389/fphar.2024.1356112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Cerebral ischemia-reperfusion (I/R) injury is the predominant causes for the poor prognosis of ischemic stroke patients after reperfusion therapy. Currently, potent therapeutic interventions for cerebral I/R injury are still very limited. Melatonin, an endogenous hormone, was found to be valid in preventing I/R injury in a variety of organs. However, a systematic review covering all neuroprotective effects of melatonin in cerebral I/R injury has not been reported yet. Thus, we perform a comprehensive overview of the influence of melatonin on cerebral I/R injury by collecting all available literature exploring the latent effect of melatonin on cerebral I/R injury as well as ischemic stroke. In this systematic review, we outline the extensive scientific studies and summarize the beneficial functions of melatonin, including reducing infarct volume, decreasing brain edema, improving neurological functions and attenuating blood-brain barrier breakdown, as well as its key protective mechanisms on almost every aspect of cerebral I/R injury, including inhibiting oxidative stress, neuroinflammation, apoptosis, excessive autophagy, glutamate excitotoxicity and mitochondrial dysfunction. Subsequently, we also review the predictive and therapeutic implications of melatonin on ischemic stroke reported in clinical studies. We hope that our systematic review can provide the most comprehensive introduction of current advancements on melatonin in cerebral I/R injury and new insights into personalized diagnosis and treatment of ischemic stroke.
Collapse
Affiliation(s)
- Chenguang Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yumei Ma
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yating Zhao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Han
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qian Wu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Changqing Mu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shutong Tan
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jian Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Shenyang, Liaoning, China
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning, China
| | - Xu Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
21
|
Tchekalarova J, Ivanova P, Krushovlieva D, Kortenska L, Angelova VT. Protective Effect of the Novel Melatonin Analogue Containing Donepezil Fragment on Memory Impairment via MT/ERK/CREB Signaling in the Hippocampus in a Rat Model of Pinealectomy and Subsequent Aβ 1-42 Infusion. Int J Mol Sci 2024; 25:1867. [PMID: 38339146 PMCID: PMC10855364 DOI: 10.3390/ijms25031867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
A reduction in melatonin function contributes to the acceleration of Alzheimer's disease (AD), and understanding the molecular processes of melatonin-related signaling is critical for intervention in AD progression. Recently, we synthesized a series of melatonin analogues with donepezil fragments and tested them in silico and in vitro. In this study, one of the most potent compounds, 3c, was evaluated in a rat model of pinealectomy (pin) followed by icvAβ1-42 infusion. Melatonin was used as the reference drug. Treatment with melatonin and 3c (10 mg/kg, i.p. for 14 days) had a beneficial effect on memory decline and the concomitant increase in hippocampal Aβ1-42 and pTAU in the pin+icvAβ1-42 rats. Melatonin supplementation facilitated non-amyloidogenic signaling via non-receptor (histone deacetylase sirtuin 1, SIRT1) and receptor-related signaling (MT/ERK/CREB). The hybrid 3c analogue up-regulated the MT1A and MT2B receptors, pERK and pCREB. Our results strongly support the hypothesis that melatonin-related analogues may become a promising drug candidate for Alzheimer's disease therapy.
Collapse
Affiliation(s)
- Jana Tchekalarova
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.I.); (D.K.); (L.K.)
| | - Petya Ivanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.I.); (D.K.); (L.K.)
| | - Desislava Krushovlieva
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.I.); (D.K.); (L.K.)
| | - Lidia Kortenska
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.I.); (D.K.); (L.K.)
| | - Violina T. Angelova
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria;
| |
Collapse
|
22
|
Shao R, Wang Y, He C, Chen L. Melatonin and its Emerging Physiological Role in Reproduction: A Review and Update. Curr Mol Med 2024; 24:449-456. [PMID: 37070447 DOI: 10.2174/1566524023666230417103201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 04/19/2023]
Abstract
Melatonin is a neuroendocrine hormone secreted by the pineal gland. The secretion of melatonin follows a circadian rhythm controlled by the suprachiasmatic nucleus, and its secretion is synchronized with the changes in light and dark periods in nature, with the highest secretion level at night. Melatonin is a critical hormone that coordinates external light stimulation and cellular responses of the body. It transmits information about the environmental light cycle, including the circadian and seasonal rhythms, to the relevant tissues and organs in the body, which, along with changes in its secretion level, ensures that its regulated functional activities are adapted in response to changes in the outside environment. Melatonin takes beneficial actions mainly through the interaction with specific membrane-bound receptors, termed MT1 and MT2. Melatonin also acts as a scavenger of free radicals via non-receptor-mediated mechanism. For more than half of acentury melatonin has been associated with vertebrate reproduction, especially in the context of seasonal breeding. Though modern humans show little remaining reproductive seasonality, the relationships between melatonin and human reproduction continue to attract extensive attention. Melatonin plays important roles in improving mitochondrial function, reducing the damage of free radicals, inducing oocyte maturation, increasing fertilization rate and promoting embryonic development, which improves the outcomes of in vitro fertilization and embryo transfer. The present article reviews the progress that has been made in our evolving understanding of the physiological role of melatonin in reproduction and its potential clinical applications in reproductive medicine.
Collapse
Affiliation(s)
- Ruifeng Shao
- Reproductive Medicine Center, Jingzhou Hospital affiliated to Yangtze University, No.60 Jingzhong Road, Jingzhou 434020, Hubei, China
| | - Ying Wang
- Reproductive Medicine Center, Jingzhou Hospital affiliated to Yangtze University, No.60 Jingzhong Road, Jingzhou 434020, Hubei, China
| | - Chihua He
- Reproductive Medicine Center, Jingzhou Hospital affiliated to Yangtze University, No.60 Jingzhong Road, Jingzhou 434020, Hubei, China
| | - Ligang Chen
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Jingzhou, No.55 Jianghan North Road, Jingzhou 434021, Hubei, China
| |
Collapse
|
23
|
Feng Y, Jiang X, Liu W, Lu H. The location, physiology, pathology of hippocampus Melatonin MT 2 receptor and MT 2-selective modulators. Eur J Med Chem 2023; 262:115888. [PMID: 37866336 DOI: 10.1016/j.ejmech.2023.115888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Melatonin, a neurohormone secreted by the pineal gland and regulated by the suprachiasmatic nucleus (SCN) of the hypothalamus, is synthesized and directly released into the cerebrospinal fluid (CSF) of the third ventricle (3rdv), where it undergoes rapid absorption by surrounding tissues to exert its physiological function. The hippocampus, a vital structure in the limbic system adjacent to the ventricles, plays a pivotal role in emotional response and memory formation. Melatonin MT1 and MT2 receptors are G protein-coupled receptors (GPCRs) that primarily mediate melatonin's receptor-dependent effects. In comparison to the MT1 receptor, the widely expressed MT2 receptor is crucial for mediating melatonin's biological functions within the hippocampus. Specifically, MT2 receptor is implicated in hippocampal synaptic plasticity and memory processes, as well as neurogenesis and axogenesis. Numerous studies have demonstrated the involvement of MT2 receptors in the pathophysiology and pharmacology of Alzheimer's disease, depression, and epilepsy. This review focuses on the anatomical localization of MT2 receptor in the hippocampus, their physiological function in this region, and their signal transduction and pharmacological roles in neurological disorders. Additionally, we conducted a comprehensive review of MT2 receptor ligands used in psychopharmacology and other MT2-selective ligands over recent years. Ultimately, we provide an outlook on future research for selective MT2 receptor drug candidates.
Collapse
Affiliation(s)
- Yueqin Feng
- Department of Ultrasound, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Xiaowen Jiang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Wenwu Liu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, PR China
| | - Hongyuan Lu
- Department of Clinical Pharmacology, China Medical University, Shenyang, PR China.
| |
Collapse
|
24
|
Turner MJ, Dietz RM. Potential Adjuncts to Therapeutic Hypothermia to Mitigate Multiorgan Injury in Perinatal Hypoxia-Ischemia. Neoreviews 2023; 24:e771-e782. [PMID: 38036441 DOI: 10.1542/neo.24-12-e771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Over the last 2 decades, therapeutic hypothermia has become the standard of care to reduce morbidity and mortality in neonates affected by moderate-to-severe hypoxic-ischemic encephalopathy (HIE). There is a significant interest in improving the neurologic outcomes of neonatal HIE, ranging from adjunctive therapy to therapeutic hypothermia. Importantly, the pathophysiologic mechanisms underlying HIE also affect multiple other organs, contributing to high morbidity and mortality in this patient population. This review focuses on the adjunct therapies currently under investigation to mitigate the impact of hypoxic-ischemic injury on the brain, kidneys, liver, heart, and gastrointestinal system.
