1
|
Sharma G, Wade Q, Graziane NM. Evaluating remyelination compounds for new applications in opioid use disorder management. J Addict Dis 2025:1-17. [PMID: 39834150 DOI: 10.1080/10550887.2025.2452691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Opioid use disorder (OUD) is associated with a reduction in brain white matter, affecting critical areas involved in decision-making, impulse control, and reward processing. The FDA has approved several drugs and natural compounds that enhance myelination, targeting oligodendrocyte progenitor cells (OPCs), directly enhancing oligodendrocyte (OL) function, or acting as cofactors for myelin production. This retrospective case study aimed to assess whether current clinical evidence supports the use of myelin-enhancing agents to promote remission in OUD. We evaluated a range of compounds with demonstrated effects on myelination, including muscarinic antagonists, cholesterol and lipid homeostatic agents, anti-aging drugs, immunomodulatory agents, anti-inflammatory medications, and others (25 medications in total), as well as 17 vitamins and supplements. Buprenorphine and methadone were used as positive controls. Sequential analyses were performed to identify individual drugs driving significant changes in remission rates (p ≤ 0.01; N ≥ 3,000) and their effects across age, sex, and Body Mass Index (BMI) categories. Three key findings emerged: (1) melatonin improved remission rates in males but showed no effect in females; (2) ibuprofen significantly increased remission rates, particularly in individuals aged 20-39 and 40-59 years; and (3) thiamin was associated with decreased remission rates in males and individuals with a BMI ranging from normal weight to obese. Additionally, buprenorphine and methadone were confirmed as effective in promoting remission. These findings highlight the importance of personalized medicine in treating OUD and suggest that further research is needed to explore individualized treatment strategies based on sex, age, and BMI.
Collapse
Affiliation(s)
- Gouri Sharma
- Department of Anesthesiology and Perioperative Medicine, Hershey, PA, USA
| | - Quinn Wade
- Department of Anesthesiology and Perioperative Medicine, Hershey, PA, USA
| | - Nicholas M Graziane
- Departments of Anesthesiology and Perioperative Medicine and Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
2
|
Song Y, Yoon M. Melatonin effects on animal behavior: circadian rhythm, stress response, and modulation of behavioral patterns. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2025; 67:1-16. [PMID: 39974791 PMCID: PMC11833209 DOI: 10.5187/jast.2024.e105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/26/2024] [Accepted: 10/29/2024] [Indexed: 02/21/2025]
Abstract
Melatonin plays a crucial role in various behavioral and physiological aspects of animals, including regulating their circadian rhythms. This review provides a comprehensive evaluation of the multifaceted effects of melatonin on animal behavior, such as temperament, stress, and aggression regulation. The focus is on the complex interactions between melatonin and the hormonal and neurotransmitter systems, highlighting how melatonin interacts with cortisol, serotonin, and dopamine to influence behavior. Additionally, it investigates the effects of melatonin on the hypothalamic-pituitary-gonada (HPG) axis and stress responses, emphasizing its potential to improve stress management and social interactions, thereby enhancing animal welfare. The review also examines the seasonal variations of melatonin and its impact on aggression and reproductive activities related to photoperiods, as well as its effects on learning and memory to suggest improvements in animal training methods and practices. Furthermore, it discusses the influence of melatonin on appetite and physical activity regulation, implying its involvement in metabolic processes. In conclusion, further research is needed to elucidate the complex mechanisms underlying the extensive influence of melatonin on animal behavior. Through this review, the aim is to integrate the overall knowledge about melatonin and animal behavioral temperament and to propose new research areas for animal management based on behavioral and hormonal regulation.
Collapse
Affiliation(s)
- Yubin Song
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea
| | - Minjung Yoon
- Department of Animal Science and
Biotechnology, Kyungpook National University, Sangju 37224,
Korea
- Department of Horse, Companion and Wild
Animal Science, Kyungpook National University, Sangju 37224,
Korea
- Research Institute for Innovative Animal
Science, Kyungpook National University, Sangju 37224,
Korea
| |
Collapse
|
3
|
Mahmood NMS, Mahmud AM, Maulood IM. The roles of angiotensin-converting enzyme 2 inhibitor, melatonin and its agonist on angiotensin II reactivity in intact and denuded rat aortic rings. J Recept Signal Transduct Res 2024; 44:35-40. [PMID: 38666646 DOI: 10.1080/10799893.2024.2345907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND The pineal product melatonin (MEL) modulates blood vessels through G protein-coupled receptors (GPCRs) called melatonin type 1 receptor (MT1R) and melatonin type 2 receptor (MT2R), in that order. The renin-angiotensin system (RAS), which breaks down angiotensin II (Ang II) to create Ang 1-7, is thought to be mostly controlled by angiotensin-converting enzyme-2 (ACE2). AIM The current work examines the involvement of ACE2 inhibitor, MEL, and ramelteon (RAM) in the vascular response to Ang II activities in the endothelial denuded (E-) and intact (E+) rat isolated thoracic aortic rings. METHOD The isometric tension was measured to evaluate the vascular Ang II contractility using dose response curve (DRC). RESULTS MEL and RAM caused a rightward shift of Ang II in endothelium E + and endothelium E- aorta. CONCLUSION According to the current study, the distribution of MEL receptors and the endothelium's condition are related to the vasomodulatory effect of MEL and ACE2 on Ang II attenuation. These physiological interactions can control vascular tone and increase Ang II reactivity denude endothelial layaer.
Collapse
Affiliation(s)
| | - Almas Mr Mahmud
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Ismail M Maulood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| |
Collapse
|
4
|
Di W, Jin Z, Lei W, Liu Q, Yang W, Zhang S, Lu C, Xu X, Yang Y, Zhao H. Protection of melatonin treatment and combination with traditional antibiotics against septic myocardial injury. Cell Mol Biol Lett 2023; 28:35. [PMID: 37101253 PMCID: PMC10134561 DOI: 10.1186/s11658-022-00415-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 12/23/2022] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Heart failure is a common complication of sepsis with a high mortality rate. It has been reported that melatonin can attenuate septic injury due to various properties. On the basis of previous reports, this study will further explore the effects and mechanisms of melatonin pretreatment, posttreatment, and combination with antibiotics in the treatment of sepsis and septic myocardial injury. METHODS AND RESULTS Our results showed that melatonin pretreatment showed an obvious protective effect on sepsis and septic myocardial injury, which was related to the attenuation of inflammation and oxidative stress, the improvement of mitochondrial function, the regulation of endoplasmic reticulum stress (ERS), and the activation of the AMPK signaling pathway. In particular, AMPK serves as a key effector for melatonin-initiated myocardial benefits. In addition, melatonin posttreatment also had a certain degree of protection, while its effect was not as remarkable as that of pretreatment. The combination of melatonin and classical antibiotics had a slight but limited effect. RNA-seq detection clarified the cardioprotective mechanism of melatonin. CONCLUSION Altogether, this study provides a theoretical basis for the application strategy and combination of melatonin in septic myocardial injury.
Collapse
Affiliation(s)
- Wencheng Di
- Department of Cardiovascular Medicine, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, 29 Bulan Road, Shenzhen, Guangdong Province, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Airforce Military Medical University, 127 Changle West Road, Xi'an, China
| | - Wangrui Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Qiong Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Wenwen Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Shaofei Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Chenxi Lu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Xiaoling Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi'an, China.
- Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, China.
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, The Airforce Military Medical University, 1 Xinsi Road, Xi'an, China.
| |
Collapse
|
5
|
Liao XX, Wu XY, Zhou YL, Li JJ, Wen YL, Zhou JJ. Gut microbiome metabolites as key actors in atherosclerosis co-depression disease. Front Microbiol 2022; 13:988643. [PMID: 36439791 PMCID: PMC9686300 DOI: 10.3389/fmicb.2022.988643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/24/2022] [Indexed: 02/26/2024] Open
Abstract
Cardiovascular diseases, mainly characterized by atherosclerosis (AS), and depression have a high comorbidity rate. However, previous studies have been conducted under a single disease, and there is a lack of studies in comorbid states to explore the commonalities in the pathogenesis of both diseases. Modern high-throughput technologies have made it clear that the gut microbiome can affect the development of the host's own disorders and have shown that their metabolites are crucial to the pathophysiology of AS and depression. The aim of this review is to summarize the current important findings on the role of gut microbiome metabolites such as pathogen-associated molecular patterns, bile acids, tryptophan metabolites, short-chain fatty acids, and trimethylamine N -oxide in depression and AS disease, with the aim of identifying potential biological targets for the early diagnosis and treatment of AS co-depression disorders.
Collapse
Affiliation(s)
- Xing-Xing Liao
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - Xiao-Yun Wu
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yu-Long Zhou
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - Jia-Jun Li
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - You-Liang Wen
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
| | - Jun-Jie Zhou
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
6
|
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: 52] [Impact Index Per Article: 17.3] [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
|
7
|
The effect of melatonin on the mouse ameloblast-lineage cell line ALCs. Sci Rep 2022; 12:8225. [PMID: 35581244 PMCID: PMC9114102 DOI: 10.1038/s41598-022-11912-3] [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: 02/02/2022] [Accepted: 04/13/2022] [Indexed: 01/17/2023] Open
Abstract
Melatonin plays a critical role in promoting the proliferation of osteoblasts and the growth and development of dental papilla cells. However, the effect and mechanism of melatonin on the growth and development of ALCs still need to be explored. CCK8 assay was used for the evaluation of cell numbers. qRT-PCR was used to identify the differentially expressed genes in ALCs after melatonin treatment. The number and morphology of ALCs were investigated by confocal microscopy. Alkaline phosphatase assay and Alizarin red S staining were used for measuring mineralization. Then, we focused on observing the crucial factors of the signaling pathway by RNA-seq and qRT-PCR. Melatonin limited the cell number of ALCs in a dose-dependent manner and promoted the production of actin fibers. A high concentration of melatonin significantly promoted the mRNA levels of enamel matrix proteins and the formation of mineralized nodules. RNA-seq data showed that Wnt signaling pathway may be involved in the differentiation of ALCs under the influence of melatonin. This study suggests that melatonin plays a regulatory role in the cell number, differentiation, and mineralization of the ALCs, and then shows the relationship between the Wnt signaling pathway with the ALCs under melatonin.
Collapse
|
8
|
Early Treatment of Acute Myocardial Infarction with Melatonin: Effects on MMP-9 and Adverse Cardiac Events. J Clin Med 2022; 11:jcm11071909. [PMID: 35407517 PMCID: PMC9000067 DOI: 10.3390/jcm11071909] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/23/2022] [Accepted: 03/27/2022] [Indexed: 01/04/2023] Open
Abstract
Background: Matrix metalloproteinase-9 (MMP-9) is crucial in tissue remodeling after an adverse cardiac event. In experimental studies, melatonin has been found to attenuate MMP-9 activation. The present study assessed the effects of systemic melatonin administration on the prognosis of patients with acute myocardial infarction (AMI) successfully treated with primary percutaneous coronary intervention, and to examine the effects on MMP-9 levels. Methods: We conducted a randomized controlled trial, enrolling patients who underwent primary percutaneous coronary intervention due to AMI. They were assigned to two groups for melatonin or placebo. The primary endpoint was a combined event of mortality and heart failure readmission at 2 years. The secondary endpoint was the levels of MMP-9 after the percutaneous coronary intervention. Results: Ninety-four patients were enrolled, 45 in the melatonin group and 49 in the control group. At 2 years of follow-up, 13 (13.8%) patients suffered the primary endpoint (3 deaths and 10 readmissions due to heart failure), 3 patients in the melatonin group and 10 in the placebo group. The difference in the restricted mean survival time was 87.5 days (p = 0.02); HR = 0.3 (95% CI 0.08–1.08; p = 0.06); Log-rank test 0.04. After controlling for confounding variables, melatonin administration reduced MMP-9 levels to 90 ng/mL (95% CI 77.3–102.6). Conclusions: This pilot study demonstrated that compared to placebo, melatonin administration was associated with better outcomes in AMI patients undergoing primary percutaneous coronary intervention.
