151
|
Yang J, Xu H, Gao R, Liu X, He J, Zhou M, Ding Y, Li F, Geng Y, Mu X, Liu T, Wang Y, Chen X. Exposure to Benzo(a)pyrene damages mitochondrial function via suppressing mitochondrial melatonin receptors in ovarian corpus luteum during early pregnancy. Chem Biol Interact 2022; 365:110085. [PMID: 35940284 DOI: 10.1016/j.cbi.2022.110085] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/03/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022]
|
152
|
Li ZR, Liu DG, Xie S, Wang YH, Han YS, Li CY, Zou MS, Jiang HX. Sleep deprivation leads to further impairment of hippocampal synaptic plasticity by suppressing melatonin secretion in the pineal gland of chronically unpredictable stress rats. Eur J Pharmacol 2022; 930:175149. [PMID: 35878808 DOI: 10.1016/j.ejphar.2022.175149] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/23/2022] [Accepted: 07/07/2022] [Indexed: 11/27/2022]
Abstract
There has been ample research showing that insomnia is a potential trigger of depression as well as a symptom of depression. These two factors contribute to behavioral problems and are closely related to the plasticity of hippocampal synapses. Although depression and insomnia impair hippocampal synaptic plasticity, the mechanism by which this happens remains a mystery. This study aimed to investigate the pathogenesis of insomnia comorbidity in depression and the regulatory effect of venlafaxine combined with melatonin on hippocampal synaptic plasticity in chronic unpredictable mild stress (CUMS) with sleep deprivation (SD) rats. Thus, rats were subjected to 14 days of chronic mild unpredictable stress, gradually acclimated to sleep deprivation on days 12-14. Followed by 21 consecutive days of sleep deprivation, 18 hours per day, with daily gavage of venlafaxine (13.5 mg/kg) + melatonin (72 mg/kg) on days 15-36. Venlafaxine + melatonin treatment improves depression-like behavior, pentobarbital sodium experimental sleep latency, and sleep duration in CUMS +SD rats. In addition to improving depressive-like behaviors, sleep deprivation also upregulates the expression of caspase-specific cysteine protein 3 (Caspase 3) in the pineal glial cells of chronic mild rats, as well as in hippocampal microglia. Expression of ionic calcium-binding adaptor 1 (iba-1), downregulates the secretion of several synaptic plasticity-related proteins, notably cAMP response element binding protein (CREB), glial cell line-derived neurotrophic factor (GDNF), and the synaptic scaffolding protein Spinophiline (Spinophiline). Hematoxylin-eosin staining showed that the structure of the pineal gland and hippocampus was damaged, and Golgi staining showed that the dendrites and spines in the DG area of the hippocampus were destroyed, vaguely aggregated or even disappeared, and the connection network could not be established. Western blot analysis further revealed a positive correlation between low melatonin levels and reduced Spinophiline protein. Interestingly, venlafaxine + melatonin reversed these events by promoting hippocampal synaptic plasticity by regulating melatonin secretion from the pineal gland. Therefore, it exerted an antidepressant effect in sleep deprivation combined with CUMS model rats. Overall, the results of this study suggest that the pathophysiology of depressive insomnia comorbidity is mediated by impaired pineal melatonin secretion and impaired hippocampal synaptic plasticity. In addition, these responses are associated with melatonin secretion from the pineal gland.
Collapse
Affiliation(s)
- Zi-Rong Li
- Department of Neurology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi, Nanning, 530022, China; State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan (incubation), Hunan University of Chinese Medicine, Hunan, Changsha, 410208, China
| | - De-Guo Liu
- Department of Breast Surgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi, Nanning, 530022, China
| | - Sheng Xie
- Prevention of Diseases with Traditional Chinese Medicine Center, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi, Nanning, 530022, China.
| | - Yu-Hong Wang
- State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan (incubation), Hunan University of Chinese Medicine, Hunan, Changsha, 410208, China.
| | - Yuan-Shan Han
- Department of Experimental Center for Medical Innovation, The First Affiliated Hospital of Hunan University of Chinese Medicine, Hunan, Changsha, 410021, China
| | - Chun-Yan Li
- State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan (incubation), Hunan University of Chinese Medicine, Hunan, Changsha, 410208, China
| | - Man-Shu Zou
- State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan (incubation), Hunan University of Chinese Medicine, Hunan, Changsha, 410208, China
| | - Hai-Xing Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Guangxi, Nanning, 530021, China
| |
Collapse
|
153
|
Gbahou F, Levin S, Tikhonova IG, Somalo Barranco G, Izabelle C, Ohana RF, Jockers R. Luminogenic HiBiT Peptide-Based NanoBRET Ligand Binding Assays for Melatonin Receptors. ACS Pharmacol Transl Sci 2022; 5:668-678. [DOI: 10.1021/acsptsci.2c00096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Florence Gbahou
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 Paris, France
| | - Sergiy Levin
- Promega Corporation, Fitchburg, Wisconsin 53711, United States
| | - Irina G. Tikhonova
- School of Pharmacy, Medical Biology Centre, Queen’s University Belfast, Belfast BT9 7BL, United Kingdom
| | | | - Charlotte Izabelle
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 Paris, France
| | | | - Ralf Jockers
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 Paris, France
| |
Collapse
|
154
|
Maleki Dana P, Sadoughi F, J Reiter R, Mohammadi S, Heidar Z, Mirzamoradi M, Asemi Z. Melatonin as an adjuvant treatment modality with doxorubicin. Biochimie 2022; 202:49-55. [PMID: 35752222 DOI: 10.1016/j.biochi.2022.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Combination chemotherapy seems to be a beneficial choice for some cancer patients particularly when the drugs target different processes of oncogenesis; patients treated with combination therapies sometimes have a better prognosis than those treated with single drug chemotherapy. However, research has shown that this is not always the case, and this approach may only increase toxicity without having a significant effect in augmenting the antitumor actions of the drugs. Doxorubicin (Dox) is one of the most common chemotherapy drugs used to treat many types of cancer, but it also has serious side effects, such as cardiotoxicity, skin necrosis, testicular toxicity, and nephrotoxicity. Many studies have examined the efficiacy of melatonin (MLT) as an anticancer agent. In fact, MLT is an anti-cancer agent that has various functions in inhibiting cancer cell proliferation, inducing apoptosis, and suppressing metastasis. Herein, we provide a comprehensive evaluation of the literature concerned with the role of MLT as an adjuvant in Dox-based chemotherapies and discuss how MLT may enhance the antitumor effects of Dox (e.g., by inducing apoptosis and suppressing metastasis) while rescuring other organs from its adverse effects, such as cardio- and nephrotoxicity.
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran.
| | - Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran.
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA.
| | - Sotoudeh Mohammadi
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Zahra Heidar
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Mirzamoradi
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran.
| |
Collapse
|
155
|
Melatonin Receptors: A Key Mediator in Animal Reproduction. Vet Sci 2022; 9:vetsci9070309. [PMID: 35878326 PMCID: PMC9320721 DOI: 10.3390/vetsci9070309] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 01/26/2023] Open
Abstract
Melatonin, a hormone produced by the mammalian pineal gland, influences various physiological activities, many of which are related to animal reproduction, including neuroendocrine function, rhythm regulation, seasonal behavior, gonadogenesis, gamete development and maturation, sexual maturation, and thermoregulation. Melatonin exerts beneficial actions mainly via binding with G-protein-coupled receptors (GPCR), termed MT1 and MT2. Melatonin receptors are crucial for mediating animal reproduction. This paper reviews the characteristics of melatonin receptors including MT1 and MT2, as well as their roles in mediating signal transduction and biological effects, with a focus on their function in animal reproduction. In addition, we briefly summarize the developments in pharmacological research regarding melatonin receptors as drug targets. It is expected that this review will provide a reference for further exploration and unveiling of melatonin receptor function in reproductive regulation.
Collapse
|
156
|
Tobeiha M, Jafari A, Fadaei S, Mirazimi SMA, Dashti F, Amiri A, Khan H, Asemi Z, Reiter RJ, Hamblin MR, Mirzaei H. Evidence for the Benefits of Melatonin in Cardiovascular Disease. Front Cardiovasc Med 2022; 9:888319. [PMID: 35795371 PMCID: PMC9251346 DOI: 10.3389/fcvm.2022.888319] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022] Open
Abstract
The pineal gland is a neuroendocrine gland which produces melatonin, a neuroendocrine hormone with critical physiological roles in the circadian rhythm and sleep-wake cycle. Melatonin has been shown to possess anti-oxidant activity and neuroprotective properties. Numerous studies have shown that melatonin has significant functions in cardiovascular disease, and may have anti-aging properties. The ability of melatonin to decrease primary hypertension needs to be more extensively evaluated. Melatonin has shown significant benefits in reducing cardiac pathology, and preventing the death of cardiac muscle in response to ischemia-reperfusion in rodent species. Moreover, melatonin may also prevent the hypertrophy of the heart muscle under some circumstances, which in turn would lessen the development of heart failure. Several currently used conventional drugs show cardiotoxicity as an adverse effect. Recent rodent studies have shown that melatonin acts as an anti-oxidant and is effective in suppressing heart damage mediated by pharmacologic drugs. Therefore, melatonin has been shown to have cardioprotective activity in multiple animal and human studies. Herein, we summarize the most established benefits of melatonin in the cardiovascular system with a focus on the molecular mechanisms of action.
Collapse
Affiliation(s)
- Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Fadaei
- Department of Internal Medicine and Endocrinology, Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, United States
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Johannesburg, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
157
|
Cecon E, Fernandois D, Renault N, Coelho CFF, Wenzel J, Bedart C, Izabelle C, Gallet S, Le Poder S, Klonjkowski B, Schwaninger M, Prevot V, Dam J, Jockers R. Melatonin drugs inhibit SARS-CoV-2 entry into the brain and virus-induced damage of cerebral small vessels. Cell Mol Life Sci 2022; 79:361. [PMID: 35697820 PMCID: PMC9191404 DOI: 10.1007/s00018-022-04390-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/11/2022] [Accepted: 05/20/2022] [Indexed: 02/07/2023]
Abstract
COVID-19 is a complex disease with short- and long-term respiratory, inflammatory and neurological symptoms that are triggered by the infection with SARS-CoV-2. Invasion of the brain by SARS-CoV-2 has been observed in humans and is postulated to be involved in post-COVID state. Brain infection is particularly pronounced in the K18-hACE2 mouse model of COVID-19. Prevention of brain infection in the acute phase of the disease might thus be of therapeutic relevance to prevent long-lasting symptoms of COVID-19. We previously showed that melatonin or two prescribed structural analogs, agomelatine and ramelteon delay the onset of severe clinical symptoms and improve survival of SARS-CoV-2-infected K18-hACE2 mice. Here, we show that treatment of K18-hACE2 mice with melatonin and two melatonin-derived marketed drugs, agomelatine and ramelteon, prevents SARS-CoV-2 entry in the brain, thereby reducing virus-induced damage of small cerebral vessels, immune cell infiltration and brain inflammation. Molecular modeling analyses complemented by experimental studies in cells showed that SARS-CoV-2 entry in endothelial cells is prevented by melatonin binding to an allosteric-binding site on human angiotensin-converting enzyme 2 (ACE2), thus interfering with ACE2 function as an entry receptor for SARS-CoV-2. Our findings open new perspectives for the repurposing of melatonergic drugs and its clinically used analogs in the prevention of brain infection by SARS-CoV-2 and COVID-19-related long-term neurological symptoms.