Collapse
Affiliation(s)
- Megan J Turner
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Pediatrics, Denver Health Medical Center, Denver, CO
| | - Robert M Dietz
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
25
|
Pilehvari S, Yavangui M, Paknahad E, Cheraghi Z, Ghorbani M. The Boosting Effects of Melatonin on the In Vitro Fertilization (IVF) of Women with Polycystic Ovary Syndrome. Chonnam Med J 2023; 59:188-193. [PMID: 37840679 PMCID: PMC10570865 DOI: 10.4068/cmj.2023.59.3.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 10/17/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most critical disorders, which affects approximately 20% of women of childbearing age and melatonin supplementation in these women can be effective. However, human studies in this area are particularly limited to IVF candidates. The aim of this clinical trial study was to evaluate the effect of melatonin on the in vitro fertilization (IVF) in PCOS involved women. In this clinical trial study, a total of 320 women with PCOS were randomly assigned to the intervention and control groups. Patients in the intervention group (n=160) received a combination of melatonin and metformin (3 mg and 500 mg, respectively) three times a day. The control group (n=160) received metformin 500 mg from the luteal phase of the cycle before the start of gonadotropin. Oocyte and embryo quality, number of oocytes, and pregnancy outcomes were compared in both groups. Our study revealed that the frequency of Metaphase II oocytes (69.9% vs. 57.9%, p<0.001) and the number of embryos of the top-quality (grade A) were higher in the group treated with melatonin (40.3% vs. 29.9%, p=0.001). The rate of clinical pregnancy and implantation were also higher in the intervention group. The odds of clinical pregnancy in the intervention group was 1.8 times (p=0.039). Moreover, oral melatonin supplementation was effective in patients with PCOS, who were candidates for IVF because of the increased quality of mature oocytes, top-quality embryos, and increased odds of clinical pregnancy.
Collapse
Affiliation(s)
- Shamim Pilehvari
- Clinical Research Development Unit of Fatemieh Hospital, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahnaz Yavangui
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Elham Paknahad
- Clinical Research Development Unit of Fatemieh Hospital, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zahra Cheraghi
- Modeling of Non-communicable Diseases Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marzieh Ghorbani
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
26
|
Munley KM, Sinkiewicz DM, Szwed SM, Demas GE. Sex and seasonal differences in neural steroid sensitivity predict territorial aggression in Siberian hamsters. Horm Behav 2023; 154:105390. [PMID: 37354601 PMCID: PMC10527453 DOI: 10.1016/j.yhbeh.2023.105390] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/01/2023] [Accepted: 05/28/2023] [Indexed: 06/26/2023]
Abstract
Many animals display marked changes in physiology and behavior on a seasonal timescale, including non-reproductive social behaviors (e.g., aggression). Previous studies from our lab suggest that the pineal hormone melatonin acts via steroid hormones to regulate seasonal aggression in Siberian hamsters (Phodopus sungorus), a species in which both males and females display increased non-breeding aggression. The neural actions of melatonin on steroids and aggressive behavior, however, are relatively unexplored. Here, we housed male and female hamsters in long-day photoperiods (LDs, characteristic of breeding season) or short-day photoperiods (SDs, characteristic of non-breeding season) and administered timed melatonin (M) or control injections. Following 10 weeks of treatment, we quantified aggressive behavior and neural steroid sensitivity by measuring the relative mRNA expression of two steroidogenic enzymes (aromatase and 5α-reductase 3) and estrogen receptor 1 in brain regions associated with aggression or reproduction [medial preoptic area (MPOA), anterior hypothalamus (AH), arcuate nucleus (ARC), and periaqueductal gray (PAG)] via quantitative PCR. Although LD-M and SD males and females displayed increased aggression and similar changes in gene expression in the ARC, there were sex-specific effects of treatment with melatonin and SDs on gene expression in the MPOA, AH, and PAG. Furthermore, males and females exhibited different relationships between neural gene expression and aggression in response to melatonin and SDs. Collectively, these findings support a role for melatonin in regulating seasonal variation in neural steroid sensitivity and aggression and reveal how distinct neuroendocrine responses may modulate a similar behavioral phenotype in male and female hamsters.
Collapse
Affiliation(s)
- Kathleen M Munley
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA; Department of Psychology, University of Houston, Houston, TX 77204, USA.
| | - David M Sinkiewicz
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA
| | - Sydney M Szwed
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Gregory E Demas
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
27
|
Gong L, Cheng F, Li X, Wang Z, Wang S, Xu R, Zhang B, Xi C. Abnormal functional connectivity in the habenula is associated with subjective hyperarousal state in chronic insomnia disorder. Front Neurol 2023; 14:1119595. [PMID: 37588671 PMCID: PMC10426801 DOI: 10.3389/fneur.2023.1119595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/13/2023] [Indexed: 08/18/2023] Open
Abstract
Background The hyperarousal process model plays a central role in the physiology of chronic insomnia disorder (CID). Recent evidence has demonstrated that the habenula is involved in the arousal and sleep-wake cycle. However, whether the intrinsic habenular functional network contributes to the underlying mechanism of CID and its relationship to the arousal state in CID remains unclear. Methods This single-centered study included 34 patients with subjective CID and 22 matched good sleep control (GSC), and underwent a series of neuropsychological tests and resting-state functional magnetic resonance imaging scans. The habenular functional network was assessed using seed-based functional connectivity (FC) analysis. The subjective arousal state was evaluated with the hyperarousal scale (HAS). Alterations in the habenular FC network and their clinical significance in patients with CID were explored. Results Compared with the GSC group, the CID group showed decreased habenular FC in the left caudate nucleus and right inferior parietal lobule and increased FC in the right habenula, bilateral calcarine cortex, and posterior cingulate cortex. The decreased FC between the left habenula and caudate nucleus was associated with an increased arousal state in the CID group. Conclusion The present results provide evidence for a dysfunctional habenular network in patients with CID. These findings extend our understanding of the neuropathological mechanisms underlying the hyperarousal model in chronic insomnia.
Collapse
Affiliation(s)
- Liang Gong
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Fang Cheng
- Department of Neurology, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xue Li
- Department of Neurology, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhiqi Wang
- Department of Neurology, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shuo Wang
- Department of Neurology, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ronghua Xu
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Bei Zhang
- Department of Neurology, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Chunhua Xi
- Department of Neurology, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
28
|
Nasini S, Tidei S, Shkodra A, De Gregorio D, Cambiaghi M, Comai S. Age-Related Effects of Exogenous Melatonin on Anxiety-like Behavior in C57/B6J Mice. Biomedicines 2023; 11:1705. [PMID: 37371801 DOI: 10.3390/biomedicines11061705] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/02/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
The synthesis of melatonin (MLT) physiologically decreases during aging. Treatment with MLT has shown anxiolytic, hypnotic, and analgesic effects, but little is known about possible age-dependent differences in its efficacy. Therefore, we studied the effects of MLT (20 mg/kg, intraperitoneal) on anxiety-like behavior (open field (OFT), elevated plus maze (EPMT), three-chamber sociability, and marble-burying (MBT) tests), and the medial prefrontal cortex (mPFC)-dorsal hippocampus (dHippo) circuit in adolescent (35-40 days old) and adult (three-five months old) C57BL/6 male mice. MLT did not show any effect in adolescents in the OFT and EPMT. In adults, compared to vehicles, it decreased locomotor activity and time spent in the center of the arena in the OFT and time spent in the open arms in the EPMT. In the MBT, no MLT effects were observed in both age groups. In the three-chamber sociability test, MLT decreased sociability and social novelty in adults, while it increased sociability in adolescents. Using local field potential recordings, we found higher mPFC-dHippo synchronization in the delta and low-theta frequency ranges in adults but not in adolescents after MLT treatment. Here, we show age-dependent differences in the effects of MLT in anxiety paradigms and in the modulation of the mPFC-dHippo circuit, indicating that when investigating the pharmacology of the MLT system, age can significantly impact the study outcomes.