Collapse
|
9
|
Wang D, Peng P, Dudek M, Hu X, Xu X, Shang Q, Wang D, Jia H, Wang H, Gao B, Zheng C, Mao J, Gao C, He X, Cheng P, Wang H, Zheng J, Hoyland JA, Meng QJ, Luo Z, Yang L. Restoring the dampened expression of the core clock molecule BMAL1 protects against compression-induced intervertebral disc degeneration. Bone Res 2022; 10:20. [PMID: 35217644 PMCID: PMC8881495 DOI: 10.1038/s41413-022-00187-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 11/09/2022] Open
Abstract
The circadian clock participates in maintaining homeostasis in peripheral tissues, including intervertebral discs (IVDs). Abnormal mechanical loading is a known risk factor for intervertebral disc degeneration (IDD). Based on the rhythmic daily loading pattern of rest and activity, we hypothesized that abnormal mechanical loading could dampen the IVD clock, contributing to IDD. Here, we investigated the effects of abnormal loading on the IVD clock and aimed to inhibit compression-induced IDD by targeting the core clock molecule brain and muscle Arnt-like protein-1 (BMAL1). In this study, we showed that BMAL1 KO mice exhibit radiographic features similar to those of human IDD and that BMAL1 expression was negatively correlated with IDD severity by systematic analysis based on 149 human IVD samples. The intrinsic circadian clock in the IVD was dampened by excessive loading, and BMAL1 overexpression by lentivirus attenuated compression-induced IDD. Inhibition of the RhoA/ROCK pathway by Y-27632 or melatonin attenuated the compression-induced decrease in BMAL1 expression. Finally, the two drugs partially restored BMAL1 expression and alleviated IDD in a diurnal compression model. Our results first show that excessive loading dampens the circadian clock of nucleus pulposus tissues via the RhoA/ROCK pathway, the inhibition of which potentially protects against compression-induced IDD by preserving BMAL1 expression. These findings underline the importance of the circadian clock for IVD homeostasis and provide a potentially effective therapeutic strategy for IDD.
Collapse
Affiliation(s)
- Dong Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Pandi Peng
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.,Medical Research Institute, Northwestern Polytechnical University, Xi'an, 710068, People's Republic of China
| | - Michal Dudek
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK.,Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester, M13 9PL, UK
| | - Xueyu Hu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Xiaolong Xu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Qiliang Shang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Di Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Haoruo Jia
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Han Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Bo Gao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Chao Zheng
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jianxin Mao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Chu Gao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Xin He
- Department of Medicine Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Pengzhen Cheng
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Huanbo Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jianmin Zheng
- Radiology Department, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Judith A Hoyland
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Qing-Jun Meng
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK.,Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester, M13 9PL, UK
| | - Zhuojing Luo
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China. .,Medical Research Institute, Northwestern Polytechnical University, Xi'an, 710068, People's Republic of China.
| | - Liu Yang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China. .,Medical Research Institute, Northwestern Polytechnical University, Xi'an, 710068, People's Republic of China.
| |
Collapse
|
10
|
Veiga ECDA, Simões RDS, Caviola LL, Abreu LC, Cavalli RC, Cipolla-Neto J, Baracat EC, Soares JM. Melatonin and the cardiovascular system in animals: systematic review and meta-analysis. Clinics (Sao Paulo) 2021; 76:e2863. [PMID: 34644731 PMCID: PMC8478132 DOI: 10.6061/clinics/2021/e2863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/16/2021] [Indexed: 12/09/2022] Open
Abstract
Melatonin, a hormone released by the pineal gland, demonstrates several effects on the cardiovascular system. Herein, we performed a systematic review and meta-analysis to verify the effects of melatonin in an experimental model of myocardial infarction. We performed a systematic review according to PRISMA recommendations and reviewed MEDLINE, Embase, and Cochrane databases. Only articles in English were considered. A systematic review of the literature published between November 2008 and June 2019 was performed. The meta-analysis was conducted using the RevMan 5.3 program provided by the Cochrane Collaboration. In total, 858 articles were identified, of which 13 were included in this review. The main results of this study revealed that melatonin benefits the cardiovascular system by reducing infarct size, improving cardiac function according to echocardiographic and hemodynamic analyses, affords antioxidant effects, improves the rate of apoptosis, decreases lactate dehydrogenase activity, enhances biometric analyses, and improves protein levels, as analyzed by western blotting and quantitative PCR. In the meta-analysis, we observed a statistically significant decrease in infarct size (mean difference [MD], -20.37 [-23.56, -17.18]), no statistical difference in systolic pressure (MD, -1.75 [-5.47, 1.97]), a statistically significant decrease in lactate dehydrogenase in animals in the melatonin group (MD, -4.61 [-6.83, -2.40]), and a statistically significant improvement in the cardiac ejection fraction (MD, -8.12 [-9.56, -6.69]). On analyzing potential bias, we observed that most studies presented a low risk of bias; two parameters were not included in the analysis, and one parameter had a high risk of bias. Melatonin exerts several effects on the cardiovascular system and could be a useful therapeutic target to combat various cardiovascular diseases.
Collapse
Affiliation(s)
- Eduardo Carvalho de Arruda Veiga
- Departamento de Obstetricia e Ginecologia, Hospital das Clinicas HCFMRP-USP, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, SP, BR
| | - Ricardo dos Santos Simões
- Departamento de Obstetricia e Ginecologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Leonardo L. Caviola
- Departamento de Obstetricia e Ginecologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Luiz Carlos Abreu
- Disciplina de escrita cientifica, Faculdade de Medicina do ABC, Santo Andre, SP, BR
| | - Ricardo Carvalho Cavalli
- Departamento de Obstetricia e Ginecologia, Hospital das Clinicas HCFMRP-USP, Faculdade de Medicina de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, SP, BR
| | - José Cipolla-Neto
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas (ICB-USP), Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Edmund Chada Baracat
- Departamento de Obstetricia e Ginecologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - José Maria Soares
- Departamento de Obstetricia e Ginecologia, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
11
|
Melatonin Improves Levels of Zn and Cu in the Muscle of Diabetic Obese Rats. Pharmaceutics 2021; 13:pharmaceutics13101535. [PMID: 34683825 PMCID: PMC8539996 DOI: 10.3390/pharmaceutics13101535] [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: 08/25/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
Melatonin improves metabolic alterations associated with obesity and its diabetes (diabesity). We intend to determine whether this improvement is exerted by changing Zn and/or Cu tissue levels in liver, muscle, pancreas, and brain, and in internal (perirenal, perigonadal, and omentum) and subcutaneous lumbar white adipose tissues (IWAT and SWAT, respectively). Male Zücker diabetic fatty (ZDF) rats and lean littermates (ZL) were orally supplemented either with melatonin (10 mg/kg body weight/day) or vehicle for 6 weeks. Zn and Cu concentrations were not significantly influenced by diabesity in the analyzed tissues (p > 0.05), with the exception of Zn in liver. In skeletal muscle Zn and Cu, and in perirenal WAT, only Zn levels increased significantly with melatonin supplementation in ZDF rats (p < 0.05). This cytoplasmic Zn enhancement would be probably associated with the upregulation of several Zn influx membrane transporters (Zips) and could explain the amelioration in the glycaemia and insulinaemia by upregulating the Akt and downregulating the inhibitor PTP1B, in obese and diabetic conditions. Enhanced Zn and Cu levels in muscle cells could be related to the reported antioxidant melatonin activity exerted by increasing the Zn, Cu-SOD, and extracellular Cu-SOD activity. In conclusion, melatonin, by increasing the muscle levels of Zn and Cu, joined with our previously reported findings improves glycaemia, insulinaemia, and oxidative stress in this diabesity animal model.
Collapse
|
12
|
Alaasam VJ, Liu X, Niu Y, Habibian JS, Pieraut S, Ferguson BS, Zhang Y, Ouyang JQ. Effects of dim artificial light at night on locomotor activity, cardiovascular physiology, and circadian clock genes in a diurnal songbird. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 282:117036. [PMID: 33838441 PMCID: PMC8184626 DOI: 10.1016/j.envpol.2021.117036] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/04/2021] [Accepted: 03/28/2021] [Indexed: 05/17/2023]
Abstract
Artificial light is transforming the nighttime environment and quickly becoming one of the most pervasive pollutants on earth. Across taxa, light entrains endogenous circadian clocks that function to synchronize behavioral and physiological rhythms with natural photoperiod. Artificial light at night (ALAN) disrupts these photoperiodic cues and has consequences for humans and wildlife including sleep disruption, physiological stress and increased risk of cardiovascular disease. However, the mechanisms underlying organismal responses to dim ALAN, resembling light pollution, remain elusive. Light pollution exists in the environment at lower levels (<5 lux) than tested in many laboratory studies that link ALAN to circadian rhythm disruption. Few studies have linked dim ALAN to both the upstream regulators of circadian rhythms and downstream behavioral and physiological consequences. We exposed zebra finches (Taeniopygia gutatta) to dim ALAN (1.5 lux) and measured circadian expression of five pacemaker genes in central and peripheral tissues, plasma melatonin, locomotor activity, and biomarkers of cardiovascular health. ALAN caused an increase in nighttime activity and, for males, cardiac hypertrophy. Moreover, downstream effects were detectable after just short duration exposure (10 days) and at dim levels that mimic the intensity of environmental light pollution. However, ALAN did not affect circulating melatonin nor oscillations of circadian gene expression in the central clock (brain) or liver. These findings suggest that dim ALAN can alter behavior and physiology without strong shifts in the rhythmic expression of molecular circadian pacemakers. Approaches that focus on ecologically-relevant ALAN and link complex biological pathways are necessary to understand the mechanisms underlying vertebrate responses to light pollution.
Collapse
Affiliation(s)
- Valentina J Alaasam
- Department of Biology, University of Nevada, Reno, Reno, NV, USA; Program of Ecology, Evolution, and Conservation Biology, University of Nevada, Reno, Reno, NV, USA.
| | - Xu Liu
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Ye Niu
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Justine S Habibian
- Department of Nutrition, University of Nevada, Reno, Reno, NV, USA; Program of Cellular and Molecular Biology, University of Nevada, Reno, Reno, NV, USA
| | - Simon Pieraut
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Brad S Ferguson
- Department of Nutrition, University of Nevada, Reno, Reno, NV, USA; Center for Biomedical Research Excellence in Molecular and Cellular Signal Transduction in the Cardiovascular System, School of Medicine, University of Nevada, Reno, Reno, NV, USA
| | - Yong Zhang
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| | - Jenny Q Ouyang
- Department of Biology, University of Nevada, Reno, Reno, NV, USA
| |
Collapse
|
13
|
Munmun F, Witt-Enderby PA. Melatonin effects on bone: Implications for use as a therapy for managing bone loss. J Pineal Res 2021; 71:e12749. [PMID: 34085304 DOI: 10.1111/jpi.12749] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/22/2021] [Accepted: 05/31/2021] [Indexed: 02/06/2023]
Abstract
Melatonin is the primary circadian output signal from the brain and is mainly synthesized in pinealocytes. The rhythm and secretion of melatonin are under the control of an endogenous oscillator located in the SCN or the master biological clock. Disruptions in circadian rhythms by shift work, aging, or light at night are associated with bone loss and increased fracture risk. Restoration of nocturnal melatonin peaks to normal levels or therapeutic levels through timed melatonin supplementation has been demonstrated to provide bone-protective actions in various models. Melatonin is a unique molecule with diverse molecular actions targeting melatonin receptors located on the plasma membrane or mitochondria or acting independently of receptors through its actions as an antioxidant or free radical scavenger to stimulate osteoblastogenesis, inhibit osteoclastogenesis, and improve bone density. Its additional actions on entraining circadian rhythms and improving quality of life in an aging population coupled with its safety profile make it an ideal therapeutic candidate for protecting against bone loss in susceptible populations. The intent of this review is to provide a focused discussion on bone loss and disorders of the bone as it relates to melatonin and conditions that modify melatonin levels with the hope that future therapies include those that include melatonin and correct those factors that modify melatonin levels like circadian disruption.
