1
|
Han D, Wang F, Jiang Q, Qiao Z, Zhuang Y, An Q, Li Y, Tang Y, Li C, Shen D. Enhancing Cardioprotection Through Neutrophil-Mediated Delivery of 18β-Glycyrrhetinic Acid in Myocardial Ischemia/Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406124. [PMID: 39264272 PMCID: PMC11558124 DOI: 10.1002/advs.202406124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/08/2024] [Indexed: 09/13/2024]
Abstract
Myocardial ischemia/reperfusion injury (MI/RI) generates reactive oxygen species (ROS) and initiates inflammatory responses. Traditional therapies targeting specific cytokines or ROS often prove inadequate. An innovative drug delivery system (DDS) is developed using neutrophil decoys (NDs) that encapsulate 18β-glycyrrhetinic acid (GA) within a hydrolyzable oxalate polymer (HOP) and neutrophil membrane vesicles (NMVs). These NDs are responsive to hydrogen peroxide (H2O2), enabling controlled GA release. Additionally, NDs adsorb inflammatory factors, thereby reducing inflammation. They exhibit enhanced adhesion to inflamed endothelial cells (ECs) and improved penetration. Once internalized by cardiomyocytes through clathrin-mediated endocytosis, NDs protect against ROS-induced damage and inhibit HMGB1 translocation. In vivo studies show that NDs preferentially accumulate in injured myocardium, reducing infarct size, mitigating adverse remodeling, and enhancing cardiac function, all while maintaining favorable biosafety profiles. This neutrophil-based system offers a promising targeted therapy for MI/RI by addressing both inflammation and ROS, holding potential for future clinical applications.
Collapse
Affiliation(s)
- Dongjian Han
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Fuhang Wang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Qingjiao Jiang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Zhentao Qiao
- Department of Vascular and Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Yuansong Zhuang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Quanxu An
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Yuhang Li
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Yazhe Tang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Chenyao Li
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| | - Deliang Shen
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhou450018China
| |
Collapse
|
2
|
Nath A, Ghosh S, Bandyopadhyay D. Role of melatonin in mitigation of insulin resistance and ensuing diabetic cardiomyopathy. Life Sci 2024; 355:122993. [PMID: 39154810 DOI: 10.1016/j.lfs.2024.122993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Addressing insulin resistance or hyperinsulinemia might offer a viable treatment approach to stop the onset of diabetic cardiomyopathy, as these conditions independently predispose to the development of the disease, which is initially characterized by diastolic abnormalities. The development of diabetic cardiomyopathy appears to be driven mainly by insulin resistance or impaired insulin signalling and/or hyperinsulinemia. Oxidative stress, hypertrophy, fibrosis, cardiac diastolic dysfunction, and, ultimately, systolic heart failure are the outcomes of these pathophysiological alterations. Melatonin is a ubiquitous indoleamine, a widely distributed compound secreted mainly by the pineal gland, and serves a variety of purposes in almost every living creature. Melatonin is found to play a leading role by improving myocardial cell metabolism, decreasing vascular endothelial cell death, reversing micro-circulation disorders, reducing myocardial fibrosis, decreasing oxidative and endoplasmic reticulum stress, regulating cell autophagy and apoptosis, and enhancing mitochondrial function. This review highlights a relationship between insulin resistance and associated cardiomyopathy. It explores the potential therapeutic strategies offered by the neurohormone melatonin, an important antioxidant that plays a leading role in maintaining glucose homeostasis by influencing the glucose transporters independently and through its receptors. The vast distribution of melatonin receptors in the body, including beta cells of pancreatic islets, asserts the role of this indole molecule in maintaining glucose homeostasis. Melatonin controls the production of GLUT4 and/or the phosphorylation process of the receptor for insulin and its intracellular substrates, activating the insulin-signalling pathway through its G-protein-coupled membrane receptors.