Collapse
Affiliation(s)
- Sofia Nasini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Sara Tidei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Atea Shkodra
- IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- School of Medicine, Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Danilo De Gregorio
- IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- School of Medicine, Vita Salute San Raffaele University, 20132 Milan, Italy
| | - Marco Cambiaghi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| | - Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Department of Psychiatry, McGill University, Montreal, QC H3A 1A1, Canada
| |
Collapse
|
29
|
Kudara M, Kato-Ishikura E, Ikegaya Y, Matsumoto N. Ramelteon administration enhances novel object recognition and spatial working memory in mice. J Pharmacol Sci 2023; 152:128-135. [PMID: 37169477 DOI: 10.1016/j.jphs.2023.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Ramelteon is used to ameliorate sleep disorders that negatively affect memory performance; however, it remains unknown whether ramelteon strengthens neutral memories, which do not involve reward or punishment. To address this, we monitored behavior of mice treated with vehicle/ramelteon while they performed a novel object recognition task and a spontaneous alternation task. Object memory performance in the novel object recognition task was improved only if ramelteon was injected before training, suggesting that ramelteon specifically enhances the acquisition of object recognition memory. Ramelteon also enhanced spatial working memory in the spontaneous alternation task. Altogether, acute ramelteon treatment enhances memory in quasi-natural contexts.
Collapse
Affiliation(s)
- Mikuru Kudara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Eriko Kato-Ishikura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Institute for AI and Beyond, 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
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
30
|
Cardinali DP, Garay A. Melatonin as a Chronobiotic/Cytoprotective Agent in REM Sleep Behavior Disorder. Brain Sci 2023; 13:brainsci13050797. [PMID: 37239269 DOI: 10.3390/brainsci13050797] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Dream-enactment behavior that emerges during episodes of rapid eye movement (REM) sleep without muscle atonia is a parasomnia known as REM sleep behavior disorder (RBD). RBD constitutes a prodromal marker of α-synucleinopathies and serves as one of the best biomarkers available to predict diseases such as Parkinson disease, multiple system atrophy and dementia with Lewy bodies. Most patients showing RBD will convert to an α-synucleinopathy about 10 years after diagnosis. The diagnostic advantage of RBD relies on the prolonged prodromal time, its predictive power and the absence of disease-related treatments that could act as confounders. Therefore, patients with RBD are candidates for neuroprotection trials that delay or prevent conversion to a pathology with abnormal α-synuclein metabolism. The administration of melatonin in doses exhibiting a chronobiotic/hypnotic effect (less than 10 mg daily) is commonly used as a first line treatment (together with clonazepam) of RBD. At a higher dose, melatonin may also be an effective cytoprotector to halt α-synucleinopathy progression. However, allometric conversion doses derived from animal studies (in the 100 mg/day range) are rarely employed clinically regardless of the demonstrated absence of toxicity of melatonin in phase 1 pharmacological studies with doses up to 100 mg in normal volunteers. This review discusses the application of melatonin in RBD: (a) as a symptomatic treatment in RBD; (b) as a possible disease-modifying treatment in α-synucleinopathies. To what degree melatonin has therapeutic efficacy in the prevention of α-synucleinopathies awaits further investigation, in particular multicenter double-blind trials.
Collapse
Affiliation(s)
- Daniel P Cardinali
- CENECON, Faculty of Medical Sciences, University of Buenos Aires, Buenos Aires C1431FWO, Argentina
| | - Arturo Garay
- Unidad de Medicina del Sueño-Sección Neurología, Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno" (CEMIC), Buenos Aires C1431FWO, Argentina
| |
Collapse
|
31
|
de Oliveira Moura Araújo E, Gavioli EC, Holanda VAD, da Silva VC, Oliveira Nunes Messias TB, Dutra LMG, de Oliveira MC, Ramos do Egypto Queiroga RDC, Guerra GCB, Soares JKB. REPEATED DONKEY MILK CONSUMPTION REDUCES ANXIETY-LIKE BEHAVIORS AND BRAIN OXIDATIVE DAMAGE TO LIPIDS IN MICE. Behav Brain Res 2023; 449:114477. [PMID: 37150444 DOI: 10.1016/j.bbr.2023.114477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 05/09/2023]
Abstract
Donkey milk (DM) is a source of bioactive compounds that can benefit neural functioning. In the present study, we investigated the effects of DM consumption on anxiolytic-related, despair-like, locomotion and coordination behaviors, as well as the provision of protection from oxidative damage to lipids and proteins in brain tissues and melatonin plasma levels. To achieve this, male mice orally received DM (4g.kg-1) or vehicle for 18 days. Their behavior was assessed in the following tests: elevated plus maze (EPM), open field and rotarod tests (OF, RR) and forced swimming test (FST). Acute treatments with diazepam (DZP, 1.5mg.kg-1, v.o.), fluoxetine (FLX, 20mg.kg-1, i.p.) and nortriptyline (NTP, 20mg.kg-1, i.p.) were used as positive controls. On the eighteenth day, the animals were euthanized and brain tissue and blood were collected to measure oxidative damage, and melatonin plasma levels. Similar to DZP, repeated DM consumption reduced exploration to open areas in the EPM test. Under our experimental conditions, conventional antidepressants reduced immobility time in the FST, and the benzodiazepine treatment impaired motor coordination in mice. No significant differences in locomotion, motor coordination and despair-related behaviors were observed in the mice treated with DM when assessed in the EPM, OF, RR and FST, respectively. Biochemical assays showed that repeated DM exposition protected against oxidative damage to lipids and increased plasma levels of melatonin. These findings suggest consumption of DM may be a promising food for the treatment of anxiety-related disorders, without depressant effects on the central nervous system.
Collapse
Affiliation(s)
| | - Elaine Cristina Gavioli
- Department of Biophysics and Pharmacology, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Valéria Costa da Silva
- Department of Biophysics and Pharmacology, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | | | - Matheus Cardoso de Oliveira
- Department of Biophysics and Pharmacology, Biosciences Centre, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | | | - Juliana Kessia Barbosa Soares
- Department of Food Engineering, Technology Centre, Federal University of Paraíba, Brazil; Center of Education and Health, Federal University of Campina Grande, Cuité, Brazil
| |
Collapse
|
32
|
Verma AK, Singh S, Rizvi SI. Therapeutic potential of melatonin and its derivatives in aging and neurodegenerative diseases. Biogerontology 2023; 24:183-206. [PMID: 36550377 DOI: 10.1007/s10522-022-10006-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Aging is associated with increasing impairments in brain homeostasis and represents the main risk factor across most neurodegenerative disorders. Melatonin, a neuroendocrine hormone that regulates mammalian chronobiology and endocrine functions is well known for its antioxidant potential, exhibiting both cytoprotective and chronobiotic abilities. Age-related decline of melatonin disrupting mitochondrial homeostasis and cytosolic DNA-mediated inflammatory reactions in neurons is a major contributory factor in the emergence of neurological abnormalities. There is scattered literature on the possible use of melatonin against neurodegenerative mechanisms in the aging process and its associated diseases. We have searched PUBMED with many combinations of key words for available literature spanning two decades. Based on the vast number of experimental papers, we hereby review recent advancements concerning the potential impact of melatonin on cellular redox balance and mitochondrial dynamics in the context of neurodegeneration. Next, we discuss a broader explanation of the involvement of disrupted redox homeostasis in the pathophysiology of age-related diseases and its connection to circadian mechanisms. Our effort may result in the discovery of novel therapeutic approaches. Finally, we summarize the current knowledge on molecular and circadian regulatory mechanisms of melatonin to overcome neurodegenerative diseases (NDDs) such as Alzheimer's, Parkinson's, Huntington's disease, and amyotrophic lateral sclerosis, however, these findings need to be confirmed by larger, well-designed clinical trials. This review is also expected to uncover the associated molecular alterations in the aging brain and explain how melatonin-mediated circadian restoration of neuronal homeodynamics may increase healthy lifespan in age-related NDDs.