Collapse
Affiliation(s)
- Erika Cecon
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Daniela Fernandois
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France
| | - Nicolas Renault
- Univ Lille, INSERM, CHU Lille, U-1286 - INFINTE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Caio Fernando Ferreira Coelho
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France
| | - Jan Wenzel
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center for Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Hamburg-Lübeck-Kiel, Hamburg, Germany
| | - Corentin Bedart
- Univ Lille, INSERM, CHU Lille, U-1286 - INFINTE - Institute for Translational Research in Inflammation, 59000, Lille, France.,Par'Immune, Bio-incubateur Eurasanté, 70 rue du Dr. Yersin, 59120, Loos-Lez-Lille, France
| | - Charlotte Izabelle
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Sarah Gallet
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France
| | - Sophie Le Poder
- UMR Virologie, INRAE, ANSES, École Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Bernard Klonjkowski
- UMR Virologie, INRAE, ANSES, École Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center for Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Hamburg-Lübeck-Kiel, Hamburg, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience and Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France
| | - Julie Dam
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014, Paris, France
| | - Ralf Jockers
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014, Paris, France.
| |
Collapse
|
158
|
Melatonergic Receptors (Mt1/Mt2) as a Potential Additional Target of Novel Drugs for Depression. Neurochem Res 2022; 47:2909-2924. [PMID: 35689787 PMCID: PMC9187850 DOI: 10.1007/s11064-022-03646-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 11/06/2022]
Abstract
A complex pathogenesis involving several physiological systems is theorized to underline the development of depressive disorders. Depression is accompanied by circadian regulation disruption and interaction with the functioning of both central and peripheral oscillators. Many aspects of melatonin function unite these systems. The use of drugs for circadian rhythm disorders could inspire a potential treatment strategy for depression. Melatonin plays an essential role in the regulation of circadian rhythms. It exerts effect by activating two types of melatonin receptors, type 1A (MT1) and 1B (MT2). These are G-protein-coupled receptors, predominantly located in the central nervous system. MT1/MT2 agonists could be a useful treatment approach according to all three prevalent theories of the pathogenesis of depression involving either monoamines, synaptic remodeling, or immune/inflammatory events. MT1/MT2 receptors can be a potential target for novel antidepressants with impact on concentrations of neurotrophins or neurotransmitters, and reducing levels of pro-inflammatory cytokines. There is an interesting cross-talk mediated via the physical association of melatonin and serotonin receptors into functional heteromers. The antidepressive and neurogenetic effects of MT1/MT2 agonists can also be caused by the inhibition of the acid sphingomyelinase, leading to reduced ceramide, or increasing monoamine oxidase A levels in the hippocampus. Compounds targeting MT1 and MT2 receptors could have potential for new anti-depressants that may improve the quality of therapeutic interventions in treating depression and relieving symptoms. In particular, a combined effect on MT1 and/or MT2 receptors and neurotransmitter systems may be useful, since the normalization of the circadian rhythm through the melatonergic system will probably contribute to improved treatment. In this review, we discuss melatonergic receptors as a potential additional target for novel drugs for depression.
Collapse
|
159
|
Palagini L, Miniati M, Massa L, Folesani F, Marazziti D, Grassi L, Riemann D. Insomnia and circadian sleep disorders in ovarian cancer: Evaluation and management of underestimated modifiable factors potentially contributing to morbidity. J Sleep Res 2022; 31:e13510. [PMID: 34716629 DOI: 10.1111/jsr.13510] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/13/2021] [Accepted: 09/26/2021] [Indexed: 12/21/2022]
Abstract
Ovarian cancer is the leading cause of gynaecological cancer deaths and the seventh most commonly diagnosed cancer among women worldwide, so that, as it is related to substantial and increasing disease burden, the management of ovarian cancer survivors should be a priority. Such issues involve prevention and management of emotional distress, anxiety/depressive symptoms, and maintenance of quality of life from initial diagnosis to post-treatment. Within this framework, sleep disturbances, in particular insomnia, are emerging as modifiable determinants of mental health, also contributing to substantial morbidity among cancer, including ovarian cancer. To this aim we conducted a systematic review according to PRISMA guidelines on prevalence and management of insomnia and circadian sleep disorders in ovarian cancer, while selecting 22 papers. Insomnia was evaluated in ovarian cancer and, while circadian sleep disturbances were poorly assessed in ovarian cancer, insomnia increased from 14% to 60% of patients. Insomnia was associated with cancer-related comorbid conditions such as emotional distress, anxiety/depressive symptoms and low quality of life. Despite this evidence, no studies have been conducted about insomnia treatment in ovarian cancer. The burden of insomnia and circadian sleep disorders in patients with ovarian cancer still needs to be addressed, and requires a call to action for the evaluation and management of these potential modifiable factors that might contribute to ovarian cancer morbidity.
Collapse
Affiliation(s)
- Laura Palagini
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
- Department of Clinical and Experimental Medicine, Psychiatric Clinic, University of Pisa, Pisa, Italy
| | - Mario Miniati
- Department of Clinical and Experimental Medicine, Psychiatric Clinic, University of Pisa, Pisa, Italy
| | - Lucia Massa
- Department of Clinical and Experimental Medicine, Psychiatric Clinic, University of Pisa, Pisa, Italy
| | - Federica Folesani
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Psychiatric Clinic, University of Pisa, Pisa, Italy
- UniCamillus - Saint Camillus University of Health Sciences, Rome, Italy
| | - Luigi Grassi
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Dieter Riemann
- Department of Psychiatry and Psychotherapy, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
160
|
Nopparat C, Boontor A, Panmanee J, Govitrapong P. Melatonin Attenuates Methamphetamine-Induced Alteration of Amyloid β Precursor Protein Cleaving Enzyme Expressions via Melatonin Receptor in Human Neuroblastoma Cells. Neurotox Res 2022; 40:1086-1095. [PMID: 35648367 DOI: 10.1007/s12640-022-00522-6] [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: 03/24/2022] [Revised: 04/26/2022] [Accepted: 05/21/2022] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is the most prominent neurodegenerative disease represented by the loss of memory and cognitive impairment symptoms and is one of the major health imperilments among the elderly. Amyloid (Aβ) deposit inside the neuron is one of the characteristic pathological hallmarks of this disease, leading to neuronal cell death. In the amyloidogenic processing, the amyloid precursor protein (APP) is cleaved by beta-secretase and γ-secretase to generate Aβ. Methamphetamine (METH) is a psychostimulant drug that causes neurodegeneration and detrimental cognitive deficits. The analogy between the neurotoxic and neurodegenerative profile of METH and AD pathology necessitates an exploration of the underlying molecular mechanisms. In the present study, we found that METH ineluctably affects APP processing, which might contribute to the marked production of Aβ in human neuroblastoma cells. Melatonin, an indolamine produced and released by the pineal gland as well as other extrapineal, has been protective against METH-induced neurodegenerative processes, thus rescuing neuronal cell death. However, the precise action of melatonin on METH has yet to be determined. We further propose to investigate the protective properties of melatonin on METH-induced APP-cleaving secretases. Pretreatment with melatonin significantly reversed METH-induced APP-cleaving secretases and Aβ production. In addition, pretreatment with luzindole, a melatonin receptor antagonist, significantly prevented the protective effect of melatonin, suggesting that the attenuation of the toxic effect on METH-induced APP processing by melatonin was mediated via melatonin receptor. The present results suggested that melatonin has a beneficial role in preventing Aβ generation in a cellular model of METH-induced AD.
Collapse
Affiliation(s)
- Chutikorn Nopparat
- Innovative Learning Center, Srinakharinwirot University, Sukhumvit 23, Bangkok, 10110, Thailand
| | - Anuttree Boontor
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| | - Jiraporn Panmanee
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom, 73170, Thailand
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, 10210, Thailand.
| |
Collapse
|
161
|
Jalodia R, Antoine D, Braniff RG, Dutta RK, Ramakrishnan S, Roy S. Opioid-Use, COVID-19 Infection, and Their Neurological Implications. Front Neurol 2022; 13:884216. [PMID: 35677336 PMCID: PMC9169980 DOI: 10.3389/fneur.2022.884216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/25/2022] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an imminent threat to human health and public safety. ACE2 and transmembrane serine protease 2 proteins on host cells provide the viral entry point to SARS-CoV-2. Although SARS-CoV-2 mainly infects the respiratory system, there have been reports of viral neurotropism and central nervous system injury as indicated by plasma biomarkers, including neurofilament light chain protein and glial fibrillary acidic protein. Even with a small proportion of infections leading to neurological manifestation, the overall number remains high. Common neurological manifestations of SARS-CoV-2 infection include anosmia, ageusia, encephalopathy, and stroke, which are not restricted to only the most severe infection cases. Opioids and opioid antagonists bind to the ACE2 receptor and thereby have been hypothesized to have therapeutic potential in treating COVID-19. However, in the case of other neurotropic viral infections such as human immunodeficiency virus (HIV), opioid use has been established to exacerbate HIV-mediated central nervous system pathogenesis. An analysis of electronic health record data from more than 73 million patients shows that people with Substance Use Disorders are at higher risk of contracting COVID-19 and suffer worse consequences then non-users. Our in-vivo and in-vitro unpublished studies show that morphine treatment causes increased expression of ACE2 in murine lung and brain tissue as early as 24 h post treatment. At the same time, we also observed morphine and lipopolysaccharides treatment lead to a synergistic increase in ACE2 expression in the microglial cell line, SIM-A9. This data suggests that opioid treatment may potentially increase neurotropism of SARS-CoV-2 infection. We have previously shown that opioids induce gut microbial dysbiosis. Similarly, gut microbiome alterations have been reported with SARS-CoV-2 infection and may play a role in predicting COVID-19 disease severity. However, there are no studies thus far linking opioid-mediated dysbiosis with the severity of neuron-specific COVID-19 infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
162
|
Abstract
STUDY DESIGN Controlled, randomized, animal study. OBJECTIVE To investigate the effect of melatonin and its receptors on radicular pain and the possible mechanisms. SUMMARY OF BACKGROUND DATA Lumbar disc herniation (LDH) may induce radicular pain, but the mechanism is not clear and therapeutic effect is still poor. Previously we report central sensitization meaning potentiation of spinal nociceptive synaptic transmission is the critical cause of radicular pain. Melatonin (Mel) has been reported to promote hippocampal synaptic transmission and thus improve learning ability. But the effect of Mel on spinal synaptic transmission and radicular pain are not clear. METHODS Rat LDH model was induced by autologous nucleus pulposus (NP) implantation. Melatonin was delivered intraperitoneally four times a day, from day 1 to day 3 after surgery. Melatonin receptor agonist and antagonists were delivered intrathecally for 3 days as well. Mechanical and thermal pain thresholds were assessed by von Frey filaments and hotplate test respectively. Electrophysiological recording was employed for survey C-fiber evoked field potentials. The protein level of N- methyl-D-aspartate submit 2A (NR2A), NR2B, melatonin receptor 1 (MT1), and receptor 2 (MT2) was evaluated by western blotting. Spinal expression of calcitonin gene related peptides (CGRP), isolectin b4 (IB4), and neurofilament-200 (NF200) was displayed by immunofluorescence staining. RESULTS Melatonin significantly increased mechanical and thermal pain thresholds, lasting at least to day 5 after surgery. Melatonin decreased C-fiber evoked field potentials; decreased spinal NR2B protein level; reduced spinal CGRP, and IB4 expression. MT2 was upregulated after NP implantation and was co-localized with neuron and microglia. MT2 receptor agonist simulated the effect of Mel, and both MT receptor broadspectrum antagonist and MT2 specific antagonist abolished the effect of MT2 receptor agonist. CONCLUSION Melatonin alleviates radicular pain from LDH by inhibiting central sensitization via binding with its receptor 2, decreasing spinal CGRP, IB4, and NR2B expression.