Collapse
Affiliation(s)
- Anupama Nath
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science, Technology and Agriculture, 92 APC Road, Kolkata 700 009, India
| | - Songita Ghosh
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science, Technology and Agriculture, 92 APC Road, Kolkata 700 009, India
| | - Debasish Bandyopadhyay
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science, Technology and Agriculture, 92 APC Road, Kolkata 700 009, India.
| |
Collapse
|
3
|
Gao Q, Dai Z, Yang X, Liu C, Liu G. Experimental study on small molecule combinations inducing reprogramming of rat fibroblasts into functional neurons. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:498-508. [PMID: 39183062 PMCID: PMC11375488 DOI: 10.3724/zdxbyxb-2024-0007] [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] [Indexed: 08/27/2024]
Abstract
OBJECTIVES To establish a methodological system for reprogramming rat embryonic fibroblasts (REF) into chemically induced neurons (ciNCs) via small molecule compounds to provide safe and effective donor cells for treatment of neurodegenerative diseases. METHODS Based on the method established by PEI Gang's research group to directly reprogram human fibroblasts into neurons, the induction medium and maturation medium was optimized by replacing the coating solution, mitigating oxidative stress injury, adding neurogenic protective factors, adjusting the concentration of trichothecenes, performing small-molecule removal experiments, and carrying out immunofluorescence and Western blotting on cells at different stages of induction to validate the effect of induction. RESULTS When the original protocol was used for induction, the cell survival rate was (34.24±2.77)%. After replacing the coating solution gelatin with matrigel, the cell survival rate increased to (45.41±4.27)%; after adding melatonin, the cell survival rate increased to (67.95±5.61)% and (23.43±1.42)% were transformed into neural-like cells; after adding the small molecule P7C3-A20, the cell survival rate was further increased to (76.27±1.41)%, and (39.72±4.75)% of the cells were transformed into neural-like cells. When the concentration of trichothecene was increased to 30 μmol/L, the proportion of neural-like cells reached (55.79±1.90)%; after the removal of SP600125, (86.96±2.15)% of the cells survived, and the rate of neural-like cell production increased to (63.43±1.60)%. With the optimized protocol, REF could be successfully induced into ciNC through the neural precursor cell stage, in which the neural precursor cells were able to highly express the neural precursor cell markers SRY-related HMG-box gene 2 (Sox2) and paired box 6 (Pax6) as well as neuron-specific marker tubulin 1 (Tuj1), while the expression of fiber-associated protein vimentin was reduced. After two weeks of induction of neural precursor cells in a maturation medium, most cells displayed neuronal-like cell morphology. The induced ciNCs were able to highly express the mature neuronal surface markers Tuj1 and microtubule-associated protein 2 (MAP2), while the expression of vimentin was reduced. CONCLUSIONS The small molecule combinations optimized in this study can reprogram REF to ciNCs under normoxic conditions.
Collapse
Affiliation(s)
- Qunwei Gao
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China.