Collapse
Affiliation(s)
- Fahima Munmun
- Division of Pharmaceutical Sciences, Duquesne University School of Pharmacy, Pittsburgh, PA, USA
| | - Paula A Witt-Enderby
- Division of Pharmaceutical Sciences, Duquesne University School of Pharmacy, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Rodrigo R, González-Montero J, Sotomayor CG. Novel Combined Antioxidant Strategy against Hypertension, Acute Myocardial Infarction and Postoperative Atrial Fibrillation. Biomedicines 2021; 9:620. [PMID: 34070760 PMCID: PMC8228412 DOI: 10.3390/biomedicines9060620] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 12/28/2022] Open
Abstract
Reactive oxygen species (ROS) play a physiological role in the modulation of several functions of the vascular wall; however, increased ROS have detrimental effects. Hence, oxidative stress has pathophysiological impacts on the control of the vascular tone and cardiac functions. Recent experimental studies reported the involvement of increased ROS in the mechanism of hypertension, as this disorder associates with increased production of pro-oxidants and decreased bioavailability of antioxidants. In addition, increased ROS exposure is found in ischemia-reperfusion, occurring in acute myocardial infarction and cardiac surgery with extracorporeal circulation, among other settings. Although these effects cause major heart damage, at present, there is no available treatment. Therefore, it should be expected that antioxidants counteract the oxidative processes, thereby being suitable against cardiovascular disease. Nevertheless, although numerous experimental studies agree with this notion, interventional trials have provided mixed results. A better knowledge of ROS modulation and their specific interaction with the molecular targets should contribute to the development of novel multitarget antioxidant effective therapeutic strategies. The complex multifactorial nature of hypertension, acute myocardial infarction, and postoperative atrial fibrillation needs a multitarget antioxidant strategy, which may give rise to additive or synergic protective effects to achieve optimal cardioprotection.
Collapse
Affiliation(s)
- Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, CP 8380453 Santiago, Chile;
| | - Jaime González-Montero
- Basic and Clinical Oncology Department, Faculty of Medicine, University of Chile, CP 8380453 Santiago, Chile;
| | - Camilo G. Sotomayor
- Clinical Hospital University of Chile, University of Chile, CP 8380453 Santiago, Chile
| |
Collapse
|
15
|
Domínguez-Rodríguez A, Abreu-González P, Báez-Ferrer N, Reiter RJ, Avanzas P, Hernández-Vaquero D. Melatonin and Cardioprotection in Humans: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front Cardiovasc Med 2021; 8:635083. [PMID: 34055929 PMCID: PMC8149621 DOI: 10.3389/fcvm.2021.635083] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/06/2021] [Indexed: 02/02/2023] Open
Abstract
Myocardial ischemia/reperfusion (IR) injury represents a critical problem associated with interventional approaches for coronary reperfusion. Pharmacological cardioprotective interventions are advocated to ameliorate IR injury. Melatonin is an anti-inflammatory and antioxidant agent with a wide range of therapeutic properties that may contribute to its cardioprotective effects. No systematic review or meta-analysis has compared melatonin vs. placebo as a cardioprotective agent in humans. The present study, based on a systematic review and meta-analysis, was carried out to assess melatonin's efficacy as a cardioprotective treatment. We performed a systematic review of the available literature. Randomized controlled trials (RCTs) were identified and information was extracted using predefined data extraction forms. The primary outcomes were (a) left ventricular ejection fraction (LVEF) and (b) blood troponin levels in patients who underwent myocardial revascularization and were randomized to melatonin or placebo. The inverse-variance random-effects method was used to pool the estimates. Heterogeneity and publication bias were assessed. Weighted mean differences or standardized mean differences were calculated. A total of 283 records were screened and seven RCTs met all the inclusion criteria. After the pooled analysis, the results on LVEF were consistent across all studies, and a significant heterogeneity was found in the results on troponin levels. The melatonin-treated patients had on average higher LVEF than the placebo-treated individuals with a weighted mean difference = 3.1% (95% CI 0.6-5.5, p = 0.01). Five works compared the levels of troponin after melatonin or placebo treatment. The melatonin-treated patients had lower levels of troponin with a standardized mean difference = -1.76 (95% CI -2.85 to -0.67, p = 0.002). The findings of this meta-analysis revealed that melatonin administration in humans as a cardioprotective agent attenuated heart dysfunction with a favorable effect on the LVEF.
Collapse
Affiliation(s)
- Alberto Domínguez-Rodríguez
- Hospital Universitario de Canarias, Servicio de Cardiología, Tenerife, Spain.,Departamento de Enfermería, Facultad de Ciencias de la Salud, Universidad de La Laguna Tenerife, San Cristóbal de La Laguna, Spain
| | - Pedro Abreu-González
- Departamento de Fisiología, Facultad de Medicina, Universidad de La Laguna, Tenerife, San Cristóbal de La Laguna, Spain
| | - Néstor Báez-Ferrer
- Hospital Universitario de Canarias, Servicio de Cardiología, Tenerife, Spain
| | - Russel J Reiter
- Department of Cell Systems and Anatomy UT Health San Antonio, Long School of Medicine, San Antonio, TX, United States
| | - Pablo Avanzas
- Área del Corazón, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Daniel Hernández-Vaquero
- Área del Corazón, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| |
Collapse
|
16
|
Nicotinamide mononucleotide and melatonin counteract myocardial ischemia-reperfusion injury by activating SIRT3/FOXO1 and reducing apoptosis in aged male rats. Mol Biol Rep 2021; 48:3089-3096. [PMID: 33866495 DOI: 10.1007/s11033-021-06351-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 04/10/2021] [Indexed: 10/21/2022]
Abstract
It has been documented that aging increases the risk of cardiovascular disease including myocardial ischemia/reperfusion (IR) injury and acute myocardial infarction. In this study, we aimed to investigate the individual or combined effects of nicotinamide mononucleotide (NMN) and melatonin (Mel) treatment on apoptotic markers, expression of SIRT3, and FOXO1, and infarct size of the aged myocardium subjected to IR injury. Sixty aged Wistar rats (22-24 months) were assigned to five groups including sham, IR, NMN+IR, Mel+IR, and NMN+Mel+IR (combination therapy). Isolated hearts were exposed to 30-min regional ischemia followed by 60-min reperfusion. NMN (100 mg/kg/day/i.p.) was injected every second day starting on day 28 before IR injury. Melatonin was added to the perfusion solution five minutes prior to and until 15 min after the start of reperfusion. The infarct size was assessed by computerized planimetry. The mRNA levels of SIRT3, FOXO1, and apoptotic genes Bax, Bcl-2, and Caspase-3 were estimated by real-time PCR. All treatments reduced infarct size as compared with the IR group. Melatonin and NMN upregulated the gene expression of Bcl-2, SIRT3, and FOXO1 and downregulated the gene expression of Bax, and Caspase-3, in comparison to the IR group. Also, the protein levels of SIRT3, quantified by Western blotting, were upregulated by the interventions. The effects of combination therapy were significantly greater than those of melatonin or NMN alone. These findings indicate that the combined administration of NMN and melatonin can protect the aged heart against IR injury by decreasing apoptosis and activating the SIRT3/FOXO1 pathway.
Collapse
|
17
|
Buijink MR, Michel S. A multi-level assessment of the bidirectional relationship between aging and the circadian clock. J Neurochem 2021; 157:73-94. [PMID: 33370457 PMCID: PMC8048448 DOI: 10.1111/jnc.15286] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022]
Abstract
The daily temporal order of physiological processes and behavior contribute to the wellbeing of many organisms including humans. The central circadian clock, which coordinates the timing within our body, is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Like in other parts of the brain, aging impairs the SCN function, which in turn promotes the development and progression of aging-related diseases. We here review the impact of aging on the different levels of the circadian clock machinery-from molecules to organs-with a focus on the role of the SCN. We find that the molecular clock is less effected by aging compared to other cellular components of the clock. Proper rhythmic regulation of intracellular signaling, ion channels and neuronal excitability of SCN neurons are greatly disturbed in aging. This suggests a disconnection between the molecular clock and the electrophysiology of these cells. The neuronal network of the SCN is able to compensate for some of these cellular deficits. However, it still results in a clear reduction in the amplitude of the SCN electrical rhythm, suggesting a weakening of the output timing signal. Consequently, other brain areas and organs not only show aging-related deficits in their own local clocks, but also receive a weaker systemic timing signal. The negative spiral completes with the weakening of positive feedback from the periphery to the SCN. Consequently, chronotherapeutic interventions should aim at strengthening overall synchrony in the circadian system using life-style and/or pharmacological approaches.
Collapse
Affiliation(s)
- M. Renate Buijink
- Department of Cellular and Chemical BiologyLaboratory for NeurophysiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Stephan Michel
- Department of Cellular and Chemical BiologyLaboratory for NeurophysiologyLeiden University Medical CenterLeidenthe Netherlands
| |
Collapse
|
18
|
The Possible Effect of Space Weather Factors on Various Physiological Systems of the Human Organism. ATMOSPHERE 2021. [DOI: 10.3390/atmos12030346] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A systematic review of heliobiological studies of the last 25 years devoted to the study of the potential influence of space weather factors on human health and well-being was carried out. We proposed three criteria (coordinates), according to which the work on solar–biospheric relations was systematized: the time scale of data sampling (years, days, hours, minutes); the level of organization of the biological system under study (population, group, individual, body system); and the degree of system response (norm, adaptation, failure of adaptation (illness), disaster (death)). This systematic review demonstrates that three parameters mentioned above are closely related in the existing heliobiological studies: the larger the selected time scale, the higher the level of estimated biological system organization and the stronger the potential response degree is. The long-term studies are devoted to the possible influence of solar activity on population disasters, i.e., significant increases in morbidity and mortality. On a daily scale, a probable effect of geomagnetic storms and other space weather events on short-term local outbreaks of morbidity is shown as well as on cases of deterioration in people functional state. On an intraday scale, in the regular functioning mode, the heart and brain rhythms of healthy people turn to be synchronized with geomagnetic field variations in some frequency ranges, which apparently is the necessary organism’s existence element. The applicability of different space weather indices at different data sampling rates, the need to take into account the contribution of meteorological factors, and the prospects for an individual approach in heliobiology are discussed. The modern important results of experiments on modeling the action of magnetic storms in laboratory conditions and the substantiation of possible theoreical mechanisms are described. These results provide an experimental and theoretical basis for studies of possible connections of space weather and human health.
Collapse
|
19
|
Boutin JA, Jockers R. Melatonin controversies, an update. J Pineal Res 2021; 70:e12702. [PMID: 33108677 DOI: 10.1111/jpi.12702] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/11/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
Abstract
Melatonin was discovered more than 60 years ago. Since then, several seminal discoveries have allowed us to define its function as a neuroendocrine hormone and its molecular targets in mammals and many other species. However, many fundamental issues have not yet been solved such as the subcellular localization of melatonin synthesis and the full spectrum of its molecular targets. In addition, a considerable number of controversies persist in the field, mainly concerning how many functions melatonin has. Altogether, this illustrates how "immature" the field still is. The intention of this opinion article is to note the controversies and limitations in the field, to initiate a discussion and to make proposals/guidelines to overcome them and move the field forward.