Collapse
|
163
|
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
|
164
|
Reiter RJ, Sharma R, Rosales-Corral S, de Campos Zuccari DAP, de Almeida Chuffa LG. Melatonin: A mitochondrial resident with a diverse skill set. Life Sci 2022; 301:120612. [PMID: 35523285 DOI: 10.1016/j.lfs.2022.120612] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/25/2022] [Accepted: 04/30/2022] [Indexed: 12/12/2022]
Abstract
Melatonin is an ancient molecule that originated in bacteria. When these prokaryotes were phagocytized by early eukaryotes, they eventually developed into mitochondria and chloroplasts. These new organelles retained the melatonin synthetic capacity of their forerunners such that all present-day animal and plant cells may produce melatonin in their mitochondria and chloroplasts. Melatonin concentrations are higher in mitochondria than in other subcellular compartments. Isolated mouse oocyte mitochondria form melatonin when they are incubated with serotonin, a necessary precursor. Oocyte mitochondria subsequently give rise to these organelles in all adult vertebrate cells where they continue to synthesize melatonin. The enzymes that convert serotonin to melatonin, i.e., arylalkylamine-N-acetyltransferase (AANAT) and acetylserotonin-O-methyltransferase, have been identified in brain mitochondria which, when incubated with serotonin, also form melatonin. Melatonin is a potent antioxidant and anti-cancer agent and is optimally positioned in mitochondria to aid in the maintenance of oxidative homeostasis and to reduce cancer cell transformation. Melatonin stimulates the transfer of mitochondria from healthy cells to damaged cells via tunneling nanotubes. Melatonin also regulates the major NAD+-dependent deacetylase, sirtuin 3, in the mitochondria. Disruptions of mitochondrial melatonin synthesis may contribute to a number of mitochondria-related diseases, as discussed in this review.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | - Sergio Rosales-Corral
- Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco CP45150, Mexico
| | | | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP-São Paulo State University, Botucatu, São Paulo 18618-689, Brazil
| |
Collapse
|
165
|
Yalçin M, Mundorf A, Thiel F, Amatriain-Fernández S, Kalthoff IS, Beucke JC, Budde H, Garthus-Niegel S, Peterburs J, Relógio A. It's About Time: The Circadian Network as Time-Keeper for Cognitive Functioning, Locomotor Activity and Mental Health. Front Physiol 2022; 13:873237. [PMID: 35547585 PMCID: PMC9081535 DOI: 10.3389/fphys.2022.873237] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 12/24/2022] Open
Abstract
A variety of organisms including mammals have evolved a 24h, self-sustained timekeeping machinery known as the circadian clock (biological clock), which enables to anticipate, respond, and adapt to environmental influences such as the daily light and dark cycles. Proper functioning of the clock plays a pivotal role in the temporal regulation of a wide range of cellular, physiological, and behavioural processes. The disruption of circadian rhythms was found to be associated with the onset and progression of several pathologies including sleep and mental disorders, cancer, and neurodegeneration. Thus, the role of the circadian clock in health and disease, and its clinical applications, have gained increasing attention, but the exact mechanisms underlying temporal regulation require further work and the integration of evidence from different research fields. In this review, we address the current knowledge regarding the functioning of molecular circuits as generators of circadian rhythms and the essential role of circadian synchrony in a healthy organism. In particular, we discuss the role of circadian regulation in the context of behaviour and cognitive functioning, delineating how the loss of this tight interplay is linked to pathological development with a focus on mental disorders and neurodegeneration. We further describe emerging new aspects on the link between the circadian clock and physical exercise-induced cognitive functioning, and its current usage as circadian activator with a positive impact in delaying the progression of certain pathologies including neurodegeneration and brain-related disorders. Finally, we discuss recent epidemiological evidence pointing to an important role of the circadian clock in mental health.
Collapse
Affiliation(s)
- Müge Yalçin
- Institute for Theoretical Biology (ITB), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Annakarina Mundorf
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Freya Thiel
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sandra Amatriain-Fernández
- Institute for Systems Medicine and Faculty of Human Sciences, MSH Medical School Hamburg, Hamburg, Germany
| | - Ida Schulze Kalthoff
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Jan-Carl Beucke
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Henning Budde
- Institute for Systems Medicine and Faculty of Human Sciences, MSH Medical School Hamburg, Hamburg, Germany
| | - Susan Garthus-Niegel
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Jutta Peterburs
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Angela Relógio
- Institute for Theoretical Biology (ITB), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
166
|
Pfeffer M, von Gall C, Wicht H, Korf HW. The Role of the Melatoninergic System in Circadian and Seasonal Rhythms—Insights From Different Mouse Strains. Front Physiol 2022; 13:883637. [PMID: 35492605 PMCID: PMC9039042 DOI: 10.3389/fphys.2022.883637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 01/01/2023] Open
Abstract
The melatoninergic system comprises the neurohormone melatonin and its molecular targets. The major source of melatonin is the pineal organ where melatonin is rhythmically produced during darkness. In mammals, melatonin biosynthesis is controlled by the central circadian rhythm generator in the suprachiasmatic nucleus (SCN) and photoreceptors in the retina. Melatonin elicits its function principally through two specific receptors called MT1 and MT2. MT1 is highly expressed in the SCN and the hypophysial pars tuberalis (PT), an important interface for control of seasonal functions. The expression of the MT2 is more widespread. The role of the melatoninergic system in the control of seasonal functions, such as reproduction, has been known for more than 4 decades, but investigations on its impact on the circadian system under normal (entrained) conditions started 2 decades later by comparing mouse strains with a fully functional melatoninergic system with mouse strains which either produce insufficient amounts of melatonin or lack the melatonin receptors MT1 and MT2. These studies revealed that an intact melatoninergic system is not required for the generation or maintenance of rhythmic behavior under physiological entrained conditions. As shown by jet lag experiments, the melatoninergic system facilitated faster re-entrainment of locomotor activity accompanied by a more rapid adaptation of the molecular clock work in the SCN. This action depended on MT2. Further studies indicated that the endogenous melatoninergic system stabilizes the locomotor activity under entrained conditions. Notably, these effects of the endogenous melatoninergic system are subtle, suggesting that other signals such as corticosterone or temperature contribute to the synchronization of locomotor activity. Outdoor experiments lasting for a whole year indicate a seasonal plasticity of the chronotype which depends on the melatoninergic system. The comparison between mice with an intact or a compromised melatoninergic system also points toward an impact of this system on sleep, memory and metabolism.
Collapse
Affiliation(s)
- Martina Pfeffer
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- *Correspondence: Martina Pfeffer,
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Helmut Wicht
- Dr. Senckenbergische Anatomie II, Fachbereich Medizin der Goethe-Universität, Frankfurt am Main, Germany
| | - Horst-Werner Korf
- Institute of Anatomy I, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
167
|
Egea J, López-Muñoz F, Fernández-Capetillo O, Reiter RJ, Romero A. Alkylating Agent-Induced Toxicity and Melatonin-Based Therapies. Front Pharmacol 2022; 13:873197. [PMID: 35401215 PMCID: PMC8984144 DOI: 10.3389/fphar.2022.873197] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Research Laboratory, Research Unit, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria-Hospital Universitario de La Princesa, Madrid, Spain
| | - Francisco López-Muñoz
- Faculty of Health, University Camilo José Cela, Madrid, Spain.,Neuropsychopharmacology Unit, Hospital Doce de Octubre Research Institute (i+12), Madrid, Spain
| | - Oscar Fernández-Capetillo
- Genomic Instability Group, Spanish National Cancer Research Centre, Madrid, Spain.,Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Russel J Reiter
- UTexas Health San Antonio, Long School of Medicine, San Antonio, TX, United States
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
168
|
Leelaviwat N, Mekraksakit P, Cross KM, Landis DM, McLain M, Sehgal L, Payne JD. Melatonin: Translation of Ongoing Studies Into Possible Therapeutic Applications Outside Sleep Disorders. Clin Ther 2022; 44:783-812. [DOI: 10.1016/j.clinthera.2022.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 12/14/2022]
|
169
|
Park H, Kim J. Activation of melatonin receptor 1 by CRISPR-Cas9 activator ameliorates cognitive deficits in an Alzheimer's disease mouse model. J Pineal Res 2022; 72:e12787. [PMID: 35133672 DOI: 10.1111/jpi.12787] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the presence of neurotoxic beta-amyloid (Aβ) in the brain. Melatonin receptors have been reported to associate with aging and AD, and their expression decreased with the progression of AD. As an alternative to AD treatment, overexpression of melatonin receptors may lead to melatonin-like effects to treat alleviate the symptoms of AD. Here, we successfully activated the type 1 melatonin receptor (Mt1) in vivo brain using a Cas9 activator as a novel AD therapeutic strategy. The Cas9 activator efficiently activated the endogenous Mt1 gene in the brain. Activation of Mt1 via Cas9 activators modulated anti-amyloidogenic and anti-inflammatory roles in 5xFAD AD mice brain. Moreover, activation of Mt1 with the CRISPR/Cas9 activator improved cognitive deficits in an AD model. These results demonstrated the therapeutic potential of melatonin receptor activation via CRISPR/Cas9 activator for AD.
Collapse
Affiliation(s)
- Hanseul Park
- Department of Chemistry, Laboratory of Stem Cells & Cell Reprogramming, Dongguk University, Seoul, Republic of Korea
| | - Jongpil Kim
- Department of Chemistry, Laboratory of Stem Cells & Cell Reprogramming, Dongguk University, Seoul, Republic of Korea
| |
Collapse
|
170
|
Melatonin-Induced Postconditioning Suppresses NMDA Receptor through Opening of the Mitochondrial Permeability Transition Pore via Melatonin Receptor in Mouse Neurons. Int J Mol Sci 2022; 23:ijms23073822. [PMID: 35409182 PMCID: PMC8998233 DOI: 10.3390/ijms23073822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/02/2022] [Accepted: 03/28/2022] [Indexed: 12/29/2022] Open
Abstract
Mitochondrial membrane potential regulation through the mitochondrial permeability transition pore (mPTP) is reportedly involved in the ischemic postconditioning (PostC) phenomenon. Melatonin is an endogenous hormone that regulates circadian rhythms. Its neuroprotective effects via mitochondrial melatonin receptors (MTs) have recently attracted attention. However, details of the neuroprotective mechanisms associated with PostC have not been clarified. Using hippocampal CA1 pyramidal cells from C57BL mice, we studied the involvement of MTs and the mPTP in melatonin-induced PostC mechanisms similar to those of ischemic PostC. We measured changes in spontaneous excitatory postsynaptic currents (sEPSCs), intracellular calcium concentration, mitochondrial membrane potential, and N-methyl-D-aspartate receptor (NMDAR) currents after ischemic challenge, using the whole-cell patch-clamp technique. Melatonin significantly suppressed increases in sEPSCs and intracellular calcium concentrations. The NMDAR currents were significantly suppressed by melatonin and the MT agonist, ramelteon. However, this suppressive effect was abolished by the mPTP inhibitor, cyclosporine A, and the MT antagonist, luzindole. Furthermore, both melatonin and ramelteon potentiated depolarization of mitochondrial membrane potentials, and luzindole suppressed depolarization of mitochondrial membrane potentials. This study suggests that melatonin-induced PostC via MTs suppressed the NMDAR that was induced by partial depolarization of mitochondrial membrane potential by opening the mPTP, reducing excessive release of glutamate and inducing neuroprotection against ischemia-reperfusion injury.