| | - Zhenjia Dai
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China
| | - Xinkang Yang
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China
| | - Changqing Liu
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu 233030, Anhui Province, China
| | - Gaofeng Liu
- School of Life Sciences, Bengbu Medical University, Bengbu 233030, Anhui Province, China. ,
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu 233030, Anhui Province, China. ,
| |
Collapse
|
4
|
Xu X, Zhou W, Wang Y, Wang Z, Zhang X, Zhang X, Tian S, Wu G. Enhanced external counterpulsation improves sleep quality in chronic insomnia: A pilot randomized controlled study. J Affect Disord 2024; 350:608-617. [PMID: 38218261 DOI: 10.1016/j.jad.2024.01.090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/05/2023] [Accepted: 01/07/2024] [Indexed: 01/15/2024]
Abstract
PURPOSE To investigate the short-term efficacy of enhanced external counterpulsation (EECP) on chronic insomnia. METHODS This is a pilot randomized, participant-blind, and sham-controlled study. Forty-six participants with chronic insomnia were randomly assigned in a 1:1 ratio to receive EECP or sham EECP intervention (total of 35 sessions with 45 min each). The primary outcome was Pittsburgh Sleep Quality Index (PSQI). The secondary outcomes included sleep diary, Hospital Anxiety and Depression Scale (HADS), Short-Form Health Survey (SF12), flow mediated dilation (FMD), serum biomarkers of melatonin, cortisol, interleukin-6, and high sensitivity C-reactive protein. Outcomes were assessed after treatment and at 3-month follow-up. RESULTS The PSQI was significantly decreased in both EECP and sham groups after 35-session intervention (13.74 to 6.96 in EECP and 13.04 to 9.48 in sham), and EECP decreased PSQI more than sham EECP (p = 0.009). PSQI in two groups kept improved at 3-month follow-up. After treatment, the total sleep time, sleep efficiency, FMD value and SF12 mental component of EECP group were significantly improved, and group differences were found for these outcomes. At follow-up, total sleep time, sleep efficiency and SF12 mental component of EECP group remained improved, and group difference for SF12 mental component was found. Post-treatment and follow-up HADS-A significantly decreased in both groups, with no differences between groups. Post-treatment serum biomarkers showed no differences within and between groups. LIMITATION Lack of objective sleep measurement. CONCLUSION EECP could improve sleep quality and mental quality of life in chronic insomnia and the therapeutic effect maintained for 3 months.
Collapse
Affiliation(s)
- Xiuli Xu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China; Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Sun Yat-sen University, Shenzhen, Guangdong 518033, China; National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, Guangdong 528478, China
| | - Wenjuan Zhou
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Yinfen Wang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Zhenyu Wang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China; Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Sun Yat-sen University, Shenzhen, Guangdong 518033, China; National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, Guangdong 528478, China
| | - Xiaocong Zhang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China; Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Sun Yat-sen University, Shenzhen, Guangdong 518033, China; National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, Guangdong 528478, China
| | - Xinxia Zhang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China; Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Shuai Tian
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China; Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Sun Yat-sen University, Shenzhen, Guangdong 518033, China; National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, Guangdong 528478, China.
| | - Guifu Wu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, China; Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Sun Yat-sen University, Shenzhen, Guangdong 518033, China; National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, Guangdong 528478, China.
| |
Collapse
|
5
|
Zhao S, Dong Y, Li Y, Wang Z, Chen Y, Dong Y. Melatonin Alleviates Lipopolysaccharide-Induced Abnormal Pregnancy through MTNR1B Regulation of m6A. Int J Mol Sci 2024; 25:733. [PMID: 38255808 PMCID: PMC10815701 DOI: 10.3390/ijms25020733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Pregnancy is a highly intricate and delicate process, where inflammation during early stages may lead to pregnancy loss or defective implantation. Melatonin, primarily produced by the pineal gland, exerts several pharmacological effects. N6-methyladenosine (m6A) is the most prevalent mRNA modification in eukaryotes. This study aimed to investigate the association between melatonin and m6A during pregnancy and elucidate the underlying protective mechanism of melatonin. Melatonin was found to alleviate lipopolysaccharide (LPS)-induced reductions in the number of implantation sites. Additionally, it mitigated the activation of inflammation, autophagy, and apoptosis pathways, thereby protecting the pregnancy process in mice. The study also revealed that melatonin regulates uterine m6A methylation levels and counteracts abnormal changes in m6A modification of various genes following LPS stimulation. Furthermore, melatonin was shown to regulate m6A methylation through melatonin receptor 1B (MTNR1B) and subsequently modulate inflammation, autophagy, and apoptosis through m6A. In conclusion, our study demonstrates that melatonin protects pregnancy by influencing inflammation, autophagy, and apoptosis pathways in an m6A-dependent manner via MTNR1B. These findings provide valuable insights into the mechanisms underlying melatonin's protective effects during pregnancy and may have implications for potential therapeutic strategies in managing pregnancy-related complications.