Collapse
Affiliation(s)
- Jean A Boutin
- Institut de Recherches Internationales SERVIER, Suresnes Cedex, France
| | - Ralf Jockers
- INSERM, CNRS, Institut Cochin, Université de Paris, Paris, France
| |
Collapse
|
20
|
Potential Role of Melatonin as an Adjuvant for Atherosclerotic Carotid Arterial Stenosis. Molecules 2021; 26:molecules26040811. [PMID: 33557283 PMCID: PMC7914857 DOI: 10.3390/molecules26040811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 12/23/2022] Open
Abstract
Carotid artery stenosis (CAS) is an atherosclerotic disease characterized by a narrowing of the artery lumen and a high risk of ischemic stroke. Risk factors of atherosclerosis, including smoking, hypertension, hyperglycemia, hyperlipidemia, aging, and disrupted circadian rhythm, may potentiate atherosclerosis in the carotid artery and further reduce the arterial lumen. Ischemic stroke due to severe CAS and cerebral ischemic/reperfusion (I/R) injury after the revascularization of CAS also adversely affect clinical outcomes. Melatonin is a pluripotent agent with potent anti-inflammatory, anti-oxidative, and neuroprotective properties. Although there is a shortage of direct clinical evidence demonstrating the benefits of melatonin in CAS patients, previous studies have shown that melatonin may be beneficial for patients with CAS in terms of reducing endothelial damage, stabilizing arterial plaque, mitigating the harm from CAS-related ischemic stroke and cerebral I/R injury, and alleviating the adverse effects of the related risk factors. Additional pre-clinical and clinical are required to confirm this speculation.
Collapse
|
21
|
Marhuenda J, Villaño D, Arcusa R, Zafrilla P. Melatonin in Wine and Beer: Beneficial Effects. Molecules 2021; 26:molecules26020343. [PMID: 33440795 PMCID: PMC7827953 DOI: 10.3390/molecules26020343] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/15/2022] Open
Abstract
Melatonin is a hormone secreted in the pineal gland with several functions, especially regulation of circadian sleep cycle and the biological processes related to it. This review evaluates the bioavailability of melatonin and resulting metabolites, the presence of melatonin in wine and beer and factors that influence it, and finally the different benefits related to treatment with melatonin. When administered orally, melatonin is mainly absorbed in the rectum and the ileum; it has a half-life of about 0.45–1 h and is extensively inactivated in the liver by phase 2 enzymes. Melatonin (MEL) concentration varies from picograms to ng/mL in fermented beverages such as wine and beer, depending on the fermentation process. These low quantities, within a dietary intake, are enough to reach significant plasma concentrations of melatonin, and are thus able to exert beneficial effects. Melatonin has demonstrated antioxidant, anticarcinogenic, immunomodulatory and neuroprotective actions. These benefits are related to its free radical scavenging properties as well and the direct interaction with melatonin receptors, which are involved in complex intracellular signaling pathways, including inhibition of angiogenesis and cell proliferation, among others. In the present review, the current evidence on the effects of melatonin on different pathophysiological conditions is also discussed.
Collapse
|
22
|
Torabi H, Mehdikhani M, Varshosaz J, Shafiee F. An innovative approach to fabricate a thermosensitive melatonin‐loaded conductive pluronic/chitosan hydrogel for myocardial tissue engineering. J Appl Polym Sci 2020. [DOI: 10.1002/app.50327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hadis Torabi
- Department of Biomedical Engineering, Faculty of Engineering University of Isfahan Isfahan Iran
| | - Mehdi Mehdikhani
- Department of Biomedical Engineering, Faculty of Engineering University of Isfahan Isfahan Iran
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center Isfahan University of Medical Sciences Isfahan Iran
- Department of Pharmaceutics School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences Isfahan Iran
| | - Fatemeh Shafiee
- Department of Pharmaceutical Biotechnology School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences Isfahan Iran
| |
Collapse
|
23
|
Wang SS, Wang C, Chen H. MicroRNAs are critical in regulating smooth muscle cell mineralization and apoptosis during vascular calcification. J Cell Mol Med 2020; 24:13564-13572. [PMID: 33089928 PMCID: PMC7754013 DOI: 10.1111/jcmm.16005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/27/2020] [Accepted: 10/01/2020] [Indexed: 02/01/2023] Open
Abstract
Vascular calcification refers to the pathological deposition of calcium and phosphate minerals into the vasculature. It is prevalent in atherosclerosis, ageing, type 2 diabetes mellitus and chronic kidney disease, thus, increasing morbidity and mortality from these conditions. Vascular calcification shares similar mechanisms with bone mineralization, with smooth muscle cells playing a critical role in both processes. In the last decade, a variety of microRNAs have been identified as key regulators for the differentiation, phenotypic switch, proliferation, apoptosis, cytokine production and matrix deposition in vascular smooth muscle cells during vascular calcification. Therefore, this review mainly discusses the roles of microRNAs in the pathophysiological mechanisms of vascular calcification in smooth muscle cells and describes several interventions against vascular calcification by regulating microRNAs. As the exact mechanisms of calcification remain not fully elucidated, having a better understanding of microRNA involvement in vascular calcification may give impetus to development of novel therapeutics for the control and treatment of vascular calcification.
Collapse
Affiliation(s)
- Shan-Shan Wang
- Department of Cardiology, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Wang
- Department of Cardiology, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Han Chen
- Department of Cardiology, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
24
|
Huang H, Li Z, Ruan Y, Feng W, Chen J, Li X, Ouyang L, Huang H. Circadian rhythm disorder: a potential inducer of vascular calcification? J Physiol Biochem 2020; 76:513-524. [PMID: 32945991 DOI: 10.1007/s13105-020-00767-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 09/14/2020] [Indexed: 12/24/2022]
Abstract
Over the past decades, circadian rhythm has drawn a great attention in cardiovascular diseases. The expressions of rhythm genes fluctuate in accordance with the diurnal changes of vascular physiology, which highlights the pivotal effect of vascular clock. Recent researches show that the circadian clock can directly regulate the synthetic and secretory function of endothelial cells and phenotypic switch of vascular smooth muscle cells to adjust vascular relaxation and contraction. Importantly, dysfunction of vascular cells is involved in vascular calcification. Secretion of osteogenic cytokines and calcified vesicles in the vessel, osteogenic phenotype switch of vascular smooth muscle cells are all implicated in the calcification process. Moreover, circadian rhythm disorder can lead to abnormal hormone secretion, oxidative stress, inflammatory reaction, and autophagy, all of which should not be ignored in vascular calcification. Vascular senescence is another pathogenetic mechanism in vascular calcification. Accelerated vascular senescence may act as an important intermediate factor to promote vascular calcification in circadian rhythm disorders. In this review, we elaborate the potential effect of circadian rhythm disorder in vascular calcification and try to provide a new direction in the prevention of vascular calcification.
Collapse
Affiliation(s)
- Haoran Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
- Department of Pediatric Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaohuai Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yuyi Ruan
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weijing Feng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxue Li
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
| | - Liu Ouyang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, No. 3025, Shennan Middle Road, Futian District, Shenzhen, 518000, China.
| |
Collapse
|
25
|
Ao L, Li L, Sun H, Chen H, Li Y, Huang H, Wang X, Guo Z, Zhou R. Transcriptomic analysis on the effects of melatonin in gastrointestinal carcinomas. BMC Gastroenterol 2020; 20:233. [PMID: 32689938 PMCID: PMC7372748 DOI: 10.1186/s12876-020-01383-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Melatonin has been shown with anticancer property and therapeutic potential for tumors. However, there lacks a systematic study on the molecular pathways of melatonin and its antitumor effects in gastrointestinal carcinomas. METHODS Using the gene expression profiles of four cancer cell lines from three types of gastrointestinal carcinomas before and after melatonin treatment, including gastric carcinoma (GC), colorectal carcinoma (CRC) and hepatocellular carcinoma (HCC), differentially expressed genes (DEGs) and biological pathways influenced by melatonin were identified. The qRT-PCR analyses were performed to validate the effects of melatonin on 5-FU resistance-related genes in CRC. RESULTS There were 17 pathways commonly altered by melatonin in the three cancer types, including FoxO signaling pathways enriched by the upregulated DEGs and cell cycle signaling pathways enriched by the downregulated DEGs, confirmed the dual role of melatonin to tumor growth, pro-apoptosis and anti-proliferation. DEGs upregulated in the three types of cancer tissues but reversely downregulated by melatonin were commonly enriched in RNA transport, spliceosome and cell cycle signaling pathways, which indicate that melatonin might exert antitumor effects through these pathways. Our results further showed that melatonin can downregulate the expression levels of 5-FU resistance-related genes, such as thymidylate synthase in GC and ATR, CHEK1, BAX and MYC in CRC. The qRT-PCR results demonstrated that melatonin enhanced the sensitivity of CRC 5-FU resistant cells by decreasing the expression of ATR. CONCLUSIONS Melatonin exerts the effects of pro-apoptosis and anti-proliferation on gastrointestinal carcinomas, and might increase the sensitivity of 5-FU in GC and CRC patients.
Collapse
Affiliation(s)
- Lu Ao
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China. .,Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.
| | - Li Li
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.,Department of Cell Biology and Genetics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Huaqin Sun
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Huxing Chen
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Yawei Li
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Haiyan Huang
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Xianlong Wang
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.,Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Zheng Guo
- Department of Bioinformatics, Fujian Key Laboratory of Medical Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.,Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Ruixiang Zhou
- Department of Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China. .,Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
26
|
Melatonin against Myocardial Ischemia-Reperfusion Injury: A Meta-analysis and Mechanism Insight from Animal Studies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1241065. [PMID: 32685084 PMCID: PMC7336233 DOI: 10.1155/2020/1241065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 12/18/2022]
Abstract
Aims Myocardial reperfusion damage after severe ischemia was an important issue during a clinical practice. However, the exacted pathogenesis involved remained unclear and also lacks effective interventions. Melatonin was identified to exert protective effects for alleviating the myocardial I/R injury. This meta-analysis was determined to evaluate the efficacy of melatonin treatment against reperfusion insult and further summarize potential molecular and cellular mechanisms. Methods and Results 15 eligible studies with 211 animals (108 received melatonin and 103 received vehicle) were included after searching the databases of PubMed, MEDLINE, Embase, and Cochrane. Pretreatment with melatonin was associated with a significant lower infarct size in comparison with vehicle in myocardial I/R damage (WMD: -20.45, 95% CI: -25.43 to -15.47, p < 0.001; I2 = 91.4%, p < 0.001). Evidence from subgroup analyses and sensitivity analysis indicated the robust and consistent cardioprotective effect of melatonin, while the metaregression also did not unmask any significant interactions between the pooled estimates and covariates (i.e., sample size, state, species, study type, route of administration, and duration of reperfusion, along with timing regimen of pretreatment). Accordingly, melatonin evidently increased EF (WMD: 17.19, 95% CI: 11.08 to 23.29, p < 0.001; I2 = 77.0%, p < 0.001) and FS (WMD: 14.18, 95% CI: 11.22 to 17.15, p < 0.001; I2 = 3.5%, p = 0.387) in the setting of reperfusion damage. Conclusions Melatonin preadministration conferred a profound cardioprotection against myocardial I/R injury in preclinical studies.