Collapse
|
171
|
Akhzari M, Barazesh M, Jalili S. Melatonin as an antioxidant agent in disease prevention: A biochemical focus. LETT ORG CHEM 2022. [DOI: 10.2174/1570178619666220325124451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Abstract:
In the recent years, free radicals and oxidative stress have been found to be associated with aging, cancer, atherosclerosis, neurodegenerative disorders, diabetes, and inflammatory diseases. Confirming the role of oxidants in numerous pathological situations including cancer, developing antioxidants as therapeutic platforms is needed. It has been well established that melatonin and its derived metabolites function as endogenous free-radical scavengers and broad spectrum antioxidants. To achieve this function, melatonin can directly detoxify reactive oxygen and reactive nitrogen species and indirectly overexpress antioxidant enzymes while suppressing the activity of pro-oxidant enzymes. Many investigations have also confirmed the role of melatonin and its derivatives in different physiological processes and therapeutic functions such as controlling the circadian rhythm and immune functions. This review aimed to focus on melatonin as a beneficial agent for the stimulation of antioxidant enzymes and inhibition of lipid peroxidation and to evaluate its contribution to protection against oxidative damages. In addition, the clinical application of melatonin in several diseases is discussed. Finally, the safety and efficacy of melatonin in clinical backgrounds is also reviewed.
Collapse
Affiliation(s)
- Morteza Akhzari
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Mahdi Barazesh
- School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| | - Sajad Jalili
- Department of Orthopedics, Faculty of Medicine, Ahvaz, Jundishapour University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
172
|
Elisi GM, Scalvini L, Lodola A, Bedini A, Spadoni G, Rivara S. In silico drug discovery of melatonin receptor ligands with therapeutic potential. Expert Opin Drug Discov 2022; 17:343-354. [PMID: 35255751 DOI: 10.1080/17460441.2022.2043846] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The neurohormone melatonin (N-acetyl-5-methoxytryptamine) regulates circadian rhythms exerting a variety of effects in the central nervous system and in periphery. These activities are mainly mediated by activation of MT1 and MT2 GPCRs. MT1/MT2 agonist compounds are used clinically for insomnia, depression, and circadian rhythm disturbances. AREA COVERED The following review describes the design strategies that have led to the identification of melatonin receptor ligands, guided by in silico approaches and molecular modeling. Initial ligand-based design, mainly relying on pharmacophore modeling and 3D-QSAR, has been flanked by structure-based virtual screening, given the recent availability of MT1 and MT2 crystal structures. Receptor ligands with different activity profiles, agonist/antagonist and subtype-selective compounds, are available. EXPERT OPINION An insight on the pharmacological characterization and therapeutic perspectives for relevant ligands is provided. In silico drug discovery has been instrumental in the design of novel ligands targeting melatonin receptors. Ligand-based approaches has led to the construction of a solid framework defining structure-activity relationships to obtain compounds with a tailored pharmacological profile. Structure-based techniques could integrate previous knowledge and provide compounds with novel chemotypes and pharmacological activity as drug candidates for disease conditions in which melatonin receptor ligands are currently being investigated, including cancer and pain.
Collapse
Affiliation(s)
- Gian Marco Elisi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parma, Italy
| | - Laura Scalvini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parma, Italy
| | - Alessio Lodola
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parma, Italy
| | - Annalida Bedini
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | - Gilberto Spadoni
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | - Silvia Rivara
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parma, Italy
| |
Collapse
|
173
|
Horcharoensuk P, Yang-En S, Chakritbudsabong W, Samatiwat P, Pramong R, Rungarunlert S, Rungsiwiwut R. Melatonin attenuates dimethyl sulfoxide- and Zika virus-induced degeneration of porcine induced neural stem cells. In Vitro Cell Dev Biol Anim 2022; 58:232-242. [PMID: 35235152 PMCID: PMC8890020 DOI: 10.1007/s11626-022-00648-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/29/2021] [Indexed: 11/29/2022]
Abstract
Domestic pigs have become increasingly popular as a model for human diseases such as neurological diseases. Drug discovery platforms have increasingly been used to identify novel compounds that combat neurodegeneration. Currently, bioactive molecules such as melatonin have been demonstrated to offer a neuroprotective effect in several studies. However, a neurodegenerative platform to study novel compounds in a porcine model has not been fully established. In this study, characterized porcine induced neural stem cells (iNSCs) were used for evaluation of the protective effect of melatonin against chemical and pathogenic stimulation. First, the effects of different concentrations of melatonin on the proliferation of porcine iNSCs were studied. Second, porcine iNSCs were treated with the appropriate concentration of melatonin prior to induced degeneration with dimethyl sulfoxide or Zika virus (ZIKV). The results demonstrated that the percentages of Ki67 expression in porcine iNSCs cultured in 0.1, 1, and 10 nM melatonin were not significantly different from that in the control groups. Melatonin at 1 nM protected porcine iNSCs from DMSO-induced degeneration, as confirmed by a dead cell exclusion assay and mitochondrial membrane potential (ΔΨm) analysis. In addition, pretreatment with melatonin reduced the percentage of dead porcine iNSCs after ZIKV infection. Melatonin increased the ΔΨm, resulting in a decrease in cell degeneration. However, pretreatment with melatonin was unable to suppress ZIKV replication in porcine iNSCs. In conclusion, the present study demonstrated the anti-degenerative effect of melatonin against DMSO- and ZIKV-induced degeneration in porcine iNSCs.
Collapse
Affiliation(s)
- Pongsatorn Horcharoensuk
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10100, Thailand
| | - Sunantha Yang-En
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10100, Thailand
| | - Warunya Chakritbudsabong
- Laboratory of Cellular Biomedicine and Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand.,Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Papavee Samatiwat
- Department of Pharmacology, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10100, Thailand
| | - Ratchadaporn Pramong
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10100, Thailand
| | - Sasitorn Rungarunlert
- Laboratory of Cellular Biomedicine and Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand.,Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Ruttachuk Rungsiwiwut
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10100, Thailand.
| |
Collapse
|
174
|
Sumaya I, Dubocovich M. Melatonin-Mediated Attenuation of Fluphenazine-Induced Hypokinesia in C57BL/6 Mice is Dependent on the Light/Dark Phase. Behav Brain Res 2022; 425:113827. [DOI: 10.1016/j.bbr.2022.113827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 02/11/2022] [Accepted: 03/02/2022] [Indexed: 11/28/2022]
|
175
|
Tian Y, Qin Z, Han Y. Suvorexant with or without ramelteon to prevent delirium: a systematic review and meta-analysis. Psychogeriatrics 2022; 22:259-268. [PMID: 34881812 DOI: 10.1111/psyg.12792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/01/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022]
Abstract
Delirium is a common and serious neurobehavioral syndrome, associated with prolonged hospital stays, and increased morbidity and mortality. As it remains unclear whether suvorexant with or without ramelteon prevents delirium in elderly hospitalized patients, we conducted a systematic review and meta-analysis to evaluate, searching the PubMed, Cochrane Library, Web of Science, EMBASE, and EBSCOhost databases for all randomized controlled trials (RCTs), case-control studies, and cohort studies that investigated the effects of suvorexant with or without ramelteon on delirium in adult hospitalized patients. The primary outcome was the incidence of delirium. Two randomized controlled trials, 7 cohort studies and 2 case-control studies involving 2594 patients were included in this meta-analysis. The results showed that both suvorexant alone (odds ratio (OR) = 0.30, 95% confidence interval (CI): 0.14-0.65, P = 0.002) and suvorexant with ramelteon (OR = 0.39, 95% CI 0.23-0.65, P = 0.0003) reduced the incidence of delirium in adult hospitalized patients. Six studies involved the use of benzodiazepines; subgroup analysis performed separately in the suvorexant alone and suvorexant with ramelteon groups indicated that when benzodiazepine was administered, suvorexant with ramelteon was effective at reducing the incidence of delirium (OR = 0.53, 95% CI 0.37-0.74, P = 0.0002), but no significant difference was observed for suvorexant alone (OR = 0.40, 95% CI 0.11-1.53, P = 0.18). The current literature thus supports the effectiveness of suvorexant with or without ramelteon for delirium prevention, although suvorexant alone failed to significantly reduce the incidence of delirium when benzodiazepine was administered. The present study was limited by the significant heterogeneity among the included studies, and caution should be exercised when interpreting the results. This study was registered in the PROSPERO database (CRD4202017964).
Collapse
Affiliation(s)
- Yu Tian
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zaisheng Qin
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yunyang Han
- Department of Anaesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
176
|
Das NK, Samanta S. The potential anti-cancer effects of melatonin on breast cancer. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Melatonin is the primary hormone of the pineal gland that is secreted at night. It regulates many physiological functions, including the sleep-wake cycle, gonadal activity, free radical scavenging, immunomodulation, neuro-protection, and cancer progression. The precise functions of melatonin are mediated by guanosine triphosphate (GTP)-binding protein (G-protein) coupled melatonin receptor 1 (MT1) and MT2 receptors. However, nuclear receptors are also associated with melatonin activity. Circadian rhythm disruption, shift work, and light exposure at night hamper melatonin production. Impaired melatonin level promotes various pathophysiological changes, including cancer. In our modern society, breast cancer is a serious problem throughout the world. Several studies have been indicated the link between low levels of melatonin and breast cancer development. Melatonin has oncostatic properties in breast cancer cells. This indolamine advances apoptosis, which arrests the cell cycle and regulates metabolic activity. Moreover, melatonin increases the treatment efficacy of cancer and can be used as an adjuvant with chemotherapeutic agents.
Collapse
Affiliation(s)
- Naba Kumar Das
- Department of Physiology, Midnapore College, Midnapore 721101, Paschim Medinipur, West Bengal, India
| | - Saptadip Samanta
- Department of Physiology, Midnapore College, Midnapore 721101, Paschim Medinipur, West Bengal, India
| |
Collapse
|
177
|
Yapislar H, Haciosmanoglu E, Sarioglu T, Ekmekcioglu C. The melatonin MT 2 receptor is involved in the anti-apoptotic effects of melatonin in rats with type 2 diabetes mellitus. Tissue Cell 2022; 76:101763. [PMID: 35247789 DOI: 10.1016/j.tice.2022.101763] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/10/2022] [Accepted: 02/18/2022] [Indexed: 01/14/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a widely prevalent chronic disease and risk factor for several other diseases, such as cardiovascular diseases, neuropathy, nephropathy, and retinopathy. Apoptosis is a homeostatic mechanism to maintain cell numbers at a certain level in tissues. Chronic high blood glucose levels might lead to mitochondrial dysfunction and trigger undesirable apoptosis in T2DM. The pineal hormone melatonin has been shown to regulate apoptosis. The aim of this study was to investigate the impact of the melatonin MT2 receptor in the role of melatonin to prevent undesirable apotosis in different tissues of diabetic rats. Male Sprague Dawley rats were randomly divided into 4 groups; 1. Control group (only vehicle), 2. Diabetic group (streptozotozin/nicotinamide treated), 3. Diabetic group treated with melatonin (500μg/kg/day), and 4. Diabetic group treated with melatonin (500 μg/kg/day for 6 weeks) and the selective MT2 receptor antagonist luzindole (0.25 g/kg/day for 6 weeks). Various tissue samples (kidney, liver, adipose tissue, pancreas) were removed after 6 weeks for immunohistochemistry and western blot analysis. Our results demonstrated an increased rate of apoptosis in different tissues of diabetic rats compared to controls with melatonin reducing the apoptotic rate in the tissues of rats with T2DM. Furthermore, the anti-apoptotic effects of melatonin were partly mediated by the melatonin MT2 receptor.