Collapse
Affiliation(s)
- Avnish Kumar Verma
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, India
| | - Sandeep Singh
- Biological Psychiatry Laboratory, Hadassah Medical Center - Hebrew University, Jerusalem, Israel
| | - Syed Ibrahim Rizvi
- Department of Biochemistry, University of Allahabad, Allahabad, 211002, India.
| |
Collapse
|
33
|
Yu Q, Guo Q, Jin S, Gao C, Zheng P, Li DP, Wu Y. Melatonin suppresses sympathetic vasomotor tone through enhancing GABAA receptor activity in the hypothalamus. Front Physiol 2023; 14:1166246. [PMID: 37064887 PMCID: PMC10090494 DOI: 10.3389/fphys.2023.1166246] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Introduction: Melatonin (5-methoxy-N-acetyl-tryptamine) is a circadian hormone synthesized and secreted by the pineal gland. In addition to regulating circadian rhythms of many physiological functions, melatonin is involved in regulating autonomic nervous function and blood pressure. Hypothalamus paraventricular nucleus (PVN), receiving melatonin projections from the superchiasmatic nucleus, is a critical brain region to regulate neuroendocrine and cardiovascular function. Here, we determined the synaptic mechanisms involved in the effect of melatonin on the sympathetic outflow and blood pressure.Methods and Results: Microinjection of melatonin into the PVN produced a depressor effect and decreased renal sympathetic nerve activity (RSNA). While microinjection of luzindole, a non-selective melatonin receptor antagonist, into the PVN did not change melatonin-induced sympathoinhibition, GABAA receptor antagonist bicuculline eliminated melatonin-induced sympathoinhibition. Furthermore, melatonin decreased firing rate of retrogradely labeled PVN neurons which project to the rostral ventrolateral medulla (RVLM), an effect was not altered by luzindole but eliminated by bicuculline. Melatonin significantly increased the amplitude of spontaneous and evoked GABAergic inhibitory synaptic currents, as well as GABA-induced currents.Conclusion: These data suggest that melatonin in the PVN suppresses sympathetic vasomotor tone through enhancing GABAA receptor activity. This study provides novel information for understanding the cellular mechanisms involved in the effect of melatonin on regulating blood pressure and sympathetic output.
Collapse
Affiliation(s)
- Qiyao Yu
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
- Office of Academic Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qi Guo
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
- Experimental Center for Teaching, Hebei Medical University, Shijiazhuang, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Chao Gao
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Peiru Zheng
- Department of Medicine, University of Missouri, Columbia, KY, United States
| | - De-Pei Li
- Department of Medicine, University of Missouri, Columbia, KY, United States
| | - Yuming Wu
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, China
- *Correspondence: Yuming Wu,
| |
Collapse
|
34
|
Khan D, Cai N, Zhu W, Li L, Guan M, Pu X, Chen Q. The role of phytomelatonin receptor 1-mediated signaling in plant growth and stress response. FRONTIERS IN PLANT SCIENCE 2023; 14:1142753. [PMID: 36968396 PMCID: PMC10036441 DOI: 10.3389/fpls.2023.1142753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
Phytomelatonin is a pleiotropic signaling molecule that regulates plant growth, development, and stress response. In plant cells, phytomelatonin is synthesized from tryptophan via several consecutive steps that are catalyzed by tryptophan decarboxylase (TDC), tryptamine 5-hydroxylase (T5H), serotonin N-acyltransferase (SNAT), and N-acetylserotonin methyltransferase (ASMT) and/or caffeic acid-3-O-methyltransferase (COMT). Recently, the identification of the phytomelatonin receptor PMTR1 in Arabidopsis has been considered a turning point in plant research, with the function and signal of phytomelatonin emerging as a receptor-based regulatory strategy. In addition, PMTR1 homologs have been identified in several plant species and have been found to regulate seed germination and seedling growth, stomatal closure, leaf senescence, and several stress responses. In this article, we review the recent evidence in our understanding of the PMTR1-mediated regulatory pathways in phytomelatonin signaling under environmental stimuli. Based on structural comparison of the melatonin receptor 1 (MT1) in human and PMTR1 homologs, we propose that the similarity in the three-dimensional structure of the melatonin receptors probably represents a convergent evolution of melatonin recognition in different species.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaojun Pu
- *Correspondence: Xiaojun Pu, ; ; Qi Chen, ;
| | - Qi Chen
- *Correspondence: Xiaojun Pu, ; ; Qi Chen, ;
| |
Collapse
|
35
|
Melatonin Activates Anti-Inflammatory Features in Microglia in a Multicellular Context: Evidence from Organotypic Brain Slices and HMC3 Cells. Biomolecules 2023; 13:biom13020373. [PMID: 36830742 PMCID: PMC9952958 DOI: 10.3390/biom13020373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Melatonin (MEL) is a neurohormone endowed with neuroprotective activity, exerted both directly on neuronal cells and indirectly through modulation of responsive glial cells. In particular, MEL's effects on microglia are receptor-mediated and in part dependent on SIRT1 activation. In the present study, we exploited the highly preserved cytoarchitecture of organotypic brain cultures (OC) to explore the effects of MEL on hippocampal microglia in a 3D context as compared to a single cell type context represented by the human HMC3 cell line. We first evaluated the expression of MEL receptor MT1 and SIRT1 and then investigated MEL action against an inflammatory stimulation with LPS: OCs were cultured for a total of 2 weeks and during this time exposed to 0.1 μg/mL of LPS for 24 h either on day 1 (LPS 1°) or on day 11 (LPS 11°). MEL was added immediately after plating and kept for the entire experiment. Under these conditions, both MEL and LPS induced amoeboid microglia. However, the same round phenotype matched different polarization features. LPS increased the number of nuclear-NF-kB+ round cells and MEL alone or in combination with LPS increased BDNF+ round microglia. In addition, MEL contrasted LPS effects on NF-kB expression. Data from HMC3 microglia confirmed MEL's anti-inflammatory effects against LPS in terms of CASP1 induction and BDNF release, identifying SIRT1 as a mediator. However, no effects were evident for MEL alone on HMC3 microglia. Overall, our results point to the importance of the multicellular context for full MEL activity, especially in a preventive view, and support the use of OCs as a favorable model to explore inflammatory responses.
Collapse
|
36
|
Suofu Y, Jauhari A, Nirmala ES, Mullins WA, Wang X, Li F, Carlisle DL, Friedlander RM. Neuronal melatonin type 1 receptor overexpression promotes M2 microglia polarization in cerebral ischemia/reperfusion-induced injury. Neurosci Lett 2023; 795:137043. [PMID: 36586530 PMCID: PMC9936831 DOI: 10.1016/j.neulet.2022.137043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/09/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
Microglial activation is readily detected following cerebral ischemia/reperfusion-induced injury. Activated microglia polarize into either classic pro-inflammatory M1 or protective M2 microglia following ischemia/reperfusion-induced injury. Melatonin is protective immediately after ischemia/reperfusion-induced brain injury. However, the ability of melatonin to affect longer-term recovery from ischemic/reperfusion-induced injury as well as its ability to modulate microglia/macrophage polarization are unknown. The goal of this study is to understand the impact of melatonin on mice 14 days after injury, as well as to understand how melatonin affects microglial polarization of neuronal MT1 activation following cerebral ischemia/reperfusion. We utilized NSEMT1-GFP transgenic mice which overexpress MT1 (melatonin type 1 receptor) in neurons. Melatonin-treated or vehicle treated wild type and NSEMT1-GFP mice underwent middle cerebral artery occlusion (MCAO)/reperfusion and followed for 14 days. Neuronal MT1 overexpression significantly reduced infarct volumes, improved motor function, and ameliorated weight loss. Additionally, melatonin treatment reduced infarct volume in NSEMT1-GFP mice as compared to untreated wild type, melatonin treated wild type, and untreated NSEMT1-GFP mice. Melatonin improved neurological function and prevented weight loss in NSEMT1-GFP mice compared with melatonin treated wild type mice. Finally, melatonin treatment in combination with MT1 overexpression reduced the numbers of Iba1+/CD16+ M1 microglia and increased the numbers of Iba1+/ CD206+ M2 microglia after ischemic injury. In conclusion, neuronal MT1 mediates melatonin-induced long-term recovery after cerebral ischemia, at least in part, by shifting microglial polarization toward the neuroprotective M2 phenotype.
Collapse
Affiliation(s)
- Yalikun Suofu
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Abhishek Jauhari
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Emilia S Nirmala
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - William A Mullins
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiaomin Wang
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fang Li
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Diane L Carlisle
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Robert M Friedlander
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
37
|
Granado MDJ, Pinato L, Santiago J, Barbalho SM, Parmezzan JEL, Suzuki LM, Cabrini ML, Spressão DRMS, Carneiro de Camargo AL, Guissoni Campos LM. Melatonin receptors and Per1 expression in the inferior olivary nucleus of the Sapajus apella monkey. Front Neurosci 2022; 16:1072772. [PMID: 36605547 PMCID: PMC9809291 DOI: 10.3389/fnins.2022.1072772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Melatonin is a transducer of photic environmental information and participates in the synchronization of various physiological and behavioral phenomena. Melatonin can act directly in several areas of the central nervous system through its membrane receptors coupled to G protein, called MT1 and MT2 receptors. In some structures, such as the retina, hypothalamus and pars tuberalis, the expression of both melatonin receptors shows circadian variations. Melatonin can act in the synchronization of the clock proteins rhythm in these areas. Using the immunohistochemistry technique, we detected the immunoexpression of the melatonin receptors and clock genes clock protein Per1 in the inferior olivary nucleus (ION) of the Sapajus apella monkey at specific times of the light-dark phase. The mapping performed by immunohistochemistry showed expressive immunoreactivity (IR) Per1 with predominance during daytime. Both melatonin receptors were expressed in the ION without a day/night difference. The presence of both melatonin receptors and the Per1 protein in the inferior olivary nucleus can indicate a functional role not only in physiological, as in sleep, anxiety, and circadian rhythm, but also a chronobiotic role in motor control mechanisms.