Collapse
|
27
|
Fu Z, Jiao Y, Wang J, Zhang Y, Shen M, Reiter RJ, Xi Q, Chen Y. Cardioprotective Role of Melatonin in Acute Myocardial Infarction. Front Physiol 2020; 11:366. [PMID: 32411013 PMCID: PMC7201093 DOI: 10.3389/fphys.2020.00366] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
Melatonin is a pleiotropic, indole secreted, and synthesized by the human pineal gland. Melatonin has biological effects including anti-apoptosis, protecting mitochondria, anti-oxidation, anti-inflammation, and stimulating target cells to secrete cytokines. Its protective effect on cardiomyocytes in acute myocardial infarction (AMI) has caused widespread interest in the actions of this molecule. The effects of melatonin against oxidative stress, promoting autophagic repair of cells, regulating immune and inflammatory responses, enhancing mitochondrial function, and relieving endoplasmic reticulum stress, play crucial roles in protecting cardiomyocytes from infarction. Mitochondrial apoptosis and dysfunction are common occurrence in cardiomyocyte injury after myocardial infarction. This review focuses on the targets of melatonin in protecting cardiomyocytes in AMI, the main molecular signaling pathways that melatonin influences in its endogenous protective role in myocardial infarction, and the developmental prospect of melatonin in myocardial infarction treatment.
Collapse
Affiliation(s)
- Zhenhong Fu
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yang Jiao
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jihang Wang
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ying Zhang
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mingzhi Shen
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, San Antonio, TX, United States
- San Antonio Cellular Therapeutics Institute, Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, TX, United States
| | - Qing Xi
- The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
28
|
Xu F, Zhong J, Lin X, Shan S, Guo B, Zheng M, Wang Y, Li F, Cui R, Wu F, Zhou E, Liao X, Liu Y, Yuan L. Melatonin alleviates vascular calcification and ageing through exosomal miR-204/miR-211 cluster in a paracrine manner. J Pineal Res 2020; 68:e12631. [PMID: 31943334 PMCID: PMC7154654 DOI: 10.1111/jpi.12631] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 12/08/2019] [Accepted: 01/06/2020] [Indexed: 12/24/2022]
Abstract
In the elderly with atherosclerosis, hypertension and diabetes, vascular calcification and ageing are ubiquitous. Melatonin (MT) has been demonstrated to impact the cardiovascular system. In this study, we have shown that MT alleviates vascular calcification and ageing, and the underlying mechanism involved. We found that both osteogenic differentiation and senescence of vascular smooth muscle cells (VSMCs) were attenuated by MT in a MT membrane receptor-dependent manner. Moreover, exosomes isolated from VSMCs or calcifying vascular smooth muscle cells (CVSMCs) treated with MT could be uptaken by VSMCs and attenuated the osteogenic differentiation and senescence of VSMCs or CVSMCs, respectively. Moreover, we used conditional medium from MT-treated VSMCs and Transwell assay to confirm exosomes secreted by MT-treated VSMCs attenuated the osteogenic differentiation and senescence of VSMCs through paracrine mechanism. We also found exosomal miR-204/miR-211 mediated the paracrine effect of exosomes secreted by VSMCs. A potential target of these two miRs was revealed to be BMP2. Furthermore, treatment of MT alleviated vascular calcification and ageing in 5/6-nephrectomy plus high-phosphate diet-treated (5/6 NTP) mice, while these effects were partially reversed by GW4869. Exosomes derived from MT-treated VSMCs were internalised into mouse artery detected by in vivo fluorescence image, and these exosomes reduced vascular calcification and ageing of 5/6 NTP mice, but both effects were largely abolished by inhibition of exosomal miR-204 or miR-211. In summary, our present study revealed that exosomes from MT-treated VSMCs could attenuate vascular calcification and ageing in a paracrine manner through an exosomal miR-204/miR-211.
Collapse
Affiliation(s)
- Feng Xu
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Jia‐Yu Zhong
- Department of GeriatricsInstitute of Aging and Age‐related Disease ResearchThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Xiao Lin
- Department of RadiologyThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Su‐Kang Shan
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Bei Guo
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Ming‐Hui Zheng
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Yi Wang
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Fuxingzi Li
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Rong‐Rong Cui
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Feng Wu
- Department of PathologyThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - En Zhou
- Department of Otorhinolaryngology Head and Neck SurgeryHunan Provincial People's HospitalChangshaChina
| | - Xiao‐Bo Liao
- Department of Cardiovascular SurgeryThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - You‐Shuo Liu
- Department of GeriatricsInstitute of Aging and Age‐related Disease ResearchThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Ling‐Qing Yuan
- Department of Metabolism and EndocrinologyNational Clinical Research Center for Metabolic DiseasesHunan Provincial Key Laboratory of Metabolic Bone DiseasesThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
29
|
Otamas A, Grant PJ, Ajjan RA. Diabetes and atherothrombosis: The circadian rhythm and role of melatonin in vascular protection. Diab Vasc Dis Res 2020; 17:1479164120920582. [PMID: 32506946 PMCID: PMC7607413 DOI: 10.1177/1479164120920582] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Obesity-related euglycaemic insulin resistance clusters with cardiometabolic risk factors, contributing to the development of both type 2 diabetes and cardiovascular disease. An increased thrombotic tendency in diabetes stems from platelet hyperactivity, enhanced activity of prothrombotic coagulation factors and impaired fibrinolysis. Furthermore, a low-grade inflammatory response and increased oxidative stress accelerate the atherosclerotic process and, together with an enhanced thrombotic environment, result in premature and more severe cardiovascular disease. The disruption of circadian cycles in man secondary to chronic obesity and loss of circadian cues is implicated in the increased risk of developing diabetes and cardiovascular disease. Levels of melatonin, the endogenous synchronizer of circadian rhythm, are reduced in individuals with vascular disease and those with deranged glucose metabolism. The anti-inflammatory, antihypertensive, antioxidative and antithrombotic activities of melatonin make it a potential therapeutic agent to reduce the risk of vascular occlusive disease in diabetes. The mechanisms behind melatonin-associated reduction in procoagulant response are not fully known. Current evidence suggests that melatonin inhibits platelet aggregation and might affect the coagulation cascade, altering fibrin clot structure and/or resistance to fibrinolysis. Large-scale clinical trials are warranted to investigate the effects of modulating the circadian clock on insulin resistance, glycaemia and cardiovascular outcome.
Collapse
Affiliation(s)
- Anastasia Otamas
- The LIGHT Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine and Leeds Teaching Hospitals Trust, University of Leeds, Leeds, UK
| | - Peter J Grant
- The LIGHT Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine and Leeds Teaching Hospitals Trust, University of Leeds, Leeds, UK
| | - Ramzi A Ajjan
- The LIGHT Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine and Leeds Teaching Hospitals Trust, University of Leeds, Leeds, UK
| |
Collapse
|
30
|
Redeker NS, Conley S, Anderson G, Cline J, Andrews L, Mohsenin V, Jacoby D, Jeon S. Effects of Cognitive Behavioral Therapy for Insomnia on Sleep, Symptoms, Stress, and Autonomic Function Among Patients With Heart Failure. Behav Sleep Med 2020; 18:190-202. [PMID: 30461315 PMCID: PMC6529289 DOI: 10.1080/15402002.2018.1546709] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background: Insomnia is common among patients with stable heart failure (HF) and associated with inflammation and altered autonomic function. Purpose: The purposes of this study were to examine the effects of cognitive behavioral therapy for insomnia (CBT-I) on the Hypothalamic Pituitary (HPA) Axis, autonomic function, inflammation, and circadian rhythmicity and the associations between these biomarkers and insomnia, sleep characteristics, symptoms, functional performance, and sleep-related cognitions. Methods: We conducted a subanalysis of a pilot randomized controlled trial (RCT, NCT02827799) whose primary aim was to test the effects of CBT-I on insomnia. We randomized 51 patients with stable Class II-IV HF to CBT-I (n = 30) or attention control (n = 21). Participants completed wrist actigraphy and self-reported insomnia severity, sleep characteristics, sleep-related cognitions, daytime symptoms, and functional performance. We measured day and nighttime urinary free cortisol, melatonin sulfate, epinephrine, and norepinephrine at baseline, and two weeks after CBT-I and computed general linear models and partial correlations. Results: CBT-I had no effects on the biomarkers, but there were statistically significant negative cross-sectional correlations between the ratio of day and night urinary free cortisol and sleep disturbance, anxiety, fatigue, depression, and negative sleep cognitions. Increases in the ratio between day and night cortisol were associated with statistically significant improvements in fatigue, depression, sleep duration, and sleep-related cognitions. Conclusions: Biomarkers of stress and autonomic function are associated with sleep, sleep-related symptoms, and cognitions among people with chronic HF. Future studies are needed to identify potential causal relationships and the impact of sleep interventions.
Collapse
Affiliation(s)
- Nancy S Redeker
- Beatrice Renfield Term Professor of Nursing, Yale School of Nursing, West Haven, Connecticut
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Khan S, Malik BH, Gupta D, Rutkofsky I. The Role of Circadian Misalignment due to Insomnia, Lack of Sleep, and Shift Work in Increasing the Risk of Cardiac Diseases: A Systematic Review. Cureus 2020; 12:e6616. [PMID: 32064196 PMCID: PMC7008727 DOI: 10.7759/cureus.6616] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Around 121.5 million people suffer from cardiovascular diseases globally. The risk of cardiovascular diseases increases with advancing age in both genders. Circadian rhythm is responsible for a streamlined functioning of various body functions. Certain functions and hormones have their peak levels according to the biological day or night of circadian rhythm. Shift work and sleep disorders like obstructive sleep apnea can cause circadian misalignment that affects different metabolic, immunological, and cardiovascular functions, which ultimately increases the risk of cardiovascular diseases. We systematically searched the online database PubMed to find papers on randomized controlled trials (RCTs) from the past five years, evaluating the role of shift work and different sleep disorders in causing circadian misalignment and its effect on the risk of cardiovascular diseases. Fifty papers were shortlisted, and after the application of various inclusion and exclusion criteria, 18 papers were chosen; and then after a thorough analysis of the text, eight papers were selected for the review. All papers were evaluated for quality. Two papers focused on the effect of shift work on cardiovascular diseases, whereas five papers evaluated the role of sleep disorders on circadian rhythm and the risk of cardiovascular diseases. Shift work and sleep-related disorders were found to cause circadian misalignment, and it was found to be associated with an increase in the risk of cardiovascular diseases. Managing these disorders can help reduce the risk of cardiovascular diseases.