Collapse
Affiliation(s)
- Hande Yapislar
- Acibadem University, School of Medicine, Department of Physiology, 34684, Istanbul, Turkey.
| | - Ebru Haciosmanoglu
- Faculty of Medicine, Department of Biophysics, Bezmialem Vakif University, Istanbul, Turkey
| | - Turkan Sarioglu
- Department of Histology and Embryology, Fundamental Sciences, Faculty of Dentistry, Istanbul Kent University Istanbul, Turkey
| | - Cem Ekmekcioglu
- Department of Environmental Health, Center for Public Health, Medical University of Vienna, 1090, Vienna, Austria
| |
Collapse
|
178
|
Munley KM, Dutta S, Jasnow AM, Demas GE. Adrenal MT 1 melatonin receptor expression is linked with seasonal variation in social behavior in male Siberian hamsters. Horm Behav 2022; 138:105099. [PMID: 34920297 PMCID: PMC8847318 DOI: 10.1016/j.yhbeh.2021.105099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/10/2021] [Accepted: 11/24/2021] [Indexed: 02/03/2023]
Abstract
Many animals exhibit pronounced changes in physiology and behavior on a seasonal basis, and these adaptations have evolved to promote survival and reproductive success. While the neuroendocrine pathways mediating seasonal reproduction are well-studied, far less is known about the mechanisms underlying seasonal changes in social behavior, particularly outside of the context of the breeding season. Our previous work suggests that seasonal changes in melatonin secretion are important in regulating aggression in Siberian hamsters (Phodopus sungorus); it is unclear, however, how melatonin acts via its receptors to modulate seasonal variation in social behavior. In this study, we infused a MT1 melatonin receptor-expressing (MT1) or control (CON) lentivirus into the adrenal glands of male Siberian hamsters. We then housed hamsters in long-day (LD) or short-day (SD) photoperiods, administered timed melatonin or control injections, and quantified aggressive and non-aggressive social behaviors (e.g., investigation, self-grooming) following 10 weeks of treatment. LD hamsters infused with the MT1 lentivirus had significantly higher adrenal mt1 expression than LD CON hamsters, as determined via quantitative PCR. While melatonin administration was necessary to induce SD-like reductions in body and relative reproductive mass, only LD hamsters infused with the MT1 lentivirus displayed SD-like changes in social behavior, including increased aggression and decreased investigation and grooming. In addition, SD CON and LD hamsters infused with the MT1 lentivirus exhibited similar relationships between adrenal mt1 expression and aggressive behavior. Together, our findings suggest a role for adrenal MT1 receptor signaling in regulating behavior, but not energetics or reproduction in seasonally breeding species.
Collapse
Affiliation(s)
- Kathleen M Munley
- Department of Biology and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA.
| | - Sohini Dutta
- Department of Psychological Sciences, Kent State University, Kent, OH 44240, USA; Department of Neuroscience, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Aaron M Jasnow
- Department of Psychological Sciences, Kent State University, Kent, OH 44240, USA; Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Gregory E Demas
- Department of Biology and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
179
|
Kory P, Meduri GU, Iglesias J, Varon J, Cadegiani FA, Marik PE. "MATH+" Multi-Modal Hospital Treatment Protocol for COVID-19 Infection: Clinical and Scientific Rationale. J Clin Med Res 2022; 14:53-79. [PMID: 35317360 PMCID: PMC8912998 DOI: 10.14740/jocmr4658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
In December 2019, coronavirus disease 2019 (COVID-19), a severe respiratory illness caused by the new coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in Wuhan, China. The greatest impact that COVID-19 had was on intensive care units (ICUs), given that approximately 20% of hospitalized cases developed acute respiratory failure (ARF) requiring ICU admission. Based on the assumption that COVID-19 represented a viral pneumonia and no anti-coronaviral therapy existed, nearly all national and international health care societies recommended "supportive care only" avoiding other therapies outside of randomized controlled trials, with a specific prohibition against the use of corticosteroids in treatment. However, early studies of COVID-19-associated ARF reported inexplicably high mortality rates, with frequent prolonged durations of mechanical ventilation (MV), even from centers expert in such supportive care strategies. These reports led the authors to form a clinical expert panel called the Front-Line COVID-19 Critical Care Alliance (www.flccc.net). The panel collaboratively reviewed the emerging clinical, radiographic, and pathological reports of COVID-19 while initiating multiple discussions among a wide clinical network of front-line clinical ICU experts from initial outbreak areas in China, Italy, and New York. Based on the shared early impressions of "what was working and what wasn't working", the increasing medical journal publications and the rapidly accumulating personal clinical experiences with COVID-19 patients, a treatment protocol was created for the hospitalized patients based on the core therapies of methylprednisolone, ascorbic acid, thiamine, heparin and non-antiviral co-interventions (MATH+). This manuscript reviews the scientific and clinical rationale behind MATH+ based on published in-vitro, pre-clinical, and clinical data in support of each medicine, with a special emphasis of studies supporting their use in the treatment of patients with viral syndromes and COVID-19 specifically.
Collapse
Affiliation(s)
- Pierre Kory
- Front Line Critical Care Consortium (FLCCC.org), Washington DC, USA
| | | | - Jose Iglesias
- Jersey Shore University Medical Center, Hackensack School of Medicine at Seton Hall, NJ, USA
| | - Joseph Varon
- University of Texas Health Science Center, Houston, TX, USA
| | | | - Paul E. Marik
- Front Line Critical Care Consortium (FLCCC.org), Washington DC, USA
| |
Collapse
|
180
|
Chen Z, Zhao S, Tian S, Yan R, Wang H, Wang X, Zhu R, Xia Y, Yao Z, Lu Q. Diurnal mood variation symptoms in major depressive disorder associated with evening chronotype: Evidence from a neuroimaging study. J Affect Disord 2022; 298:151-159. [PMID: 34715183 DOI: 10.1016/j.jad.2021.10.087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 09/16/2021] [Accepted: 10/23/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is often accompanied with classic diurnal mood variation (DMV) symptoms. Patients with DMV symptoms feel a mood improvement and prefer activities at dusk or in the evening, which is consistent with the evening chronotype. Their neural alterations are unclear. In this study, we aimed to explore the neuropathological mechanisms underlying the circadian rhythm of mood and the association with chronotype in MDD. METHODS A total of 126 depressed patients, including 48 with DMV, 78 without, and 67 age/gender-matched healthy controls (HC) were recruited and underwent a resting-state functional magnetic resonance imaging. Spontaneous neural activity was investigated using amplitude of low-frequency fluctuation (ALFF) and region of interest (ROI)-based functional connectivity (FC) analyses were conducted. The Morningness-Eveningness Questionnaire (MEQ) was utilized to evaluate participant chronotypes and Pearson correlations were calculated between altered ALFF/FC values and MEQ scores in patients with MDD. RESULTS Compared with NMV, DMV group exhibited lower MEQ scores, and increased ALFF values in the right orbital superior frontal gyrus (oSFG). We observed that increased FC between the left suprachiasmatic nucleus (SCN) and supramarginal gyrus (SMG). ALFF in the oSFG and FC of rSCN-SMG were negatively correlated with MEQ scores. LIMITATION Some people's chronotypes information is missing. CONCLUSION Patients with DMV tended to be evening type and exhibited abnormal brain functions in frontal lobes. The synergistic changes between frontotemporal lobe, SCN-SMG maybe the characteristic of patients with DMV symptoms.
Collapse
Affiliation(s)
- Zhilu Chen
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shuai Zhao
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shui Tian
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Rui Yan
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China; Nanjing Brain Hospital, Medical School of Nanjing University, Nanjing 210093, China
| | - Huan Wang
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xumiao Wang
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Rongxin Zhu
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yi Xia
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhijian Yao
- Department of Psychiatry, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China; School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, China; Nanjing Brain Hospital, Medical School of Nanjing University, Nanjing 210093, China.
| | - Qing Lu
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210096, China; Child Development and Learning Science, Key Laboratory of Ministry of Education, 210096, China.
| |
Collapse
|
181
|
Yang XB, Zu HB, Zhao YF, Yao K. Agomelatine Prevents Amyloid Plaque Deposition, Tau Phosphorylation, and Neuroinflammation in APP/PS1 Mice. Front Aging Neurosci 2022; 13:766410. [PMID: 35153715 PMCID: PMC8828541 DOI: 10.3389/fnagi.2021.766410] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/20/2021] [Indexed: 01/09/2023] Open
Abstract
Agomelatine, an agonist of melatonergic MT1 and MT2 receptors and a selective 5-hydroxytryptamine 2C receptor antagonist, is widely applied in treating depression and insomnia symptoms in several neurogenerative diseases. However, the neuroprotective effect of agomelatine in Alzheimer’s disease (AD) is less known. In this study, a total of 30 mice were randomly divided into three groups, namely, wild type (WT), APP/PS1, and agomelatine (50 mg/kg). After 30 days, the Morris water maze was performed to test the cognitive ability of mice. Then, all mice were sacrificed, and the hippocampus tissues were collected for ELISA, Western blot, and immunofluorescence analysis. In this study, we found that agomelatine attenuated spatial memory deficit, amyloid-β (Aβ) deposition, tau phosphorylation, and neuroinflammation in the hippocampus of APP/PS1 mice. Further study demonstrated that agomelatine treatment upregulated the protein expression of DHCR24 and downregulated P-Akt, P-mTOR, p-p70s6k, Hes1, and Notch1 expression. In summary, our results identified that agomelatine could improve cognitive impairment and ameliorate AD-like pathology in APP/PS1 mice via activating DHCR24 signaling and inhibiting Akt/mTOR and Hes1/Notch1 signaling pathway. Agomelatine may become a promising drug candidate in the therapy of AD.
Collapse
|
182
|
Structural basis of the ligand binding and signaling mechanism of melatonin receptors. Nat Commun 2022; 13:454. [PMID: 35075127 PMCID: PMC8786939 DOI: 10.1038/s41467-022-28111-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022] Open
Abstract
Melatonin receptors (MT1 and MT2 in humans) are family A G protein-coupled receptors that respond to the neurohormone melatonin to regulate circadian rhythm and sleep. Numerous efforts have been made to develop drugs targeting melatonin receptors for the treatment of insomnia, circadian rhythm disorder, and cancer. However, designing subtype-selective melatonergic drugs remains challenging. Here, we report the cryo-EM structures of the MT1-Gi signaling complex with 2-iodomelatonin and ramelteon and the MT2-Gi signaling complex with ramelteon. These structures, together with the reported functional data, reveal that although MT1 and MT2 possess highly similar orthosteric ligand-binding pockets, they also display distinctive features that could be targeted to design subtype-selective drugs. The unique structural motifs in MT1 and MT2 mediate structural rearrangements with a particularly wide opening on the cytoplasmic side. Gi is engaged in the receptor core shared by MT1 and MT2 and presents a conformation deviating from those in other Gi complexes. Together, our results provide new clues for designing melatonergic drugs and further insights into understanding the G protein coupling mechanism.
Collapse
|
183
|
Melatonin secretion, molecular expression and evolution of MT1/2 in two Lasiopodomys species. Mamm Biol 2022. [DOI: 10.1007/s42991-021-00204-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
184
|
Pohanka M. New uses of Melatonin as a Drug, a Review. Curr Med Chem 2022; 29:3622-3637. [PMID: 34986763 DOI: 10.2174/0929867329666220105115755] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/25/2021] [Accepted: 11/21/2021] [Indexed: 11/22/2022]
Abstract
Melatonin is a simple compound with a proper chemical name N-acetyl-5-methoxy tryptamine and known as a hormone controlling circadian rhythm. Humans produce melatonin at night which is the reason for sleeping in the night and awakening over the day. Melatonin interacts with melatonin receptors MT1 and MT2 but it was also revealed that melatonin is a strong antioxidant and it also has a role in regulation of cell cycle. Currently, melatonin is used as a drug for some types of sleep disorder but the recent research points to the fact that melatonin can also serve for the other purposes including prophylaxis or therapy of lifestyle diseases, cancer, neurodegenerative disorders and exposure to chemicals. This review summarizes basic facts and direction of the current research on melatonin. The actual literature was scrutinized for the purpose of this review.