Collapse
Affiliation(s)
- Shisu Zhao
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Yanjun Dong
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Yuanyuan Li
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Zixu Wang
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Yaoxing Chen
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
| | - Yulan Dong
- Laboratory of Neurobiology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (S.Z.); (Y.D.); (Y.L.); (Z.W.)
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, China Agricultural University, Beijing 100193, China
| |
Collapse
|
6
|
Reiter RJ, Sharma R, Chuffa LGDA, Simko F, Dominguez-Rodriguez A. Mitochondrial Melatonin: Beneficial Effects in Protecting against Heart Failure. Life (Basel) 2024; 14:88. [PMID: 38255703 PMCID: PMC10820220 DOI: 10.3390/life14010088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular disease is the cause of physical infirmity and thousands of deaths annually. Typically, during heart failure, cardiomyocyte mitochondria falter in terms of energy production and metabolic processing. Additionally, inflammation and the accumulation of non-contractile fibrous tissue contribute to cardiac malfunction. Melatonin, an endogenously produced molecule, experimentally reduces the initiation and progression of atherosclerotic lesions, which are often the basis of coronary artery disease. The current review critically analyzes published data related to the experimental use of melatonin to forestall coronary artery pathologies. Collectively, these studies document melatonin's anti-atherosclerotic actions in reducing LDL oxidation and triglyceride levels, lowering endothelial malfunction, limiting adhesion molecule formation, preventing macrophage polarization to the M1 pro-inflammatory phenotype, changing cellular metabolism, scavenging destructive reactive oxygen species, preventing the proliferation and invasion of arterial smooth muscle cells into the lesioned area, restricting the ingrowth of blood vessels from the vasa vasorum, and solidifying the plaque cap to reduce the chance of its rupture. Diabetic hyperglycemia, which aggravates atherosclerotic plaque formation, is also inhibited by melatonin supplementation in experimental animals. The potential value of non-toxic melatonin as a possible inhibitor of cardiac pathology in humans should be seriously considered by performing clinical trials using this multifunctional molecule.
Collapse
Affiliation(s)
- Russel J. Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Ramaswamy Sharma
- Applied Biomedical Sciences, School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX 78235, USA
| | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology-IBB/UNESP, Institute of Biosciences of Botucatu, Universidade Estadual Paulista, Botucatu 18618-689, São Paulo, Brazil;
| | - Fedor Simko
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia;
| | | |
Collapse
|
7
|
Ma Y, Ma J, Lu L, Xiong X, Shao Y, Ren J, Yang J, Liu J. Melatonin Restores Autophagic Flux by Activating the Sirt3/TFEB Signaling Pathway to Attenuate Doxorubicin-Induced Cardiomyopathy. Antioxidants (Basel) 2023; 12:1716. [PMID: 37760018 PMCID: PMC10525655 DOI: 10.3390/antiox12091716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Doxorubicin (DOX) chemotherapy in cancer patients increases the risk of the occurrence of cardiac dysfunction and even results in congestive heart failure. Despite the great progress of pathology in DOX-induced cardiomyopathy, the underlying molecular mechanisms remain elusive. Here, we investigate the protective effects and the underlying mechanisms of melatonin in DOX-induced cardiomyopathy. Our results clearly show that oral administration of melatonin prevented the deterioration of cardiac function caused by DOX treatment, which was evaluated by left ventricular ejection fraction and fractional shortening as well as cardiac fibrosis. The ejection fraction and fractional shortening in the DOX group were 49.48% and 25.5%, respectively, while melatonin treatment increased the ejection fraction and fractional shortening to 60.33 and 31.39 in wild-type mice. Cardiac fibrosis in the DOX group was 3.97%, while melatonin reduced cardiac fibrosis to 1.95% in wild-type mice. Sirt3 is a mitochondrial deacetylase and shows protective effects in diverse cardiovascular diseases. Therefore, to test whether Sirt3 is a key factor in protection, Sirt3 knockout mice were used, and it was found that the protective effects of melatonin in DOX-induced cardiomyopathy were partly abolished. Further analysis revealed that Sirt3 and its downstream molecule TFEB were downregulated in response to DOX treatment, while melatonin administration was able to significantly enhance the expressions of Sirt3 and TFEB. Our in vitro study demonstrated that melatonin enhanced lysosomal function by increasing the Sirt3-mediated increase at the TFEB level, and the accumulation of autolysosomes induced by DOX treatment was attenuated. Thus, autophagic flux disrupted by DOX treatment was restored by melatonin supplementation. In summary, our results demonstrate that melatonin protects the heart against DOX injury by the restoration of autophagic flux via the activation of the Sirt3/TFEB signaling pathway.