Collapse
Affiliation(s)
- Safeera Khan
- Family Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Bilal Haider Malik
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Deepti Gupta
- Reproductive Medicine, Saint Mary's Hospital, Manchester, GBR
| | - Ian Rutkofsky
- Psychiatry, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
32
|
Ding S, Lin N, Sheng X, Zhao Y, Su Y, Xu L, Tong R, Yan Y, Fu Y, He J, Gao Y, Yuan A, Ye L, Reiter RJ, Pu J. Melatonin stabilizes rupture-prone vulnerable plaques via regulating macrophage polarization in a nuclear circadian receptor RORα-dependent manner. J Pineal Res 2019; 67:e12581. [PMID: 31009101 DOI: 10.1111/jpi.12581] [Citation(s) in RCA: 86] [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: 12/01/2018] [Revised: 03/18/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
Abstract
Rupture of vulnerable plaques is the main trigger of acute cardio-cerebral vascular events, but mechanisms responsible for transforming a stable atherosclerotic into a vulnerable plaque remain largely unknown. Melatonin, an indoleamine hormone secreted by the pineal gland, plays pleiotropic roles in the cardiovascular system; however, the effect of melatonin on vulnerable plaque rupture and its underlying mechanisms remains unknown. Here, we generated a rupture-prone vulnerable carotid plaque model induced by endogenous renovascular hypertension combined with low shear stress in hypercholesterolemic ApoE-/- mice. Melatonin (10 mg/kg/d by oral administration for 9 weeks) significantly prevented vulnerable plaque rupture, with lower incidence of intraplaque hemorrhage (42.9% vs. 9.5%, P = 0.014) and of spontaneous plaque rupture with intraluminal thrombus formation (38.1% vs. 9.5%, P = 0.029). Mechanistic studies indicated that melatonin ameliorated intraplaque inflammation by suppressing the differentiation of intraplaque macrophages toward the proinflammatory M1 phenotype, and circadian nuclear receptor retinoid acid receptor-related orphan receptor-α (RORα) mediated melatonin-exerted vasoprotection against vulnerable plaque instability and intraplaque macrophage polarization. Further analysis in human monocyte-derived macrophages confirmed the role of melatonin in regulating macrophage polarization by regulating the AMPKα-STATs pathway in a RORα-dependent manner. In summary, our data provided the first evidence that melatonin-RORα axis acts as a novel endogenous protective signaling pathway in the vasculature, regulates intraplaque inflammation, and stabilizes rupture-prone vulnerable plaques.
Collapse
MESH Headings
- Animals
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Humans
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Melatonin/pharmacology
- Mice
- Mice, Knockout, ApoE
- Nuclear Receptor Subfamily 1, Group F, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Signal Transduction/drug effects
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Song Ding
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Nan Lin
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Xincheng Sheng
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Yichao Zhao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Yuanyuan Su
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Longwei Xu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Renyang Tong
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Yang Yan
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Yanan Fu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Jie He
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Yu Gao
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Ancai Yuan
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore City, Singapore
| | - Russel J Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, Texas
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
33
|
Lu J, Sun Z, Fang Y, Zheng J, Xu S, Xu W, Shi L, Mei S, Wu H, Liang F, Zhang J. Melatonin Suppresses Microglial Necroptosis by Regulating Deubiquitinating Enzyme A20 After Intracerebral Hemorrhage. Front Immunol 2019; 10:1360. [PMID: 31258534 PMCID: PMC6587666 DOI: 10.3389/fimmu.2019.01360] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/29/2019] [Indexed: 11/19/2022] Open
Abstract
Cell death is deeply involved in pathophysiology of brain injury after intracerebral hemorrhage (ICH). Necroptosis, one of the recently discovered forms of cell death, plays an important role in various diseases, including ICH. Previous studies have suggested that a considerable number of neurons undergoes necroptosis after ICH. However, necroptosis of microglia after ICH has not been reported to date. The present study demonstrated for the first time that necroptosis occurred in the microglia surrounding the hematoma after ICH in C57 mice, and melatonin, a hormone that is predominantly synthesized in and secreted from the pineal gland, exerted a neuroprotective effect by suppressing this process. When we further explored the potential underlying mechanism, we found that melatonin inhibits RIP3-mediated necroptosis by regulating the deubiquitinating enzyme A20 (also known as TNFAIP3) expression after ICH. In summary, we have demonstrated the role of microglial necroptosis in the pathogenesis of ICH. More importantly, A20 was identified as a novel target of melatonin, which opens perspectives for future research.
Collapse
Affiliation(s)
- Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zeyu Sun
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingwei Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shenbin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ligen Shi
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuhao Mei
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Liang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Brain Research Institute, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
34
|
Misaka T, Yoshihisa A, Yokokawa T, Sato T, Oikawa M, Kobayashi A, Yamaki T, Sugimoto K, Kunii H, Nakazato K, Takeishi Y. Plasma levels of melatonin in dilated cardiomyopathy. J Pineal Res 2019; 66:e12564. [PMID: 30715754 PMCID: PMC6593840 DOI: 10.1111/jpi.12564] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/05/2019] [Accepted: 01/19/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Melatonin is a multifunctional indolamine and has a cardioprotective role in a variety of cardiovascular processes via antioxidant, anti-inflammatory, antihypertensive, antithrombotic, and antilipemic effects. It has been reported that lower levels of circulating melatonin are significantly associated with a higher risk of acute myocardial infarction (AMI) and later cardiac remodeling. However, levels of melatonin in patients with dilated cardiomyopathy (DCM) and associations between melatonin levels and cardiac function remain unclear. METHODS AND RESULTS We measured and compared plasma levels of melatonin in 61 control subjects, 81 AMI patients, and 77 DCM patients. Plasma levels of melatonin were progressively decreased from 71.9 pg/mL in the control group to 52.6 pg/mL in the DCM group and 21.9 pg/mL in the AMI group. Next, we examined associations of melatonin levels with parameters of laboratory data, echocardiography, and right-heart catheterization. In the DCM patients, circulating melatonin showed significant correlations with both high-sensitivity troponin T (R = -0.422, P < 0.001) and cardiac output (R = 0.431, P = 0.003), but not with B-type natriuretic peptide (BNP), left ventricular ejection fraction (LVEF), pulmonary artery wedge pressure, or pulmonary artery pressure. CONCLUSION Patients with not only AMI but also DCM had lower circulating melatonin levels. Circulating melatonin levels appear to correlate with myocardial injury and cardiac output in DCM patients.
Collapse
Affiliation(s)
- Tomofumi Misaka
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
- Department of Advanced Cardiac TherapeuticsFukushima Medical UniversityFukushimaJapan
| | - Akiomi Yoshihisa
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
- Department of Advanced Cardiac TherapeuticsFukushima Medical UniversityFukushimaJapan
| | - Tetsuro Yokokawa
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
- Department of Pulmonary HypertensionFukushima Medical UniversityFukushimaJapan
| | - Takamasa Sato
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
| | - Masayoshi Oikawa
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
| | - Atsushi Kobayashi
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
| | - Takayoshi Yamaki
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
| | - Koichi Sugimoto
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
- Department of Pulmonary HypertensionFukushima Medical UniversityFukushimaJapan
| | - Hiroyuki Kunii
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
| | - Kazuhiko Nakazato
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
| | - Yasuchika Takeishi
- Department of Cardiovascular MedicineFukushima Medical UniversityFukushimaJapan
| |
Collapse
|
35
|
Seifalian A, Hart A. Circadian Rhythms: Will It Revolutionise the Management of Diseases? J Lifestyle Med 2019; 9:1-11. [PMID: 30918828 PMCID: PMC6425903 DOI: 10.15280/jlm.2019.9.1.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 09/26/2018] [Indexed: 11/22/2022] Open
Abstract
The Nobel Prize for Medicine in 2017 was awarded to Michael Young, Michael Rosbash and Jeffrey Hall for their discoveries into the molecular mechanisms controlling circadian rhythms (CR). The aims of this paper were to present the mechanisms behind the CRs and discuss the impact this could have on human health. We argued that further research in this field has the potential to revolutionise healthcare through understanding the influence on the pathogenesis of disease, including in cardiovascular, mental and neurological health, as well as influence on cognitive function. The research has shown that intrinsic CRs have physiological and biochemical influences on the body, which may affect the efficiency of drug absorption due to the altered activity of enzymes. There is strong data to suggest CR disturbances, due to either shift work, sleep disorders or frequent travel between time zones, has negative impact on health. This article aims to summarise the extent of this impact and analyse CRs as a potential therapeutic target, as well as describing the pathophysiology and mechanisms driving the course of disease among people with CR disorders. These new discoveries may revolutionise the way in which treatment is provided in the future with more focus on lifestyle changes to provide treatment and more optimal precision medicine. Pharmaceutical companies and healthcare staff must consider the significant message provided from this data and use the information to optimise drug delivery and treatment provision. The facts of CRs role in healthcare can no longer be ignored.
Collapse
Affiliation(s)
- Amelia Seifalian
- University College London Medical School, London, United Kingdom
| | - Ashley Hart
- University College London Medical School, London, United Kingdom
| |
Collapse
|
36
|
Kopáni M, Vraníková B, Kosnáč D, Zeman M, Šišovský V, Polakovičová S, Biró C. Pineal gland calcification under hypoxic conditions. Physiol Res 2019; 68:S405-S413. [DOI: 10.33549/physiolres.934378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The pineal gland (glandula pinealis) is neuroendocrine gland located at the epithalamus of the brain secreting melatonin. The aim of this study was to explore effects of prenatal hypoxia in rats at the age of 33 weeks on the occurrence of pineal gland calcification. Distribution and chemical composition of calcerous material by light, scanning and transmission electron microscopy was investigated. Melatonin concentrations in blood plasma by direct radioimmunoassay were measured. Rats were exposed to prenatal hypoxia for 12 h at day 20 of development and second group to prenatal hypoxia for 2x8 h at days 19 and 20 of development. Vacuoles of intracellular edema in the pineal samples after 12 h hypoxia were found. Their size ranges up to 30 µm. Some of them were filled with the flocculent and fibrous material. Samples of pineal glands after 2 x 8 h hypoxia revealed the pericellular edema of pinealocytes. The amount of calcium rich particles in 2 x 8 h hypoxia group was lower than in 12 h hypoxia group. Plasma melatonin levels did not differ between control and both hypoxia groups. We concluded that calcification is a process induced by osteoblasts and osteocytes with melatonin as a promotor and it is favored under hypoxic conditions.
Collapse
Affiliation(s)
- M. Kopáni
- , Institute of Medical Physics, Biophysics, Informatics and Telemedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | | | | | | | | | | | | |
Collapse
|
37
|
Agorastos A, Nicolaides NC, Bozikas VP, Chrousos GP, Pervanidou P. Multilevel Interactions of Stress and Circadian System: Implications for Traumatic Stress. Front Psychiatry 2019; 10:1003. [PMID: 32047446 PMCID: PMC6997541 DOI: 10.3389/fpsyt.2019.01003] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
The dramatic fluctuations in energy demands by the rhythmic succession of night and day on our planet has prompted a geophysical evolutionary need for biological temporal organization across phylogeny. The intrinsic circadian timing system (CS) represents a highly conserved and sophisticated internal "clock," adjusted to the 24-h rotation period of the earth, enabling a nyctohemeral coordination of numerous physiologic processes, from gene expression to behavior. The human CS is tightly and bidirectionally interconnected to the stress system (SS). Both systems are fundamental for survival and regulate each other's activity in order to prepare the organism for the anticipated cyclic challenges. Thereby, the understanding of the temporal relationship between stressors and stress responses is critical for the comprehension of the molecular basis of physiology and pathogenesis of disease. A critical loss of the harmonious timed order at different organizational levels may affect the fundamental properties of neuroendocrine, immune, and autonomic systems, leading to a breakdown of biobehavioral adaptative mechanisms with increased stress sensitivity and vulnerability. In this review, following an overview of the functional components of the SS and CS, we present their multilevel interactions and discuss how traumatic stress can alter the interplay between the two systems. Circadian dysregulation after traumatic stress exposure may represent a core feature of trauma-related disorders mediating enduring neurobiological correlates of trauma through maladaptive stress regulation. Understanding the mechanisms susceptible to circadian dysregulation and their role in stress-related disorders could provide new insights into disease mechanisms, advancing psychochronobiological treatment possibilities and preventive strategies in stress-exposed populations.