Collapse
Affiliation(s)
- Miroslav Pohanka
- Faculty of Military Health Sciences, University of Defense, Trebesska 1575, Hradec Kralove CZ-50001, Czech Republic
| |
Collapse
|
185
|
Giannetto C, Rajaei SM, Abdous A, Ostadhasan H, Alagha HE, Faghihi H, Piccione G, Omidi R, Fazio F. Effects of long-term oral administration of melatonin on tear production, intraocular pressure, and tear and serum melatonin concentrations in healthy dogs. J Am Vet Med Assoc 2022; 260:524-529. [PMID: 34986123 DOI: 10.2460/javma.21.03.0114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the effects of long-term (30-day) oral administration of melatonin on tear production, intraocular pressure (IOP), and concentration of melatonin in the tears and serum of healthy dogs. ANIMALS 20 healthy sexually intact adult male dogs. PROCEDURES 10 dogs were given melatonin (0.3 mg/kg, PO, q 24 h, administered in food at 9 am), and 10 dogs were given a placebo. Tear and serum melatonin concentrations, IOP, and tear production (determined with a Schirmer tear test) were recorded before (baseline) and 30 minutes, 3 hours, and 5 hours after administration of melatonin or the placebo on day 1 and 30 minutes after administration of melatonin or the placebo on days 8, 15, and 30. RESULTS Data collection time had significant effects on tear production, IOP, and tear melatonin concentration but not on serum melatonin concentration. Treatment (melatonin vs placebo) had a significant effect on tear melatonin concentration, but not on tear production, IOP, or serum melatonin concentration; however, tear melatonin concentration was significantly different between groups only 30 minutes after administration on day 1 and not at other times. CLINICAL RELEVANCE In healthy dogs, long-term administration of melatonin at a dosage of 0.3 mg/kg, PO, every 24 hours did not have any clinically important effects on tear production, IOP, or serum or tear melatonin concentrations.
Collapse
Affiliation(s)
- Claudia Giannetto
- 1Department of Veterinary Sciences, Polo Universitario Annunziata, University of Messina, Messina, Italy
| | - Seyed Mehdi Rajaei
- 2Ophthalmology Section, Negah Veterinary Centre, Tehran, Iran.,3Department of Clinical Sciences, College of Veterinary Medicine, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Arman Abdous
- 3Department of Clinical Sciences, College of Veterinary Medicine, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Hesam Ostadhasan
- 3Department of Clinical Sciences, College of Veterinary Medicine, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Hannah Emami Alagha
- 3Department of Clinical Sciences, College of Veterinary Medicine, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Houman Faghihi
- 2Ophthalmology Section, Negah Veterinary Centre, Tehran, Iran
| | - Giuseppe Piccione
- 1Department of Veterinary Sciences, Polo Universitario Annunziata, University of Messina, Messina, Italy
| | - Roghiyeh Omidi
- 4Stem Cell Preparation Unit, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Francesco Fazio
- 1Department of Veterinary Sciences, Polo Universitario Annunziata, University of Messina, Messina, Italy
| |
Collapse
|
186
|
Woo SM, Seo SU, Min KJ, Kwon TK. Melatonin induces apoptotic cell death through Bim stabilization by Sp1-mediated OTUD1 upregulation. J Pineal Res 2022; 72:e12781. [PMID: 34826170 DOI: 10.1111/jpi.12781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/19/2021] [Accepted: 11/15/2021] [Indexed: 12/15/2022]
Abstract
Melatonin, secreted by the pineal gland, regulates the circadian rhythms and also plays an oncostatic role in cancer cells. Previously, we showed that melatonin induces the expression of Bim, a pro-apoptotic Bcl-2 protein, at both the transcriptional and post-translational levels. In the present study, we investigated the molecular mechanisms underlying the melatonin-mediated Bim upregulation through post-translational regulation. We found that ovarian tumor domain-containing protein 1 (OTUD1), a deubiquitinase belonging to the OTU protein family, is upregulated by melatonin at the mRNA and protein levels. OTUD1 knockdown inhibited melatonin-induced Bim upregulation and apoptosis in cancer cells. OTUD1 directly interacted with Bim and inhibited its ubiquitination. Melatonin-induced OTUD1 upregulation caused deubiquitination at the lysine 3 residue of Bim, resulting in its stabilization. In addition, melatonin-induced activation of Sp1 was found to be involved in OTUD1 upregulation at the transcriptional level, and pharmacological inhibition and genetic ablation of Sp1 (siRNA) interrupted melatonin-induced OTUD1-mediated Bim upregulation. Furthermore, melatonin reduced tumor growth and induced upregulation of OTUD1 and Bim in a mouse xenograft model. Notably, Bim expression levels correlated with OTUD1 levels in patients with renal clear cell carcinoma. Thus, our results demonstrated that melatonin induces apoptosis by stabilizing Bim via Sp1-mediated OTUD1 upregulation.
Collapse
Affiliation(s)
- Seon Min Woo
- Department of Immunology, School of Medicine, Keimyung University, Daegu, South Korea
| | - Seung Un Seo
- Department of Immunology, School of Medicine, Keimyung University, Daegu, South Korea
| | - Kyoung-Jin Min
- Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), New Drug Development Center, Daegu, South Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, South Korea
- Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, South Korea
| |
Collapse
|
187
|
Cipolla-Neto J, Amaral FG, Soares JM, Gallo CC, Furtado A, Cavaco JE, Gonçalves I, Santos CRA, Quintela T. The Crosstalk between Melatonin and Sex Steroid Hormones. Neuroendocrinology 2022; 112:115-129. [PMID: 33774638 DOI: 10.1159/000516148] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/24/2021] [Indexed: 11/19/2022]
Abstract
Melatonin, an indolamine mainly released from the pineal gland, is associated with many biological functions, namely, the modulation of circadian and seasonal rhythms, sleep inducer, regulator of energy metabolism, antioxidant, and anticarcinogenic. Although several pieces of evidence also recognize the influence of melatonin in the reproductive physiology, the crosstalk between melatonin and sex hormones is not clear. Here, we review the effects of sex differences in the circulating levels of melatonin and update the current knowledge on the link between sex hormones and melatonin. Furthermore, we explore the effects of melatonin on gonadal steroidogenesis and hormonal control in females. The literature review shows that despite the strong evidence that sex differences impact on the circadian profiles of melatonin, reports are still considerably ambiguous, and these differences may arise from several factors, like the use of contraceptive pills, hormonal status, and sleep deprivation. Furthermore, there has been an inconclusive debate about the characteristics of the reciprocal relationship between melatonin and reproductive hormones. In this regard, there is evidence for the role of melatonin in gonadal steroidogenesis brought about by research that shows that melatonin affects multiple transduction pathways that modulate Sertoli cell physiology and consequently spermatogenesis, and also estrogen and progesterone production. From the outcome of our research, it is possible to conclude that understanding the correlation between melatonin and reproductive hormones is crucial for the correction of several complications occurring during pregnancy, like preeclampsia, and for the control of climacteric symptoms.
Collapse
Affiliation(s)
- José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - José Maria Soares
- Laboratório de Ginecologia Estrutural e Molecular (LIM 58), Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, HCFMUSP, São Paulo, Brazil
| | | | - André Furtado
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - José Eduardo Cavaco
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | - Telma Quintela
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
188
|
Li D, Fang P, Liu H, Chen L, Fu Y, Liu J, Xie B, Liu Y, Ye H, Gu P. The Clinical Effect of Blue Light Therapy on Patients with Delayed Sleep-Wake Phase Disorder. Nat Sci Sleep 2022; 14:75-82. [PMID: 35082544 PMCID: PMC8784911 DOI: 10.2147/nss.s344616] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To investigate the feasibility and patient acceptance of applying blue light glasses to treat delayed sleep-wake phase disorder (DSWPD). METHODS Fifteen patients with DSWPD were collected as the observation group and 15 healthy people as the control group. The patients wore blue light glasses with a continuous radiation wavelength of about 470 nm for 1h to 2h during the period from 06:30 to 09:00 in the morning after waking up, respectively. Assessment of Hamilton Anxiety Scale 14 items (HAMA14), Hamilton Depression Rating Scale 24 items (HAMD24), Pittsburgh Sleep Quality Index (PSQI), Epworth Sleepiness Scale (ESS), Morningness-Eveningness Questionnaire (MEQ), and Insomnia Severity Index (ISI) scores before and after 1 week of treatment. Pearson correlation was used to analyze the correlation between the efficacy of patients with sleep-wake phase delay disorder and HAMA14, HAMD24, PSQI, ISI, ESS, MEQ, SL (sleep time), TST (total sleep time), TTiB (total time in bed), SQ (sleep quality), TOA (total arousal time), WASO (wake after sleep onset), AAT (average arousal time), and SE (sleep efficiency percent). Multi-factor logistic regression analysis of factors influencing the efficacy of patients with sleep-wake phase delay disorder. RESULTS After treatment, PSQI-G scores, number of nighttime awakenings and time of awakening recorded in the sleep diary decreased significantly in the observation group (P < 0.05), and subjective sleep quality and MEQ scores increased (P < 0.05). MEQ score shifted from "moderate night type" to "intermediate type", sleep-wake phase tended to shift forward. The total PSQI score and Pittsburgh Sleep Quality Index Global (PSQI-G) score were significantly lower in the control group after treatment (P < 0.05). By Pearson correlation analysis, the efficacy of patients with sleep-wake phase delay disorder was significantly correlated with HAMA14, HAMD24, PSQI, ISI, ESS, MEQ, SL, TST, TTiB, SQ, TOA, WASO, AAT, and SE. Multifactorial logistic regression analysis revealed that the factors influencing the efficacy of patients with sleep-wake phase delay disorder were PSQI, ISI, ESS, MEQ, SL, TST, TTiB, SQ, TOA, WASO, AAT, and SE. CONCLUSION Blue light therapy has a positive effect on improving subjective sleep quality, reducing the number of nocturnal awakenings and the duration of nocturnal awakenings, improving daytime function, and shifting the sleep phase forward in patients with DSWPD. Blue light therapy improves subjective sleep quality and daytime function the following day in normal individuals.
Collapse
Affiliation(s)
- Dong Li
- Department of Vertigo Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Penghong Fang
- Department of Neurology, Xiangya Changde Hospital, Changde, 415000, Hunan, People's Republic of China
| | - Huimiao Liu
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Lili Chen
- Department of Neurology, Tangshan Workers' Hospital, Tangshan, 063000, Hebei, People's Republic of China
| | - Ying Fu
- Department of Brain Function, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Junqian Liu
- Department of Brain Function, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Bingchuan Xie
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Yihan Liu
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Hongyuan Ye
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000, Hebei, People's Republic of China
| | - Ping Gu
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| |
Collapse
|
189
|
Glatfelter GC, Sosa J, Hudson RL, Dubocovich ML. Methods to Assess Melatonin Receptor-Mediated Phase-Shift and Re-entrainment of Rhythmic Behaviors in Mouse Models. Methods Mol Biol 2022; 2550:391-411. [PMID: 36180708 DOI: 10.1007/978-1-0716-2593-4_39] [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] [Indexed: 06/01/2023]
Abstract
The neurohormone melatonin facilitates entrainment of biological rhythms to environmental light-dark conditions as well as phase-shifts of circadian rhythms in constant conditions via activation of the MT1 and/or MT2 receptors expressed within the suprachiasmatic nucleus of the hypothalamus. The efficacy of melatonin and related agonists to modulate biological rhythms can be assessed using two well-validated mouse models of rhythmic behaviors. These models serve as predictive measures of therapeutic efficacy for treatment of circadian phase disorders caused by internal (e.g., clock gene mutations, blindness, depression, seasonal affective disorder) or external (e.g., shift work, travel across time zones) causes in humans. Here we provide background and detailed protocols for quantitative assessment of the magnitude and efficacy of melatonin receptor ligands in mouse circadian phase-shift and re-entrainment paradigms. The utility of these models in the discovery of novel therapeutics acting on melatonin receptors will also be discussed.