Collapse
Affiliation(s)
- Marcos Donizete Junior Granado
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Luciana Pinato
- Department of Speech, Language and Hearing Sciences, São Paulo State University (UNESP), Marília, Brazil
| | - Jeferson Santiago
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Sandra Maria Barbalho
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Jessica Ellen Lima Parmezzan
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Lenita Mayumi Suzuki
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Mayara Longui Cabrini
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | | | - Ana Letícia Carneiro de Camargo
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil
| | - Leila Maria Guissoni Campos
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Medical School, University of Marilia (UNIMAR), Marília, Brazil,*Correspondence: Leila Maria Guissoni Campos,
| |
Collapse
|
38
|
Posa L, De Gregorio D, Lopez-Canul M, He Q, Darcq E, Rullo L, Pearl-Dowler L, Luongo L, Candeletti S, Romualdi P, Kieffer BL, Gobbi G. Supraspinal melatonin MT 2 receptor agonism alleviates pain via a neural circuit that recruits mu opioid receptors. J Pineal Res 2022; 73:e12825. [PMID: 35996205 DOI: 10.1111/jpi.12825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022]
Abstract
Melatonin, through its G protein-coupled receptor (GPCR) (MTNR1B gene) MT2 , is implicated in analgesia, but the relationship between MT2 receptors and the opioid system remains elusive. In a model of rodent neuropathic pain (spared nerve injured [SNI]), the selective melatonin MT2 agonist UCM924 reversed the allodynia (a pain response to a non-noxious stimulus), and this effect was nullified by the pharmacological blockade or genetic inactivation of the mu opioid receptor (MOR), but not the delta opioid receptor (DOR). Indeed, SNI MOR, but not DOR knockout mice, did not respond to the antiallodynic effects of the UCM924. Similarly, the nonselective opioid antagonist naloxone and the selective MOR antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP) blocked the effects of UCM924 in SNI rats, but not the DOR antagonist naltrindole (NTI). Electrophysiological recordings in the rostral-ventromedial medulla (RVM) revealed that the typical reduction of the firing activity of pronociceptive ON-cells, and the enhancement of the firing of the antinociceptive OFF-cells, induced by the microinjection of the MT2 agonist UCM924 into the ventrolateral periaqueductal gray (vlPAG) were blocked by MOR, but not DOR, antagonism. Immunohistochemistry studies showed that MT2 receptors are expressed in both excitatory (CaMKIIα+ ) and inhibitory (GAD65+ ) neuronal cell bodies in the vlPAG (~2.16% total), but not RVM. Only 0.20% of vlPAG neurons coexpressed MOR and MT2 receptors. Finally, UCM924 treatment induced an increase in the enkephalin precursor gene (PENK) in the PAG of SNI mice. Collectively, the melatonin MT2 receptor agonism requires MORs to exert its antiallodynic effects, mostly through an interneuronal circuit involving MOR and MT2 receptors.
Collapse
Affiliation(s)
- Luca Posa
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada
- Division of Neuroscience, Vita-Salute San Raffaele University, Italy, Milano
| | - Martha Lopez-Canul
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada
| | - Qianzi He
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada
| | - Emmanuel Darcq
- Department of Psychiatry, School of Medicine, Douglas Hospital Research Center, McGill University, Quebec, Montreal, Canada
| | - Laura Rullo
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Leora Pearl-Dowler
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Brigitte Lina Kieffer
- Department of Psychiatry, School of Medicine, Douglas Hospital Research Center, McGill University, Quebec, Montreal, Canada
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, Quebec, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
- McGill University, Health Center (MUHC), Montreal, Quebec, Canada
| |
Collapse
|
39
|
Molloy EJ, El-Dib M, Juul SE, Benders M, Gonzalez F, Bearer C, Wu YW, Robertson NJ, Hurley T, Branagan A, Michael Cotten C, Tan S, Laptook A, Austin T, Mohammad K, Rogers E, Luyt K, Bonifacio S, Soul JS, Gunn AJ. Neuroprotective therapies in the NICU in term infants: present and future. Pediatr Res 2022:10.1038/s41390-022-02295-2. [PMID: 36195634 PMCID: PMC10070589 DOI: 10.1038/s41390-022-02295-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/09/2022] [Accepted: 08/18/2022] [Indexed: 01/13/2023]
Abstract
Outcomes of neonatal encephalopathy (NE) have improved since the widespread implementation of therapeutic hypothermia (TH) in high-resource settings. While TH for NE in term and near-term infants has proven beneficial, 30-50% of infants with moderate-to-severe NE treated with TH still suffer death or significant impairments. There is therefore a critical need to find additional pharmacological and non-pharmacological interventions that improve the outcomes for these children. There are many potential candidates; however, it is unclear whether these interventions have additional benefits when used with TH. Although primary and delayed (secondary) brain injury starting in the latent phase after HI are major contributors to neurodisability, the very late evolving effects of tertiary brain injury likely require different interventions targeting neurorestoration. Clinical trials of seizure management and neuroprotection bundles are needed, in addition to current trials combining erythropoietin, stem cells, and melatonin with TH. IMPACT: The widespread use of therapeutic hypothermia (TH) in the treatment of neonatal encephalopathy (NE) has reduced the associated morbidity and mortality. However, 30-50% of infants with moderate-to-severe NE treated with TH still suffer death or significant impairments. This review details the pathophysiology of NE along with the evidence for the use of TH and other beneficial neuroprotective strategies used in term infants. We also discuss treatment strategies undergoing evaluation at present as potential adjuvant treatments to TH in NE.
Collapse
Affiliation(s)
- Eleanor J Molloy
- Paediatrics, Trinity College Dublin, Trinity Research in Childhood Centre (TRICC), Dublin, Ireland. .,Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland. .,Neonatology, CHI at Crumlin, Dublin, Ireland. .,Neonatology, Coombe Women's and Infants University Hospital, Dublin, Ireland.
| | - Mohamed El-Dib
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Manon Benders
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Fernando Gonzalez
- Department of Neurology, Division of Child Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Cynthia Bearer
- Division of Neonatology, Department of Pediatrics, Rainbow Babies & Children's Hospital, Cleveland, OH, USA.,Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yvonne W Wu
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Nicola J Robertson
- Institute for Women's Health, University College London, London, UK.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Tim Hurley
- Paediatrics, Trinity College Dublin, Trinity Research in Childhood Centre (TRICC), Dublin, Ireland.,Neonatology, Coombe Women's and Infants University Hospital, Dublin, Ireland
| | - Aoife Branagan
- Paediatrics, Trinity College Dublin, Trinity Research in Childhood Centre (TRICC), Dublin, Ireland.,Neonatology, Coombe Women's and Infants University Hospital, Dublin, Ireland
| | | | - Sidhartha Tan
- Pediatrics, Division of Neonatology, Children's Hospital of Michigan, Detroit, MI, USA.,Wayne State University School of Medicine, Detroit, MI, 12267, USA.,Pediatrics, Division of Neonatology, Central Michigan University, Mount Pleasant, MI, USA
| | - Abbot Laptook
- Department of Pediatrics, Women and Infants Hospital, Brown University, Providence, RI, USA
| | - Topun Austin
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Khorshid Mohammad
- Section of Neonatology, Department of Pediatrics, University of Calgary, Calgary, AB, Canada
| | - Elizabeth Rogers
- Department of Pediatrics, University of California, San Francisco Benioff Children's Hospital, San Francisco, CA, USA
| | - Karen Luyt
- Translational Health Sciences, University of Bristol, Bristol, UK.,Neonatology, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Sonia Bonifacio
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 750 Welch Road, Suite 315, Palo Alto, CA, 94304, USA
| | - Janet S Soul
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alistair J Gunn
- Departments of Physiology and Paediatrics, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | |
Collapse
|
40
|
Huang X, Tanveer M, Min Y, Shabala S. Melatonin as a regulator of plant ionic homeostasis: implications for abiotic stress tolerance. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:5886-5902. [PMID: 35640481 DOI: 10.1093/jxb/erac224] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/23/2022] [Indexed: 06/15/2023]
Abstract
Melatonin is a highly conserved and ubiquitous molecule that operates upstream of a broad array of receptors in animal systems. Since melatonin was discovered in plants in 1995, hundreds of papers have been published revealing its role in plant growth, development, and adaptive responses to the environment. This paper summarizes the current state of knowledge of melatonin's involvement in regulating plant ion homeostasis and abiotic stress tolerance. The major topics covered here are: (i) melatonin's control of H+-ATPase activity and its implication for plant adaptive responses to various abiotic stresses; (ii) regulation of the reactive oxygen species (ROS)-Ca2+ hub by melatonin and its role in stress signaling; and (iii) melatonin's regulation of ionic homeostasis via hormonal cross-talk. We also show that the properties of the melatonin molecule allow its direct scavenging of ROS, thus preventing negative effects of ROS-induced activation of ion channels. The above 'desensitization' may play a critical role in preventing stress-induced K+ loss from the cytosol as well as maintaining basic levels of cytosolic Ca2+ required for optimal cell operation. Future studies should focus on revealing the molecular identity of transporters that could be directly regulated by melatonin and providing a bioinformatic analysis of evolutionary aspects of melatonin sensing and signaling.