Collapse
Affiliation(s)
- Yanyan Ma
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China
| | - Jipeng Ma
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China
| | - Linhe Lu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China
| | - Xiang Xiong
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China
| | - Yalan Shao
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Yang
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| |
Collapse
|
8
|
Suzen S, Saso L. Melatonin as mitochondria-targeted drug. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:249-276. [PMID: 37437980 DOI: 10.1016/bs.apcsb.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Oxidative damage is associated to numerous diseases as well as aging development. Mitochondria found in most eukaryotic organisms to create the energy of the cell, generate free radicals during its action and they are chief targets of the oxidants. Mitochondrial activities outspread outside the borders of the cell and effect human physiology by modulating interactions among cells and tissues. Therefore, it has been implicated in several human disorders and conditions. Melatonin (MLT) is an endogenously created indole derivative that modifies several tasks, involving mitochondria-associated activities. These possessions make MLT a powerful defender against a selection of free radical-linked disorders. MLT lessens mitochondrial anomalies causing from extreme oxidative stress and may improve mitochondrial physiology. It is a potent and inducible antioxidant for mitochondria. MLT is produced in mitochondria of conceivably of all cells and it also appears to be a mitochondria directed antioxidant which has related defensive properties as the synthesized antioxidant molecules. This chapter summarizes the suggestion that MLT is produced in mitochondria as well as disorders of mitochondrial MLT production that may associate to a number of mitochondria-linked diseases. MLT as a mitochondria-targeted drug is also discussed.
Collapse
Affiliation(s)
- Sibel Suzen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Tandogan, Ankara, Turkey.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
9
|
Chen Y, Zhang SP, Gong WW, Zheng YY, Shen JR, Liu X, Gu YH, Shi JH, Meng GL. Novel Therapeutic Potential of Retinoid-Related Orphan Receptor α in Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24043462. [PMID: 36834872 PMCID: PMC9959049 DOI: 10.3390/ijms24043462] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/18/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
The retinoid-related orphan receptor α (RORα) is one subfamily of nuclear hormone receptors (NRs). This review summarizes the understanding and potential effects of RORα in the cardiovascular system and then analyzes current advances, limitations and challenges, and further strategy for RORα-related drugs in cardiovascular diseases. Besides regulating circadian rhythm, RORα also influences a wide range of physiological and pathological processes in the cardiovascular system, including atherosclerosis, hypoxia or ischemia, myocardial ischemia/reperfusion injury, diabetic cardiomyopathy, hypertension, and myocardial hypertrophy. In terms of mechanism, RORα was involved in the regulation of inflammation, apoptosis, autophagy, oxidative stress, endoplasmic reticulum (ER) stress, and mitochondrial function. Besides natural ligands for RORα, several synthetic RORα agonists or antagonists have been developed. This review mainly summarizes protective roles and possible mechanisms of RORα against cardiovascular diseases. However, there are also several limitations and challenges of current research on RORα, especially the difficulties on the transformability from the bench to the bedside. By the aid of multidisciplinary research, breakthrough progress on RORα-related drugs to combat cardiovascular disorder may appear.