Collapse
Affiliation(s)
- Grant C Glatfelter
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
- Designer Drug Research Unit, National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, USA
| | - Jennifer Sosa
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Randall L Hudson
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences University at Buffalo, Buffalo, NY, USA
| | - Margarita L Dubocovich
- Department of Pharmacology and Toxicology, Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences University at Buffalo (SUNY), Buffalo, NY, USA.
| |
Collapse
|
190
|
Romero A, Ramos E, López-Muñoz F, Gil-Martín E, Escames G, Reiter RJ. Coronavirus Disease 2019 (COVID-19) and Its Neuroinvasive Capacity: Is It Time for Melatonin? Cell Mol Neurobiol 2022; 42:489-500. [PMID: 32772307 PMCID: PMC7415199 DOI: 10.1007/s10571-020-00938-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/05/2020] [Indexed: 01/08/2023]
Abstract
The world faces an exceptional new public health concern caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), subsequently termed the coronavirus disease 2019 (COVID-19) by the World Health Organization (WHO). Although the clinical symptoms mostly have been characterized, the scientific community still doesn´t know how SARS-CoV-2 successfully reaches and spreads throughout the central nervous system (CNS) inducing brain damage. The recent detection of SARS-CoV-2 in the cerebrospinal fluid (CSF) and in frontal lobe sections from postmortem examination has confirmed the presence of the virus in neural tissue. This finding reveals a new direction in the search for a neurotherapeutic strategy in the COVID-19 patients with underlying diseases. Here, we discuss the COVID-19 outbreak in a neuroinvasiveness context and suggest the therapeutic use of high doses of melatonin, which may favorably modulate the immune response and neuroinflammation caused by SARS-CoV-2. However, clinical trials elucidating the efficacy of melatonin in the prevention and clinical management in the COVID-19 patients should be actively encouraged.
Collapse
Affiliation(s)
- Alejandro Romero
- grid.4795.f0000 0001 2157 7667Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Eva Ramos
- grid.4795.f0000 0001 2157 7667Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Francisco López-Muñoz
- grid.449750.b0000 0004 1769 4416Faculty of Health Sciences, University Camilo José Cela, C/ Castillo de Alarcón 49, 28692 Villanueva de la Cañada, Madrid, Spain ,grid.144756.50000 0001 1945 5329Neuropsychopharmacology Unit, Hospital 12 de Octubre Research Institute (i+12), Avda. Córdoba, s/n, 28041 Madrid, Spain ,grid.410919.40000 0001 2152 2367Portucalense Institute of Neuropsychology and Cognitive and Behavioural Neurosciences (INPP), Portucalense University, R. Dr. António Bernardino de Almeida 541, 4200-072 Porto, Portugal ,grid.413448.e0000 0000 9314 1427Thematic Network for Cooperative Health Research (RETICS), Addictive Disorders Network, Health Institute Carlos III, MICINN and FEDER, Madrid, Spain
| | - Emilio Gil-Martín
- grid.6312.60000 0001 2097 6738Nutrition and Food Science Group, Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, 36310 Vigo, Spain
| | - Germaine Escames
- grid.4489.10000000121678994Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain ,grid.4489.10000000121678994Department of Physiology, University of Granada, 18016 Granada, Spain ,grid.507088.2Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Instituto de Investigación Biosanitaria CIBERFES, IBS. Granada, Granada Hospital Complex, 18016 Granada, Spain
| | - Russel J. Reiter
- grid.267309.90000 0001 0629 5880Department of Cellular and Structural Biology, University of Texas Health Science at San Antonio, San Antonio, TX 78229 USA
| |
Collapse
|
191
|
Cecon E, Izabelle C, Poder SL, Real F, Zhu A, Tu L, Ghigna MR, Klonjkowski B, Bomsel M, Jockers R, Dam J. Therapeutic potential of melatonin and melatonergic drugs on K18-hACE2 mice infected with SARS-CoV-2. J Pineal Res 2022; 72:e12772. [PMID: 34586649 PMCID: PMC8646885 DOI: 10.1111/jpi.12772] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/01/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022]
Abstract
As the COVID-19 pandemic grows, several therapeutic candidates are being tested or undergoing clinical trials. Although prophylactic vaccination against SARS-CoV-2 infection has been shown to be effective, no definitive treatment exists to date in the event of infection. The rapid spread of infection by SARS-CoV-2 and its variants fully warrants the continued evaluation of drug treatments for COVID-19, especially in the context of repurposing of already available and safe drugs. Here, we explored the therapeutic potential of melatonin and melatonergic compounds in attenuating COVID-19 pathogenesis in mice expressing human ACE2 receptor (K18-hACE2), strongly susceptible to SARS-CoV-2 infection. Daily administration of melatonin, agomelatine, or ramelteon delays the occurrence of severe clinical outcome with improvement of survival, especially with high melatonin dose. Although no changes in most lung inflammatory cytokines are observed, treatment with melatonergic compounds limits the exacerbated local lung production of type I and type III interferons, which is likely associated with the observed improved symptoms in treated mice. The promising results from this preclinical study should encourage studies examining the benefits of repurposing melatonergic drugs to treat COVID-19 and related diseases in humans.
Collapse
Affiliation(s)
- Erika Cecon
- Institut CochinINSERMCNRSUniversité de ParisParisFrance
| | | | - Sophie Le Poder
- UMR VirologieINRAEANSESÉcole Nationale Vétérinaire d'AlfortMaisons‐AlfortFrance
| | - Fernando Real
- Institut CochinINSERMCNRSUniversité de ParisParisFrance
| | - Aiwei Zhu
- Institut CochinINSERMCNRSUniversité de ParisParisFrance
| | - Ly Tu
- School of Medicine Le Kremlin‐BicêtreHôpital Marie Lannelongue, INSERM UMRS 999Université Paris‐SaclayLe Plessis‐RobinsonFrance
| | - Maria Rosa Ghigna
- School of Medicine Le Kremlin‐BicêtreHôpital Marie Lannelongue, INSERM UMRS 999Université Paris‐SaclayLe Plessis‐RobinsonFrance
| | - Bernard Klonjkowski
- UMR VirologieINRAEANSESÉcole Nationale Vétérinaire d'AlfortMaisons‐AlfortFrance
| | | | - Ralf Jockers
- Institut CochinINSERMCNRSUniversité de ParisParisFrance
| | - Julie Dam
- Institut CochinINSERMCNRSUniversité de ParisParisFrance
| |
Collapse
|
192
|
Role of Melatonin in the Management of Sleep and Circadian Disorders in the Context of Psychiatric Illness. Curr Psychiatry Rep 2022; 24:623-634. [PMID: 36227449 PMCID: PMC9633504 DOI: 10.1007/s11920-022-01369-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW We present a review of research on the role of melatonin in the management of sleep and circadian disorders, stressing current overall view of the knowledge across psychiatric disorders. RECENT FINDINGS Dysregulation of sleep and circadian rhythms has been established in several psychiatric and neurocognitive disorders for long. Recent research confirms this finding consistently across disorders. The secretion of melatonin in schizophrenia and neurocognitive disorders is reduced due to a smaller volume and enlarged calcification of the pineal gland. On the other hand, melatonin dysregulation in bipolar disorder may be more dynamic and caused by light-sensitive melatonin suppression and delayed melatonin secretion. In both cases, exogenous melatonin seems indicated to correct the dysfunction. However, a very limited number of well-designed trials with melatonin to correct sleep and circadian rhythms exist in psychiatric disorders, and the evidence for efficacy is robust only in autism, attention deficit hyperactivity disorder (ADHD), and neurocognitive disorders. This topic has mainly not been of interest for recent work and well-designed trials with objective circadian parameters are few. Overall, recent studies in psychiatric disorders reported that melatonin can be effective in improving sleep parameters such as sleep onset latency, sleep efficiency, and sleep quality. Recent meta-analysis suggests that optimal dosage and dosing time might be important to maximize the efficacy of melatonin. The knowledge base is sufficient to propose well-designed, larger trials with circadian parameters as inclusion and outcome criteria. Based on the partly fragmentary information, we propose testing efficacy in disorders with neurocognitive etiopathology with later and higher dosing, and affective and anxiety disorders with lower and earlier dosing of melatonin. Melatonin is promising for the correction of sleep and circadian abnormalities in psychiatric disorders. However, research results on its effect are still few and need to be accumulated. For effective use of melatonin, it is necessary to consider the appropriate dosage and administration time, depending on the individual abnormality of sleep and circadian rhythms.
Collapse
|
193
|
Nikolaev G, Robeva R, Konakchieva R. Membrane Melatonin Receptors Activated Cell Signaling in Physiology and Disease. Int J Mol Sci 2021; 23:ijms23010471. [PMID: 35008896 PMCID: PMC8745360 DOI: 10.3390/ijms23010471] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023] Open
Abstract
The pineal hormone melatonin has attracted great scientific interest since its discovery in 1958. Despite the enormous number of basic and clinical studies the exact role of melatonin in respect to human physiology remains elusive. In humans, two high-affinity receptors for melatonin, MT1 and MT2, belonging to the family of G protein-coupled receptors (GPCRs) have been cloned and identified. The two receptor types activate Gi proteins and MT2 couples additionally to Gq proteins to modulate intracellular events. The individual effects of MT1 and MT2 receptor activation in a variety of cells are complemented by their ability to form homo- and heterodimers, the functional relevance of which is yet to be confirmed. Recently, several melatonin receptor genetic polymorphisms were discovered and implicated in pathology-for instance in type 2 diabetes, autoimmune disease, and cancer. The circadian patterns of melatonin secretion, its pleiotropic effects depending on cell type and condition, and the already demonstrated cross-talks of melatonin receptors with other signal transduction pathways further contribute to the perplexity of research on the role of the pineal hormone in humans. In this review we try to summarize the current knowledge on the membrane melatonin receptor activated cell signaling in physiology and pathology and their relevance to certain disease conditions including cancer.
Collapse
Affiliation(s)
- Georgi Nikolaev
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 1504 Sofia, Bulgaria;
- Correspondence:
| | - Ralitsa Robeva
- Department of Endocrinology, Faculty of Medicine, Medical University, 1431 Sofia, Bulgaria;
| | - Rossitza Konakchieva
- Faculty of Biology, Sofia University “St. Kliment Ohridski”, 1504 Sofia, Bulgaria;
| |
Collapse
|
194
|
Palagini L, Miniati M, Riemann D, Zerbinati L. Insomnia, Fatigue, and Depression: Theoretical and Clinical Implications of a Self-reinforcing Feedback Loop in Cancer. Clin Pract Epidemiol Ment Health 2021; 17:257-263. [PMID: 35444704 PMCID: PMC8985470 DOI: 10.2174/1745017902117010257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 08/01/2021] [Accepted: 08/09/2021] [Indexed: 11/22/2022]
Abstract
Introduction:
Insomnia is emerging as a modifiable major risk factor for mental and physical problems, including cancer, and it may contribute to cancer-related fatigue and depression. Since both fatigue and depression may favor insomnia as well, we may hypothesize a self-reinforcing feedback loop among these factors in cancer.
Methods:
With the aim of discussing this hypothesis, PubMed, PsycINFO, and Embase electronic databases were searched for literature published according to the PRISMA method with several combinations of terms such as “insomnia” and “cancer” and “fatigue” and “depression”. On this basis, we conducted a narrative review about theoretical aspects of insomnia in the context of cancer and about its role in cancer-related fatigue and depression.