Collapse
Affiliation(s)
- Agorastos Agorastos
- Department of Psychiatry, Division of Neurosciences, Faculty of Medical Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.,VA Center of Excellence for Stress and Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA, United States
| | - Nicolas C Nicolaides
- First Department of Pediatrics, Division of Endocrinology, Metabolism and Diabetes, School of Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Vasilios P Bozikas
- Department of Psychiatry, Division of Neurosciences, Faculty of Medical Sciences, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George P Chrousos
- First Department of Pediatrics, Division of Endocrinology, Metabolism and Diabetes, School of Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece.,Unit of Developmental & Behavioral Pediatrics, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Panagiota Pervanidou
- Unit of Developmental & Behavioral Pediatrics, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| |
Collapse
|
38
|
Lu J, Sun Z, Fang Y, Zheng J, Xu S, Xu W, Shi L, Mei S, Wu H, Liang F, Zhang J. Melatonin Suppresses Microglial Necroptosis by Regulating Deubiquitinating Enzyme A20 After Intracerebral Hemorrhage. Front Immunol 2019. [PMID: 31258534 DOI: 10.3389/fimmu.2019.01360/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Abstract
Cell death is deeply involved in pathophysiology of brain injury after intracerebral hemorrhage (ICH). Necroptosis, one of the recently discovered forms of cell death, plays an important role in various diseases, including ICH. Previous studies have suggested that a considerable number of neurons undergoes necroptosis after ICH. However, necroptosis of microglia after ICH has not been reported to date. The present study demonstrated for the first time that necroptosis occurred in the microglia surrounding the hematoma after ICH in C57 mice, and melatonin, a hormone that is predominantly synthesized in and secreted from the pineal gland, exerted a neuroprotective effect by suppressing this process. When we further explored the potential underlying mechanism, we found that melatonin inhibits RIP3-mediated necroptosis by regulating the deubiquitinating enzyme A20 (also known as TNFAIP3) expression after ICH. In summary, we have demonstrated the role of microglial necroptosis in the pathogenesis of ICH. More importantly, A20 was identified as a novel target of melatonin, which opens perspectives for future research.
Collapse
Affiliation(s)
- Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zeyu Sun
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingwei Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shenbin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ligen Shi
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuhao Mei
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Liang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Brain Research Institute, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Xu T, Lu B. The effects of phytochemicals on circadian rhythm and related diseases. Crit Rev Food Sci Nutr 2018; 59:882-892. [DOI: 10.1080/10408398.2018.1493678] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Tao Xu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Baiyi Lu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Postharvest Handling of Ministry of Agriculture and Rural affairs, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
40
|
Yang L, Zhang Y, Ma Y, Du J, Gu L, Zheng L, Zhang X. Effect of melatonin on EGF- and VEGF-induced monolayer permeability of HUVECs. Am J Physiol Heart Circ Physiol 2018; 316:H1178-H1191. [PMID: 30575440 DOI: 10.1152/ajpheart.00542.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Melatonin is a natural hormone involved in the regulation of circadian rhythm, immunity, and cardiovascular function. In the present study, we focused on the mechanism of melatonin in the regulation of vascular permeability. We found that melatonin could inhibit both VEGF- and EGF-induced monolayer permeability of human umbilical vein endothelial cells (HUVECs) and change the tyrosine phosphorylation of vascular-endothelial (VE-)cadherin, which was related to endothelial barrier function. In addition, phospho-AKT (Ser473) and phospho-ERK(1/2) played significant roles in the regulation of VE-cadherin phosphorylation. Both the phosphatidylinositol 3-kinase/AKT inhibitor LY49002 and MEK/ERK inhibitor U0126 could inhibit the permeability of HUVECs, but with different effects on tyrosine phosphorylation of VE-cadherin. Melatonin can influence the two growth factor-induced phosphorylation of AKT (Ser473) but not ERK(1/2). Our results show that melatonin can inhibit growth factor-induced monolayer permeability of HUVECs by influencing the phosphorylation of AKT and VE-cadherin. Melatonin can be a potential treatment for diseases associated with abnormal vascular permeability. NEW & NOTEWORTHY We found that melatonin could inhibit both EGF- and VEGF-induced monolayer permeability of human umbilical vein endothelial cells, which is related to phosphorylation of vascular-endothelial cadherin. Blockade of phosphatidylinositol 3-kinase/AKT and MEK/ERK pathways could inhibit the permeability of human umbilical vein endothelial cells, and phosphorylation of AKT (Ser473) might be a critical event in the changing of monolayer permeability and likely has cross-talk with the MEK/ERK pathway.
Collapse
Affiliation(s)
- Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University , Changzhou, Jiangsu , China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University , Nanjing, Jiangsu , China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University , Nanjing, Jiangsu , China
| | - Yadong Ma
- Department of Physiology, Nanjing Medical University , Nanjing, Jiangsu , China
| | - Jun Du
- Department of Physiology, Nanjing Medical University , Nanjing, Jiangsu , China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University , Nanjing, Jiangsu , China
| | - Luo Gu
- Department of Physiology, Nanjing Medical University , Nanjing, Jiangsu , China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University , Nanjing, Jiangsu , China
| | - Lu Zheng
- General Laboratory, The Third Affiliated Hospital of Soochow University , Changzhou, Jiangsu , China
| | - Xiaoying Zhang
- Department of Cardiothoracic surgery and the General Laboratory, The Third Affiliated Hospital of Soochow University , Changzhou, Jiangsu , China
| |
Collapse
|
41
|
Strohmaier S, Devore EE, Zhang Y, Schernhammer ES. A Review of Data of Findings on Night Shift Work and the Development of DM and CVD Events: a Synthesis of the Proposed Molecular Mechanisms. Curr Diab Rep 2018; 18:132. [PMID: 30343445 PMCID: PMC6209035 DOI: 10.1007/s11892-018-1102-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Night shift work has become highly prevalent in our 24/7 societies, with up to 18% of the US work force working alternate shift schedules. However, studies indicate that there may be adverse health effects of chronic night work across diverse populations. These effects are likely due to misalignment of the circadian system with work schedules, mediated by the system's primary marker melatonin as well as other downstream molecules. RECENT FINDINGS Melatonin has multiple biologic actions that are relevant to cardiometabolic disease, including modulation of oxidative stress, inflammation, and (via the melatonin receptor) vasoconstriction. Behavioral traits, such as chronotype and meal timing, have recently been shown to interact with the effects of night work on cardiometabolic health. Together with recent findings suggesting a role for circadian genes in cardiometabolic risk, the interactions of night shift work and behavioral traits are likely to facilitate novel treatment and prevention approaches for cardiovascular disease and type 2 diabetes, incorporating aspects of clock and timing.
Collapse
Affiliation(s)
- S. Strohmaier
- 0000 0000 9259 8492grid.22937.3dDepartment of Epidemiology, Center for Public Health, Medical University of Vienna, Kinderspitalgasse 15, 1090 Vienna, Austria
- 000000041936754Xgrid.38142.3cChanning Division of Network Medicine, Harvard Medical School, Boston, MA USA
| | - E. E. Devore
- 000000041936754Xgrid.38142.3cChanning Division of Network Medicine, Harvard Medical School, Boston, MA USA
| | - Y. Zhang
- 000000041936754Xgrid.38142.3cChanning Division of Network Medicine, Harvard Medical School, Boston, MA USA
| | - E. S. Schernhammer
- 0000 0000 9259 8492grid.22937.3dDepartment of Epidemiology, Center for Public Health, Medical University of Vienna, Kinderspitalgasse 15, 1090 Vienna, Austria
- 000000041936754Xgrid.38142.3cChanning Division of Network Medicine, Harvard Medical School, Boston, MA USA
- 000000041936754Xgrid.38142.3cDepartment of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA USA
| |
Collapse
|
42
|
Melatonin Inhibits the Progression of Hepatocellular Carcinoma through MicroRNA Let7i-3p Mediated RAF1 Reduction. Int J Mol Sci 2018; 19:ijms19092687. [PMID: 30201903 PMCID: PMC6163650 DOI: 10.3390/ijms19092687] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/06/2018] [Accepted: 09/08/2018] [Indexed: 02/07/2023] Open
Abstract
Melatonin is the main pineal hormone that relays light/dark-cycle information to the circadian system. Recent studies have examined the intrinsic antitumor activity of melatonin in various cancers, including hepatocellular carcinoma (HCC), the primary life-threatening malignancy in both sexes in Taiwan. However, the detailed regulatory mechanisms underlying melatonin’s anti-HCC activity remain incompletely understood. Here, we investigated the mechanisms by which the anti-HCC activity of melatonin is regulated. Human hepatoma cell lines were treated with 1 and 2 mM melatonin, and functional assays were used to dissect melatonin’s antitumor effect in HCC; small-RNA sequencing was performed to identify the microRNAs (miRNAs) involved in the anti-HCC activity of melatonin; and quantitative RT-PCR and Western blotting were used to elucidate how miRNAs regulate melatonin-mediated HCC suppression. Melatonin treatment at both doses strongly inhibited the proliferation, migration and invasion capacities of Huh7 and HepG2 cell lines, and melatonin treatment markedly induced the expression of the miRNA let7i-3p in cells. Notably, transfection of cells with a let7i-3p mimic drastically reduced RAF1 expression and activation of mitogen-activated protein kinase signaling downstream from RAF1, and rescue-assay results demonstrated that melatonin inhibited HCC progression by modulating let7i-3p-mediated RAF1 suppression. Our findings support the view that melatonin treatment holds considerable promise as a therapy for HCC.
Collapse
|
43
|
Melatonin Ameliorates the Progression of Atherosclerosis via Mitophagy Activation and NLRP3 Inflammasome Inhibition. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9286458. [PMID: 30254716 PMCID: PMC6142770 DOI: 10.1155/2018/9286458] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/24/2018] [Accepted: 07/11/2018] [Indexed: 01/24/2023]
Abstract
The NLRP3 (nucleotide-binding domain and leucine-rich repeat pyrin domain containing 3) inflammasome-mediated inflammatory responses are critically involved in the progression of atherosclerosis (AS), which is the essential cause for cardiovascular diseases. Melatonin has anti-inflammatory properties. However, little is known about the potential effects of melatonin in the pathological process of AS. Herein, we demonstrate that melatonin suppressed prolonged NLRP3 inflammasome activation in atherosclerotic lesions by reactive oxygen species (ROS) scavenging via mitophagy in macrophages. The atherosclerotic mouse model was induced with a high-fat diet using ApoE−/− mice. Melatonin treatment markedly attenuated AS plaque size and vulnerability. Furthermore, melatonin decreased NLRP3 inflammasome activation and the consequent IL-1β secretion within atherosclerotic lesions. Despite the unchanged protein expression, the silent information regulator 3 (Sirt3) activity was elevated in the atherosclerotic lesions in melatonin-treated mice. In ox-LDL-treated macrophages, melatonin attenuated the NLRP3 inflammasome activation and the inflammatory factors secretion, while this protective effect was abolished by either Sirt3 silence or autophagy inhibitor 3-MA. Mitochondrial ROS (mitoROS), which was a recognized inducer for NLRP3 inflammasome, was attenuated by melatonin through the induction of mitophagy. Both Sirt3-siRNA and autophagy inhibitor 3-MA partially abolished the beneficial effects of melatonin on mitoROS clearance and NLRP3 inflammasome activation, indicating the crucial role of Sirt3-mediated mitophagy. Furthermore, we demonstrated that melatonin protected against AS via the Sirt3/FOXO3a/Parkin signaling pathway. In conclusion, the current study demonstrated that melatonin prevented atherosclerotic progression, at least in part, via inducing mitophagy and attenuating NLRP3 inflammasome activation, which was mediated by the Sirt3/FOXO3a/Parkin signaling pathway. Collectively, our study provides insight into melatonin as a new target for therapeutic intervention for AS.