Collapse
Affiliation(s)
- Xin Huang
- International Research Center for Environmental Membrane Biology, Foshan University, Foshan, Guangdong, China
| | - Mohsin Tanveer
- Tasmanian Institute of Agriculture, University of Tasmania, Tas, Hobart, Australia
| | - Yu Min
- International Research Center for Environmental Membrane Biology, Foshan University, Foshan, Guangdong, China
| | - Sergey Shabala
- International Research Center for Environmental Membrane Biology, Foshan University, Foshan, Guangdong, China
- Tasmanian Institute of Agriculture, University of Tasmania, Tas, Hobart, Australia
- School of Biological Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
41
|
Sundram S, Malviya R, Awasthi R. Genetic Causes of Alzheimer's Disease and the Neuroprotective Role of Melatonin in its Management. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 22:CNSNDDT-EPUB-126085. [PMID: 36056839 DOI: 10.2174/1871527321666220901125730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/15/2022] [Accepted: 07/16/2022] [Indexed: 06/15/2023]
Abstract
Dementia is a global health concern owing to its complexity, which also poses a great challenge to pharmaceutical scientists and neuroscientists. The global dementia prevalence is approximately 47 million, which may increase by three times by 2050. Alzheimer's disease (AD) is the most common cause of dementia. AD is a severe age-related neurodegenerative disorder characterized by short-term memory loss, aphasia, mood imbalance, and executive function. The etiology of AD is still unknown, and the exact origin of the disease is still under investigation. Aggregation of Amyloid β (Aβ) plaques or neurotoxic Aβo oligomers outside the neuron is the most common cause of AD development. Amyloid precursor protein (APP) processing by β secretase and γ secretase produces abnormal Aβ monomers. This aggregation of Aβ and NFT is promoted by various genes like BACE1, ADAM10, PIN1, GSK-3, APOE, PPARα, etc. Identification of these genes can discover several therapeutic targets that can be useful in studying pathogenesis and underlying treatments. Melatonin modulates the activities of these genes, thereby reducing Aβ production and increasing its clearance. Melatonin also reduces the expression of APP by attenuating cAMP, thereby enhancing the non-amyloidogenic process. Present communication explored and discussed the neuroprotective role of melatonin against Aβ-dependent AD pathogenesis. The manuscript also discussed potential molecular and genetic mechanisms of melatonin in the production and clearance of Aβ that could ameliorate neurotoxicity.
Collapse
Affiliation(s)
- Sonali Sundram
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Sector-125, Noida 201313, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rajendra Awasthi
- Department of Pharmaceutical Sciences, School of Health Science and Technology, University of Petroleum and Energy Studies (UPES), Energy Acres, Bidholi, Via-Prem Nagar, Dehradun - 248 007, Uttarakhand, India
| |
Collapse
|
42
|
Roy TK, Uniyal A, Tiwari V. Multifactorial pathways in burn injury-induced chronic pain: novel targets and their pharmacological modulation. Mol Biol Rep 2022; 49:12121-12132. [PMID: 35842856 DOI: 10.1007/s11033-022-07748-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022]
Abstract
Burn injuries are among the highly prevalent medical conditions worldwide that occur mainly in children, military veterans and victims of fire accidents. It is one of the leading causes of temporary as well as permanent disabilities in patients. Burn injuries are accompanied by pain that persists even after recovery from tissue damage which puts immense pressure on the healthcare system. The pathophysiology of burn pain is poorly understood due to its complex nature and lack of considerable preclinical and clinical shreds of evidence, that creates a substantial barrier to the development of new analgesics. Burns damage the skin layers supplied with nociceptors such as NAV1.7, TRPV1, and TRPA1. Burn injury-mediated co-localization and simultaneous activation of TRPA1 and TRPV1 in nociceptive primary afferent C-fibers which contributes to the development and maintenance of chronic pain. Burn injuries are accompanied by central sensitization, a key feature of pain pathophysiology mainly driven by a series of cascades involving aberrations in the glutamatergic system, microglial activation, release of neuropeptides, cytokines, and chemokines. Activation of p38 mitogen-activated protein kinase, altered endogenous opioid signaling, and distorted genomic expression are other pathophysiological factors responsible for the development and maintenance of burn pain. Here we discuss comprehensive literature on molecular mechanisms of burn pain and potential targets that could be translated into near future therapeutics.
Collapse
Affiliation(s)
- Tapas Kumar Roy
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, 221005, Varanasi, U.P, India
| | - Ankit Uniyal
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, 221005, Varanasi, U.P, India
| | - Vinod Tiwari
- Neuroscience & Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, 221005, Varanasi, U.P, India.
| |
Collapse
|
43
|
Giannos P, Prokopidis K, Forbes SC, Celoch K, Candow DG, Tartar JL. Gene Expression Changes of Murine Cortex Homeostasis in Response to Sleep Deprivation Hint Dysregulated Aging-like Transcriptional Responses. Brain Sci 2022; 12:825. [PMID: 35884632 PMCID: PMC9313387 DOI: 10.3390/brainsci12070825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
Sleep deprivation leads to the deterioration in the physiological functioning of the brain, cognitive decline, and many neurodegenerative diseases, all of which progress with advancing age. Sleep insufficiency and impairments in cognitive function are characterized by progressive neuronal losses in the cerebral cortex. In this study, we analyze gene expression profiles following sleep-deprived murine models and circadian matched controls to identify genes that might underlie cortical homeostasis in response to sleep deprivation. Screening of the literature resulted in three murine (Mus musculus) gene expression datasets (GSE6514, GSE78215, and GSE33491) that included cortical tissue biopsies from mice that are sleep deprived for 6 h (n = 15) and from circadian controls that are left undisturbed (n = 15). Cortical differentially expressed genes are used to construct a network of encoded proteins that are ranked based on their interactome according to 11 topological algorithms. The analysis revealed three genes-NFKBIA, EZR, and SGK1-which exhibited the highest multi-algorithmic topological significance. These genes are strong markers of increased brain inflammation, cytoskeletal aberrations, and glucocorticoid resistance, changes that imply aging-like transcriptional responses during sleep deprivation in the murine cortex. Their potential role as candidate markers of local homeostatic response to sleep loss in the murine cortex warrants further experimental validation.