Results:
Twenty-one papers were selected according to inclusion/exclusion criteria. Insomnia is frequent in cancer, and it is associated with cancer-related comorbid conditions such as emotional distress, depressive symptoms, and cancer-related fatigue. The hyperactivation of stress and inflammatory systems, which sustain insomnia, may contribute to cancer-related depression and fatigue. A deleterious feedback loop may be created, and it may perpetuate not only insomnia but also these cancer-related comorbid conditions.
Conclusion:
Although the understanding of the causal relationship between insomnia/ depression/fatigue in individuals with cancer is limited, we may hypothesize that these symptoms can exacerbate and maintain each other. When insomnia is established in cancer, it may lead to a vicious cycle with fatigue and depression and may contribute to adverse cancer outcomes. Interventions targeting insomnia could provide a promising approach not only for insomnia but also for cancer-related symptoms among cancer patients.
Collapse
|
195
|
Elisi GM, Scalvini L, Lodola A, Mor M, Rivara S. Free-Energy Simulations Support a Lipophilic Binding Route for Melatonin Receptors. J Chem Inf Model 2021; 62:210-222. [PMID: 34932329 PMCID: PMC8757440 DOI: 10.1021/acs.jcim.1c01183] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
![]()
The effects of the
neurohormone melatonin are mediated by the activation
of the GPCRs MT1 and MT2 in a variety of tissues.
Crystal structures suggest ligand access to the orthosteric binding
site of MT1 and MT2 receptors through a lateral
channel between transmembrane (TM) helices IV and V. We investigated
the feasibility of this lipophilic entry route for 2-iodomelatonin,
a nonselective agonist with a slower dissociation rate from the MT2 receptor, applying enhanced sampling simulations and free-energy
calculations. 2-Iodomelatonin unbinding was investigated with steered
molecular dynamics simulations which revealed different trajectories
passing through the gap between TM helices IV and V for both receptors.
For one of these unbinding trajectories from the MT1 receptor,
an umbrella-sampling protocol with path-collective variables provided
a calculated energy barrier consistent with the experimental dissociation
rate. The side-chain flexibility of Tyr5.38 was significantly different
in the two receptor subtypes, as assessed by metadynamics simulations,
and during ligand unbinding it frequently assumes an open conformation
in the MT1 but not in the MT2 receptor, favoring
2-iodomelatonin egress. Taken together, our simulations are consistent
with the possibility that the gap between TM IV and V is a way of
connecting the orthosteric binding site and the membrane core for
lipophilic melatonin receptor ligands. Our simulations also suggest
that the open state of Tyr5.38 generates a small pocket on the surface
of MT1 receptor, which could participate in the recognition
of MT1-selective ligands and may be exploited in the design
of new selective compounds.
Collapse
Affiliation(s)
- Gian Marco Elisi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I-43124 Parma, Italy
| | - Laura Scalvini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I-43124 Parma, Italy
| | - Alessio Lodola
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I-43124 Parma, Italy
| | - Marco Mor
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I-43124 Parma, Italy.,Microbiome Research Hub, University of Parma, I-43124 Parma, Italy
| | - Silvia Rivara
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Parco Area delle Scienze 27/A, I-43124 Parma, Italy
| |
Collapse
|
196
|
Huang YS, Lo CH, Tsai PH, Hou YC, Chang YT, Guo CY, Hsieh HY, Lu KC, Shih HM, Wu CC. Downregulation of AANAT by c-Fos in tubular epithelial cells with membranous nephropathy. Biochem Biophys Res Commun 2021; 584:32-38. [PMID: 34763165 DOI: 10.1016/j.bbrc.2021.10.079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/27/2021] [Accepted: 10/30/2021] [Indexed: 10/19/2022]
Abstract
Melatonin is a hormone majorly secreted by the pineal gland and contributes to a various type of physiological functions in mammals. The melatonin production is tightly limited to the AANAT level, yet the most known molecular mechanisms underlying AANAT gene transcription is limited in the pinealocyte. Here, we find that c-Fos and cAMP-response element-binding protein (CREB) decreases and increases the AANAT transcriptional activity in renal tubular epithelial cell, respectively. Notably, c-Fos knockdown significantly upregulates melatonin levels in renal tubular cells. Functional results indicate that AANAT expression is decreased by c-Fos and resulted in enhancement of cell damage in albumin-injury cell model. We further find an inverse correlation between c-Fos and AANAT levels in renal tubular cells from experimental membranous nephropathy (MN) samples and clinical MN specimens. Our finding provides the molecular basis of c-Fos in transcriptionally downregulating expression of AANAT and melatonin, and elucidate the protective role of AANAT in preventing renal tubular cells death in albumin-injury cell model and MN progression.
Collapse
Affiliation(s)
- Yen-Sung Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
| | - Chang-Han Lo
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital Penghu Branch, National Defense Medical Center, Penghu County, 88056, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Ping-Huang Tsai
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Yi-Chou Hou
- Division of Nephrology, Department of Medicine, Cardinal-Tien Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City, 24205, Taiwan.
| | - Yu-Tien Chang
- School of Public Health, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Cheng-Yi Guo
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Hsin-Yi Hsieh
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, 23142, Taiwan.
| | - Hsiu-Ming Shih
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
| | - Chia-Chao Wu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan; Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, 11490, Taiwan.
| |
Collapse
|
197
|
Wang YQ, Jiang YJ, Zou MS, Liu J, Zhao HQ, Wang YH. Antidepressant actions of melatonin and melatonin receptor agonist: Focus on pathophysiology and treatment. Behav Brain Res 2021; 420:113724. [PMID: 34929236 DOI: 10.1016/j.bbr.2021.113724] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/15/2021] [Accepted: 12/15/2021] [Indexed: 12/27/2022]
Abstract
Depression has become one of the most commonly prevalent neuropsychiatric disorders, and the main characteristics of depression are sleep disorders and melatonin secretion disorders caused by circadian rhythm disorders. Abnormal endogenous melatonin alterations can contribute to the occurrence and development of depression. However, molecular mechanisms underlying this abnormality remain ambiguous. The present review summarizes the mechanisms underlying the antidepressant effects of melatonin, which is related to its functions in the regulation of the hypothalamic-pituitary-adrenal axis, inhibition of neuroinflammation, inhibition of oxidative stress, alleviation of autophagy, and upregulation of neurotrophic, promotion of neuroplasticity and upregulation of the levels of neurotransmitters, etc. Also, melatonin receptor agonists, such as agomelatine, ramelteon, piromelatine, tasimelteon, and GW117, have received considerable critical attention and are highly implicated in treating depression and comorbid disorders. This review focuses on melatonin and various melatonin receptor agonists in the pathophysiology and treatment of depression, aiming to provide further insight into the pathogenesis of depression and explore potential targets for novel agent development.
Collapse
Affiliation(s)
- Ye-Qing Wang
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Ya-Jie Jiang
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Man-Shu Zou
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jian Liu
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Hong-Qing Zhao
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Yu-Hong Wang
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
198
|
NRM 2021 Abstract Booklet. J Cereb Blood Flow Metab 2021; 41:11-309. [PMID: 34905986 PMCID: PMC8851538 DOI: 10.1177/0271678x211061050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
199
|
Phiboonchaiyanan PP, Puthongking P, Chawjarean V, Harikarnpakdee S, Sukprasansap M, Chanvorachote P, Priprem A, Govitrapong P. Melatonin and its derivative disrupt cancer stem-like phenotypes of lung cancer cells via AKT downregulation. Clin Exp Pharmacol Physiol 2021; 48:1712-1723. [PMID: 34396568 DOI: 10.1111/1440-1681.13572] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/28/2021] [Accepted: 08/11/2021] [Indexed: 12/27/2022]
Abstract
Cancer stem cells (CSCs), a small subpopulation of tumour cells, have properties of self-renewal and multipotency, which drive cancer progression and resistance to current treatments. Compounds potentially targeting CSCs have been recently developed. This study shows how melatonin, an endogenous hormone synthesised by the pineal gland, and its derivative suppress CSC-like phenotypes of human non-small cell lung cancer (NSCLC) cell lines, H460, H23, and A549. The effects of MLT and its derivative, acetyl melatonin (ACT), on CSC-like phenotypes were investigated using assays for anchorage-independent growth, three-dimensional spheroid formation, scratch wound healing ability, and CSC marker and upstream protein signalling expression. Enriched CSC spheroids were used to confirm the effect of both compounds on lung cancer cells. MLT and ACT inhibited CSC-like behaviours by suppression of colony and spheroid formation in NSCLC cell lines. Their effects on spheroid formation were confirmed in CSC-enriched H460 cells. CSC markers, CD133 and ALDH1A1, were depleted by both compounds. The behaviour and factors associated to epithelial-mesenchymal transition, as indicated by cell migration and the protein vimentin, were also decreased by MLT and ACT. Mechanistically, MLT and ACT decreased the expression of stemness proteins Oct-4, Nanog, and β-catenin by reducing active AKT (phosphorylated AKT). Suppression of the AKT pathway was not mediated through melatonin receptors. This study demonstrates a novel role, and its underlying mechanism, for MLT and its derivative ACT in suppression of CSC-like phenotypes in NSCLC cells, indicating that they are potential candidates for lung cancer treatment.
Collapse
Affiliation(s)
- Preeyaporn Plaimee Phiboonchaiyanan
- College of Pharmacy, Rangsit University, Pathumthani, Thailand
- Cosmeceutical Research, Development and Testing Center, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Ploenthip Puthongking
- Melatonin Research Group, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Verisa Chawjarean
- College of Pharmacy, Rangsit University, Pathumthani, Thailand
- Cosmeceutical Research, Development and Testing Center, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Saraporn Harikarnpakdee
- College of Pharmacy, Rangsit University, Pathumthani, Thailand
- Cosmeceutical Research, Development and Testing Center, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Monruedee Sukprasansap
- Food Toxicology Unit, Institute of Nutrition, Mahidol University, Nakhon Pathom, Thailand
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Aroonsri Priprem
- Faculty of Pharmacy, Mahasarakham University, Maha Sarakham, Thailand
| | | |
Collapse
|
200
|
Melatonin and Pathological Cell Interactions: Mitochondrial Glucose Processing in Cancer Cells. Int J Mol Sci 2021; 22:ijms222212494. [PMID: 34830375 PMCID: PMC8621753 DOI: 10.3390/ijms222212494] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/06/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022] Open
Abstract
Melatonin is synthesized in the pineal gland at night. Since melatonin is produced in the mitochondria of all other cells in a non-circadian manner, the amount synthesized by the pineal gland is less than 5% of the total. Melatonin produced in mitochondria influences glucose metabolism in all cells. Many pathological cells adopt aerobic glycolysis (Warburg effect) in which pyruvate is excluded from the mitochondria and remains in the cytosol where it is metabolized to lactate. The entrance of pyruvate into the mitochondria of healthy cells allows it to be irreversibly decarboxylated by pyruvate dehydrogenase (PDH) to acetyl coenzyme A (acetyl-CoA). The exclusion of pyruvate from the mitochondria in pathological cells prevents the generation of acetyl-CoA from pyruvate. This is relevant to mitochondrial melatonin production, as acetyl-CoA is a required co-substrate/co-factor for melatonin synthesis. When PDH is inhibited during aerobic glycolysis or during intracellular hypoxia, the deficiency of acetyl-CoA likely prevents mitochondrial melatonin synthesis. When cells experiencing aerobic glycolysis or hypoxia with a diminished level of acetyl-CoA are supplemented with melatonin or receive it from another endogenous source (pineal-derived), pathological cells convert to a more normal phenotype and support the transport of pyruvate into the mitochondria, thereby re-establishing a healthier mitochondrial metabolic physiology.
Collapse
|