Collapse
|
44
|
Zhong J, Liu Y. Melatonin and age-related cardiovascular diseases. Aging Med (Milton) 2018; 1:197-203. [PMID: 31942497 PMCID: PMC6880684 DOI: 10.1002/agm2.12036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 08/13/2018] [Indexed: 12/29/2022] Open
Abstract
The pineal gland is a neuroendocrine gland closely related to human aging. Melatonin is a kind of indole neuroendocrine hormone secreted by the pineal gland, which is essential for maintaining physiological function. Many researches found that melatonin plays a key role in anti-aging-related cardiovascular diseases. In this paper, the latest advances in the study of melatonin and aging-related cardiovascular diseases are reviewed, and their related physiological functions and mechanisms are discussed.
Collapse
Affiliation(s)
- Jiayu Zhong
- Department of GeriatricsThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| | - Youshuo Liu
- Department of GeriatricsThe Second Xiang‐Ya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
45
|
Jiki Z, Lecour S, Nduhirabandi F. Cardiovascular Benefits of Dietary Melatonin: A Myth or a Reality? Front Physiol 2018; 9:528. [PMID: 29867569 PMCID: PMC5967231 DOI: 10.3389/fphys.2018.00528] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022] Open
Abstract
The role of the diet as well as the impact of the dietary habits on human health and disease is well established. Apart from its sleep regulatory effect, the indoleamine melatonin is a well-established antioxidant molecule with multiple health benefits. Convincing evidence supports the presence of melatonin in plants and foods with the intake of such foods affecting circulating melatonin levels in humans. While numerous actions of both endogenous melatonin and melatonin supplementation are well described, little is known about the influence of the dietary melatonin intake on human health. In the present review, evidence for the cardiovascular health benefits of melatonin supplementation and dietary melatonin is discussed. Current knowledge on the biological significance as well as the underlying physiological mechanism of action of the dietary melatonin is also summarized. Whether dietary melatonin constitutes an alternative preventive treatment for cardiovascular disease is addressed.
Collapse
Affiliation(s)
- Zukiswa Jiki
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Frederic Nduhirabandi
- Cardioprotection Group, Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
46
|
Li T, Ni L, Zhao Z, Liu X, Lai Z, Di X, Xie Z, Song X, Wang X, Zhang R, Liu C. Melatonin attenuates smoking-induced hyperglycemia via preserving insulin secretion and hepatic glycogen synthesis in rats. J Pineal Res 2018; 64:e12475. [PMID: 29437243 PMCID: PMC5947659 DOI: 10.1111/jpi.12475] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/02/2018] [Indexed: 12/11/2022]
Abstract
Epidemiology survey indicated that cigarette smoking is a risk factor of diabetes. However, the precise mechanisms remain to be clarified. In this study, we found that smoking caused metabolic malfunctions on pancreas and liver in experimental animal model. These were indicated by hyperglycemia, increased serum hemoglobin A1c level and decreased insulin secretion, inhibition of liver glycogen synthase (LGS), and hepatic glycogen synthesis. Mechanistic studies revealed that all these alterations were caused by the inflammatory reaction and reactive oxygen species (ROS) induced by the smoking. Melatonin treatment significantly preserved the functions of both pancreas and liver by reducing β cell apoptosis, CD68-cell infiltration, ROS production, and caspase-3 expression. The siRNA-knockdown model identified that the protective effects of melatonin were mediated by melatonin receptor-2 (MT2). This study uncovered potentially underlying mechanisms related to the association between smoking and diabetes. In addition, it is, for first time, to report that melatonin effectively protects against smoking-induced glucose metabolic alterations and the signal transduction pathway of melatonin is mainly mediated by its MT2 receptor. These observations provide solid evidence for the clinically use of melatonin to reduce smoking-related diabetes, and the therapeutic regimens are absent currently.
Collapse
Affiliation(s)
- Tianjia Li
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Leng Ni
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhewei Zhao
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinnong Liu
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhichao Lai
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiao Di
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhibo Xie
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xitao Song
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xuebin Wang
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Rui Zhang
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Changwei Liu
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
47
|
Altered Circadian Timing System-Mediated Non-Dipping Pattern of Blood Pressure and Associated Cardiovascular Disorders in Metabolic and Kidney Diseases. Int J Mol Sci 2018; 19:ijms19020400. [PMID: 29385702 PMCID: PMC5855622 DOI: 10.3390/ijms19020400] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/12/2018] [Accepted: 01/20/2018] [Indexed: 12/15/2022] Open
Abstract
The morning surge in blood pressure (BP) coincides with increased cardiovascular (CV) events. This strongly suggests that an altered circadian rhythm of BP plays a crucial role in the development of CV disease (CVD). A disrupted circadian rhythm of BP, such as the non-dipping type of hypertension (i.e., absence of nocturnal BP decline), is frequently observed in metabolic disorders and chronic kidney disease (CKD). The circadian timing system, controlled by the central clock in the suprachiasmatic nucleus of the hypothalamus and/or by peripheral clocks in the heart, vasculature, and kidneys, modulates the 24 h oscillation of BP. However, little information is available regarding the molecular and cellular mechanisms of an altered circadian timing system-mediated disrupted dipping pattern of BP in metabolic disorders and CKD that can lead to the development of CV events. A more thorough understanding of this pathogenesis could provide novel therapeutic strategies for the management of CVD. This short review will address our and others' recent findings on the molecular mechanisms that may affect the dipping pattern of BP in metabolic dysfunction and kidney disease and its association with CV disorders.
Collapse
|
48
|
Simko F, Pechanova O, Repova K, Aziriova S, Krajcirovicova K, Celec P, Tothova L, Vrankova S, Balazova L, Zorad S, Adamcova M. Lactacystin-Induced Model of Hypertension in Rats: Effects of Melatonin and Captopril. Int J Mol Sci 2017; 18:E1612. [PMID: 28757582 PMCID: PMC5578004 DOI: 10.3390/ijms18081612] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/10/2017] [Accepted: 07/13/2017] [Indexed: 12/15/2022] Open
Abstract
Lactacystin is a proteasome inhibitor that interferes with several factors involved in heart remodelling. The aim of this study was to investigate whether the chronic administration of lactacystin induces hypertension and heart remodelling and whether these changes can be modified by captopril or melatonin. In addition, the lactacystin-model was compared with NG-nitro-l-arginine-methyl ester (L-NAME)- and continuous light-induced hypertension. Six groups of three-month-old male Wistar rats (11 per group) were treated for six weeks as follows: control (vehicle), L-NAME (40 mg/kg/day), continuous light (24 h/day), lactacystin (5 mg/kg/day) alone, and lactacystin with captopril (100 mg/kg/day), or melatonin (10 mg/kg/day). Lactacystin treatment increased systolic blood pressure (SBP) and induced fibrosis of the left ventricle (LV), as observed in L-NAME-hypertension and continuous light-hypertension. LV weight and the cross-sectional area of the aorta were increased only in L-NAME-induced hypertension. The level of oxidative load was preserved or reduced in all three models of hypertension. Nitric oxide synthase (NOS) activity in the LV and kidney was unchanged in the lactacystin group. Nuclear factor-kappa B (NF-κB) protein expression in the LV was increased in all treated groups in the cytoplasm, however, in neither group in the nucleus. Although melatonin had no effect on SBP, only this indolamine (but not captopril) reduced the concentration of insoluble and total collagen in the LV and stimulated the NO-pathway in the lactacystin group. We conclude that chronic administration of lactacystin represents a novel model of hypertension with collagenous rebuilding of the LV, convenient for testing antihypertensive drugs or agents exerting a cardiovascular benefit beyond blood pressure reduction.
Collapse
Affiliation(s)
- Fedor Simko
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 81108 Bratislava, Slovakia.
- 3rd Clinic of Internal Medicine, Faculty of Medicine, Comenius University, 83305 Bratislava, Slovakia.
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 84505 Bratislava, Slovakia.
| | - Olga Pechanova
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 81371 Bratislava, Slovakia.
| | - Kristina Repova
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 81108 Bratislava, Slovakia.
| | - Silvia Aziriova
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 81108 Bratislava, Slovakia.
| | - Kristina Krajcirovicova
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 81108 Bratislava, Slovakia.
| | - Peter Celec
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Sasinkova 4, 81108 Bratislava, Slovakia.
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia.
| | - Lubomira Tothova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia.
| | - Stanislava Vrankova
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 81371 Bratislava, Slovakia.
| | - Lucia Balazova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 84505 Bratislava, Slovakia.
| | - Stefan Zorad
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 84505 Bratislava, Slovakia.
| | - Michaela Adamcova
- Department of Physiology, Faculty of Medicine, Charles University, 50003 Hradec Kralove, Czech Republic.
| |
Collapse
|
49
|
Wang TH, Wu CH, Yeh CT, Su SC, Hsia SM, Liang KH, Chen CC, Hsueh C, Chen CY. Melatonin suppresses hepatocellular carcinoma progression via lncRNA-CPS1-IT-mediated HIF-1α inactivation. Oncotarget 2017; 8:82280-82293. [PMID: 29137263 PMCID: PMC5669889 DOI: 10.18632/oncotarget.19316] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/10/2017] [Indexed: 12/27/2022] Open
Abstract
Melatonin is the primary pineal hormone that relays light/dark cycle information to the circadian system. It was recently reported to exert intrinsic antitumor activity in various cancers. However, the regulatory mechanisms underlying the antitumor activity of melatonin are poorly understood. Moreover, a limited number of studies have addressed the role of melatonin in hepatocellular carcinoma (HCC), a major life-threatening malignancy in both sexes in Taiwan. In this study, we investigated the antitumor effects of melatonin in HCC and explored the regulatory mechanisms underlying these effects. We observed that melatonin significantly inhibited the proliferation, migration, and invasion of HCC cells and significantly induced the expression of the transcription factor FOXA2 in HCC cells. This increase in FOXA2 expression resulted in upregulation of lncRNA-CPS1 intronic transcript 1 (CPS1-IT1), which reduced HIF-1α activity and consequently resulted in the suppression of epithelial-mesenchymal transition (EMT) progression and HCC metastasis. Furthermore, the results of the in vivo experiments confirmed that melatonin exerts tumor suppressive effects by reducing tumor growth. In conclusion, our findings suggested that melatonin inhibited HCC progression by reducing lncRNA-CPS1-IT1-mediated EMT suppression and indicated that melatonin could be a promising treatment for HCC.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan.,Liver Research Center, Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Chi-Hao Wu
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Kung-Hao Liang
- Liver Research Center, Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Chin-Chuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Tao-Yuan, Taiwan
| | - Chuen Hsueh
- Tissue Bank, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Tao-Yuan, Taiwan
| | - Chi-Yuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.,Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| |
Collapse
|
50
|
Tian T, Wang Z, Zhang J. Pathomechanisms of Oxidative Stress in Inflammatory Bowel Disease and Potential Antioxidant Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4535194. [PMID: 28744337 PMCID: PMC5506473 DOI: 10.1155/2017/4535194] [Citation(s) in RCA: 411] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/22/2017] [Accepted: 05/31/2017] [Indexed: 12/22/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal disease whose incidence has risen worldwide in recent years. Accumulating evidence shows that oxidative stress plays an essential role in the pathogenesis and progression of IBD. This review highlights the generation of reactive oxygen species (ROS) and antioxidant defense mechanisms in the gastrointestinal (GI) tract, the involvement of oxidative stress signaling in the initiation and progression of IBD and its relationships with genetic susceptibility and the mucosal immune response. In addition, potential therapeutic strategies for IBD that target oxidative stress signaling are reviewed and discussed. Though substantial progress has been made in understanding the role of oxidative stress in IBD in humans and experimental animals, the underlying mechanisms are still not well defined. Thus, further studies are needed to validate how oxidative stress signaling is involved in and contributes to the development of IBD.
Collapse
Affiliation(s)
- Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Ziling Wang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|