Collapse
Affiliation(s)
- Panagiotis Giannos
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, South Kensington, London SW7 2AZ, UK
- Society of Meta-Research and Biomedical Innovation, London W12 0BZ, UK;
| | - Konstantinos Prokopidis
- Society of Meta-Research and Biomedical Innovation, London W12 0BZ, UK;
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 3BX, UK
| | - Scott C. Forbes
- Department of Physical Education Studies, Faculty of Education, Brandon University, Brandon, MB R7A 6A9, Canada;
| | - Kamil Celoch
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (K.C.); (J.L.T.)
| | - Darren G. Candow
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, SK S4S 0A2, Canada;
| | - Jaime L. Tartar
- Department of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL 33314, USA; (K.C.); (J.L.T.)
| |
Collapse
|
44
|
Merlo S, Caruso GI, Bonfili L, Spampinato SF, Costantino G, Eleuteri AM, Sortino MA. Microglial polarization differentially affects neuronal vulnerability to the β-amyloid protein: Modulation by melatonin. Biochem Pharmacol 2022; 202:115151. [PMID: 35750198 DOI: 10.1016/j.bcp.2022.115151] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/02/2022]
Abstract
Microglial cells play a central but yet debated role in neuroinflammatory events occurring in Alzheimer's disease (AD). We here explored how microglial features are modulated by melatonin following β-amyloid (Aβ42)-induced activation and examined the cross-talk with Aβ-challenged neuronal cells. Human microglial HMC3 cells were exposed to Aβ42 (200 nM) in the presence of melatonin (MEL; 1 μM) added since the beginning (MELco) or after a 72 h-exposure to Aβ42 (MELpost). In both conditions, MEL favored an anti-inflammatory activation and rescued SIRT1 and BDNF expression/release. Caspase-1 up-regulation and phospho-ERK induction following a prolonged exposure to Aβ42 were prevented by MEL. In addition, MEL partially restored proteasome functionality that was altered by long-term Aβ42 treatment, re-establishing both 20S and 26S chymotrypsin-like activity. Differentiated neuronal-like SH-SY5Y cells were exposed to Aβ42 (200 nM for 24 h) in basal medium or in the presence of conditioned medium (CM) collected from microglia exposed for different times to Aβ42 alone or in combination with MELco or MELpost. Aβ42 significantly reduced pre-synaptic proteins synaptophysin and VAMP2 and mean neuritic length. These effects were prevented by CM from anti-inflammatory microglia (Aβ42 for 6 h), or from MELco and MELpost microglia, but the reduction of neuritic length was not rescued when the SIRT1 inhibitor EX527 was added. In conclusion, our data add to the concept that melatonin shows a promising anti-inflammatory action on microglia that is retained even after pro-inflammatory activation, involving modulation of proteasome function and translating into neuroprotective microglial effects.
Collapse
Affiliation(s)
- Sara Merlo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy.
| | - Grazia Ilaria Caruso
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy.
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, via Gentile III da Varano, 62032 Camerino, MC, Italy.
| | - Simona Federica Spampinato
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via P. Giuria 13, Turin 10125, Italy.
| | - Giuseppe Costantino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy.
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, via Gentile III da Varano, 62032 Camerino, MC, Italy.
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy.
| |
Collapse
|
45
|
Bdair H, Singleton TA, Ross K, Jolly D, Kang MS, Aliaga A, Tuznik M, Kaur T, Yous S, Soucy JP, Massarweh G, Scott PJH, Koeppe R, Spadoni G, Bedini A, Rudko DA, Gobbi G, Benkelfat C, Rosa-Neto P, Brooks AF, Kostikov A. Radiosynthesis and In Vivo Evaluation of Four Positron Emission Tomography Tracer Candidates for Imaging of Melatonin Receptors. ACS Chem Neurosci 2022; 13:1382-1394. [PMID: 35420022 DOI: 10.1021/acschemneuro.1c00678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Melatonin is a neurohormone that modulates several physiological functions in mammals through the activation of melatonin receptor type 1 and 2 (MT1 and MT2). The melatonergic system is an emerging therapeutic target for new pharmacological interventions in the treatment of sleep and mood disorders; thus, imaging tools to further investigate its role in the brain are highly sought-after. We aimed to develop selective radiotracers for in vivo imaging of both MT1 and MT2 by positron emission tomography (PET). We identified four previously reported MT ligands with picomolar affinities to the target based on different scaffolds which were also amenable for radiolabeling with either carbon-11 or fluorine-18. [11C]UCM765, [11C]UCM1014, [18F]3-fluoroagomelatine ([18F]3FAGM), and [18F]fluoroacetamidoagomelatine ([18F]FAAGM) have been synthesized in high radiochemical purity and evaluated in wild-type rats. All four tracers showed moderate to high brain permeability in rats with maximum standardized uptake values (SUVmax of 2.53, 1.75, 3.25, and 4.47, respectively) achieved 1-2 min after tracer administration, followed by a rapid washout from the brain. Several melatonin ligands failed to block the binding of any of the PET tracer candidates, while in some cases, homologous blocking surprisingly resulted in increased brain retention. Two 18F-labeled agomelatine derivatives were brought forward to PET scans in non-human primates and autoradiography on human brain tissues. No specific binding has been detected in blocking studies. To further investigate pharmacokinetic properties of the putative tracers, microsomal stability, plasma protein binding, log D, and membrane bidirectional permeability assays have been conducted. Based on the results, we conclude that the fast first pass metabolism by the enzymes in liver microsomes is the likely reason of the failure of our PET tracer candidates. Nevertheless, we showed that PET imaging can serve as a valuable tool to investigate the brain permeability of new therapeutic compounds targeting the melatonergic system.
Collapse
Affiliation(s)
- Hussein Bdair
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
- McGill University, Department of Psychiatry, Irving Ludmer Psychiatry Research and Training Building, Montreal, Quebec H3A 1A1, Canada
| | - Thomas A. Singleton
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
| | - Karen Ross
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
| | - Dean Jolly
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montreal, Quebec H4H 1R3, Canada
| | - Arturo Aliaga
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montreal, Quebec H4H 1R3, Canada
| | - Marius Tuznik
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
| | - Tanpreet Kaur
- University of Michigan Medical School, Department of Radiology, Ann Arbor, Michigan 48109-5610, United States
| | - Saïd Yous
- University of Lille, Lille Neurosciences and Cognition Research Center, Lille, Hauts-de-France FR 59000, France
| | - Jean-Paul Soucy
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
- Concordia University, PERFORM Centre, Montreal, Québec H4B 1R6, Canada
| | - Gassan Massarweh
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
| | - Peter J. H. Scott
- University of Michigan Medical School, Department of Radiology, Ann Arbor, Michigan 48109-5610, United States
| | - Robert Koeppe
- University of Michigan Medical School, Department of Radiology, Ann Arbor, Michigan 48109-5610, United States
| | - Gilberto Spadoni
- University Carlo Bo, Department Biomolecular Science, Urbino IT 61029, Italy
| | - Annalida Bedini
- University Carlo Bo, Department Biomolecular Science, Urbino IT 61029, Italy
| | - David A. Rudko
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Gabriella Gobbi
- McGill University, Department of Psychiatry, Irving Ludmer Psychiatry Research and Training Building, Montreal, Quebec H3A 1A1, Canada
| | - Chawki Benkelfat
- McGill University, Department of Psychiatry, Irving Ludmer Psychiatry Research and Training Building, Montreal, Quebec H3A 1A1, Canada
| | - Pedro Rosa-Neto
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montreal, Quebec H4H 1R3, Canada
| | - Allen F. Brooks
- University of Michigan Medical School, Department of Radiology, Ann Arbor, Michigan 48109-5610, United States
| | - Alexey Kostikov
- McGill University, McConnell Brain Imaging Centre, Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, Montreal, Quebec H4H 1R3, Canada
- McGill University, Department of Chemistry, Montreal, Quebec H3A 0B8, Canada
| |
Collapse
|
46
|
Jantzie L, Muthukumar S, Kitase Y, Vasan V, Fouda MA, Hamimi S, Burkhardt C, Burton VJ, Gerner G, Scafidi J, Ye X, Northington F, Robinson S. Infantile Cocktail of Erythropoietin and Melatonin Restores Gait in Adult Rats with Preterm Brain Injury. Dev Neurosci 2022; 44:266-276. [PMID: 35358965 PMCID: PMC10066804 DOI: 10.1159/000524394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/11/2022] [Indexed: 11/19/2022] Open
Abstract
Cerebral palsy (CP) is the most common cause of physical disability for children worldwide. Many infants and toddlers are not diagnosed with CP until they fail to achieve obvious motor milestones. Currently, there are no effective pharmacologic interventions available for infants and toddlers to substantially improve their trajectory of neurodevelopment. Because children with CP from preterm birth also exhibit a sustained immune system hyper-reactivity, we hypothesized that neuro-immunomodulation with a regimen of repurposed endogenous neurorestorative medications, erythropoietin (EPO) and melatonin (MLT), could improve this trajectory. Thus, we administered EPO + MLT to rats with CP during human infant-toddler equivalency to determine whether we could influence gait patterns in mature animals. After a prenatal injury on embryonic day 18 (E18) that mimics chorioamnionitis at ∼25 weeks human gestation, rat pups were born and raised with their dam. Beginning on postnatal day 15 (P15), equivalent to human infant ∼1 year, rats were randomized to receive either a regimen of EPO + MLT or vehicle (sterile saline) through P20. Gait was assessed in young adult rats at P30 using computerized digital gait analyses including videography on a treadmill. Results indicate that gait metrics of young adult rats treated with an infantile cocktail of EPO + MLT were restored compared to vehicle-treated rats (p < 0.05) and similar to sham controls. These results provide reassuring evidence that pharmacological interventions may be beneficial to infants and toddlers who are diagnosed with CP well after the traditional neonatal window of intervention.
Collapse
Affiliation(s)
- Lauren Jantzie
- Dept. of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Dept. of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
- Dept. of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Dept. of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD
| | - Sankar Muthukumar
- Dept. of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yuma Kitase
- Dept. of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vikram Vasan
- Dept. of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mohammed A. Fouda
- Dept. of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sarah Hamimi
- Dept. of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Dept. of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Vera Joanna Burton
- Dept. of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Center for Infant Neurodevelopment, Kennedy Krieger Institute, Baltimore, MD
| | - Gwendolyn Gerner
- Center for Infant Neurodevelopment, Kennedy Krieger Institute, Baltimore, MD
| | - Joseph Scafidi
- Dept. of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
- Center for Infant Neurodevelopment, Kennedy Krieger Institute, Baltimore, MD
| | - Xiaobu Ye
- Dept. of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Frances Northington
- Dept. of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shenandoah Robinson
- Dept. of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Dept. of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
- Dept. of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
47
|
Filippova YE, Malishevskaya TN, Kolomeichuk SN, Gubin DG, Vlasova AS. The severity of endothelial dysfunction, oxidative stress, lipid metabolism disorders, decreased elastic properties and tone of peripheral vessels in patients with different POAG course variants, depending on the polymorphism of the genes of the biological clock. RUSSIAN OPHTHALMOLOGICAL JOURNAL 2022; 15:78-88. [DOI: 10.21516/2072-0076-2022-15-1-78-88] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
The purpose is to find out the relations of the vascular endothelium dysfunction, atherosclerotic damage to the vessels of the upper and lower limbs, the state of the antioxidant and prooxidant systems, and the lipid profile in patients with different POAG course — carriers of biological clock gene polymorphism.Materials and methods. 47 patients with advanced POAG underwent an examination that included visometry, biomicroscopy, ophthalmoscopy, perimetry, tonometry, thermometry, as well as determination of lipid metabolism, indicators of oxidative stress and antioxidant system. All patients were also tested for the degree of endothelial dysfunction by reactive hyperemia method, and the elastic properties and vascular wall tone by the volumetric sphygmomanometry method. POAG progression was determined by static automated perimetry and optical coherence tomography. The hospitalized patients were examined for daily melatonin profile and key biological clock genes typed by real-time polymerase chain reaction. The genetic material was taken by buccal scraping. Polymorphic variants were identified for CLOCK rs1801260 3111T/C and MTNR1B genes. 16 patients (S-POAG — 8, A-POAG — 8) were tested for melatonin taken from saliva samples at different times of the day in laboratory conditions with controlled nutrition and lighting using the DLMO protocol.Results. The predictors of glaucoma progression were found to be pronounced endothelial dysfunction and oxidative stress, which contribute to the acceleration of atherogenesis and a decrease in the elastic properties of the vascular wall of peripheral vessels. Clock 3111t-c circadian gene polymorphism was found to be interrelated with lipid metabolism disorders and increased lipid peroxidation in patients with POAG progression. Carriers of the G allele of the MTNR1B gene tend to have a higher level of triglyceride (TG) which grows in the evening hours if glaucoma is progressing. We showed that the phasal and amplitude characteristics of daily melatonin level, rather than its average level, may affect the state of the vascular wall of peripheral vessels in POAG patients.Conclusion. POAG progression is associated with pronounced oxidative stress, weakened antioxidant protection and dyslipoproteinemia. Differences in the lipid profile (dyslipidemia) and the indicators of oxidative stress and antioxidant protection in patients with stable and progressing glaucoma course are mutually related with Clock 3111t-c gene polymorphism (CG genotype).
Collapse
Affiliation(s)
| | | | - S. N. Kolomeichuk
- Tyumen State Medical University; Karelian Scientific Center of the Russian Academy of Sciences
| | - D. G. Gubin
- Tyumen State Medical University; Tyumen Cardiology Research Center, branch of the Tomsk Medical Research Center of the Russian Academy of Sciences
| | - A. S. Vlasova
- West Siberian Institute of Postgraduate Medical Education; Regional Ocular Health Clinic
| |
Collapse
|
48
|
Zisapel N. Assessing the potential for drug interactions and long term safety of melatonin for the treatment of insomnia in children with autism spectrum disorder. Expert Rev Clin Pharmacol 2022; 15:175-185. [PMID: 35285365 DOI: 10.1080/17512433.2022.2053520] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Melatonin preparations are emerging first-line pharmacotherapy for insomnia in children and adolescents with autism spectrum disorder (ASD), but quality, formulation, consistency, dosing, and limited long-term safety data are of concern. The recent approval of pediatric-appropriate prolonged-release melatonin (Ped-PRM) addresses these aspects. AREAS COVERED A systematic search of PubMed and web of science for prospective, randomized, and controlled trials (RCTs) of melatonin preparations vs placebo in children and adolescents with ASD and the European public assessment report on Ped-PRM was conducted. EXPERT OPINION Melatonin is rapidly absorbed and undergoes first pass hepatic metabolism by cytochrome CYP1A2; over 80% is excreted in the urine as 6-sulfatoxymelatonin (inactive). Immediate-release melatonin (IRM) is short-acting (3-4 h), whereas PRM provides therapeutic levels throughout the night. Drugs interacting with CYP1A2 are likely to slow-down melatonin metabolism. High variability in bioavailability among subjects calls for dose optimization. Melatonin was essentially safe for short-term use (up to 3 months). Long-term data available for Ped-PRM demonstrate fatigue (6.3%), somnolence (6.3%), and mood swings (4.2%) with no evidence of effects on height, BMI, or pubertal development, tolerance or withdrawal effects following long-term use of this product. Studies on long-term safety of IRM and oversight of melatonin supplement manufacture are warranted.
Collapse
Affiliation(s)
- Nava Zisapel
- Department of Neurobiology, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel.,Research and Development unit, Neurim Pharmaceuticals Ltd, Tel Aviv, Israel
| |
Collapse
|
49
|
Li L, Gang X, Wang J, Gong X. Role of melatonin in respiratory diseases (Review). Exp Ther Med 2022; 23:271. [PMID: 35251337 PMCID: PMC8892605 DOI: 10.3892/etm.2022.11197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/27/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Lijie Li
- Department of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Xiaochao Gang
- Department of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Jiajia Wang
- Department of Pediatrics, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Xiaoyan Gong
- Department of Respiratory Medicine, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
50
|
Cardinali DP, Brown GM, Pandi-Perumal SR. Melatonin's Benefits and Risks as a Therapy for Sleep Disturbances in the Elderly: Current Insights. Nat Sci Sleep 2022; 14:1843-1855. [PMID: 36267165 PMCID: PMC9578490 DOI: 10.2147/nss.s380465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/03/2022] [Indexed: 01/19/2023] Open
Abstract
Aging is accompanied by circadian changes, including disruptive alterations in the sleep/wake cycle, as well as the beginning of low-degree inflammation ("inflammaging"), a scenario that leads to several chronic illnesses, including cancer, and metabolic, cardiovascular, and neurological dysfunctions. As a result, any effective approach to healthy aging must consider both the correction of circadian disturbance and the control of low-grade inflammation. One of the most important prerequisites for healthy aging is the preservation of robust circadian rhythmicity (particularly of the sleep/wake cycle). Sleep disturbance disrupts various activities in the central nervous system, including waste molecule elimination. Melatonin is a chemical with extraordinary phylogenetic conservation found in all known aerobic creatures whose alteration plays an important role in sleep changes with aging. Every day, the late afternoon/nocturnal surge in pineal melatonin helps to synchronize both the central circadian pacemaker found in the hypothalamic suprachiasmatic nuclei (SCN) and a plethora of peripheral cellular circadian clocks. Melatonin is an example of an endogenous chronobiotic substance that can influence the timing and amplitude of circadian rhythms. Moreover, melatonin is also an excellent anti-inflammatory agent, buffering free radicals, down-regulating proinflammatory cytokines, and reducing insulin resistance, among other things. We present both scientific and clinical evidence that melatonin is a safe drug for treating sleep disturbances in the elderly.
Collapse
Affiliation(s)
- Daniel P Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Gregory M Brown
- Molecular Brain Science Research Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | | |
Collapse
|