Collapse
Affiliation(s)
- Yun Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Shu-Ping Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Wei-Wei Gong
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yang-Yang Zheng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Jie-Ru Shen
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases, Nantong University, Nantong 226001, China
| | - Xiao Liu
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases, Nantong University, Nantong 226001, China
| | - Yun-Hui Gu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Jia-Hai Shi
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Research Institution of Translational Medicine in Cardiothoracic Diseases, Nantong University, Nantong 226001, China
- Correspondence: (J.-H.S.); (G.-L.M.); Tel.: +86-513-8116-0901 (J.-H.S.); +86-513-8505-1726 (G.-L.M.); Fax: +86-513-8116-0901 (J.-H.S.); +86-513-8505-1728 (G.-L.M.)
| | - Guo-Liang Meng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China
- Correspondence: (J.-H.S.); (G.-L.M.); Tel.: +86-513-8116-0901 (J.-H.S.); +86-513-8505-1726 (G.-L.M.); Fax: +86-513-8116-0901 (J.-H.S.); +86-513-8505-1728 (G.-L.M.)
| |
Collapse
|
10
|
Liu Y, Wang D, Li T, Xu L, Li Z, Bai X, Tang M, Wang Y. Melatonin: A potential adjuvant therapy for septic myopathy. Biomed Pharmacother 2023; 158:114209. [PMID: 36916434 DOI: 10.1016/j.biopha.2022.114209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/24/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Septic myopathy, also known as ICU acquired weakness (ICU-AW), is a characteristic clinical symptom of patients with sepsis, mainly manifested as skeletal muscle weakness and muscular atrophy, which affects the respiratory and motor systems of patients, reduces the quality of life, and even threatens the survival of patients. Melatonin is one of the hormones secreted by the pineal gland. Previous studies have found that melatonin has anti-inflammatory, free radical scavenging, antioxidant stress, autophagic lysosome regulation, mitochondrial protection, and other multiple biological functions and plays a protective role in sepsis-related multiple organ dysfunction. Given the results of previous studies, we believe that melatonin may play an excellent regulatory role in the repair and regeneration of skeletal muscle atrophy in septic myopathy. Melatonin, as an over-the-counter drug, has the potential to be an early, complementary treatment for clinical trials. Based on previous research results, this article aims to critically discuss and review the effects of melatonin on sepsis and skeletal muscle depletion.
Collapse
Affiliation(s)
- Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Dongfang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Ligang Xu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Manli Tang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| | - Yuchang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
11
|
Akhlagh S, Jouybar R, Zohoori K, Khademi S, Mani A, reza Akhlagh S, Asadpour E. The effect of melatonin on cognitive functions following coronary artery bypass grafting: A triple-blind randomized-controlled trial. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2023; 28:14. [PMID: 37064795 PMCID: PMC10098135 DOI: 10.4103/jrms.jrms_118_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 08/23/2022] [Accepted: 09/26/2022] [Indexed: 03/18/2023]
Abstract
Background Cognitive dysfunction presents one of the chief causes of postoperative morbidity. Melatonin as a neurohormone can improve neurocognitive functioning and sleep disorders. We evaluated the effect of melatonin on the postoperative cognitive function of patients undergoing coronary artery bypass grafting (CABG). Materials and Methods A triple-blind randomized-controlled trial was conducted on 66 CABG candidates in Namazee Hospital (Shiraz, Iran). Patients were assigned equally into two groups receiving melatonin 10 mg or a placebo daily for 4 weeks before surgery and 2 days after surgery in the intensive care unit. The Mini-Mental State Examination (MMSE), Tower of London (ToL), and Wechsler Adults Intelligence Scale-Revised (WAIS-R) cognitive function tests were performed in both groups 4 weeks before surgery (time point 1), 2 days after surgery (time point 2), and 6 weeks after initial administration of melatonin (time point 3). Results The mean change score (time point 3-time point 1) differed significantly between the two groups in the MMSE (P ≤ 0.001), ToL total score (P = 0.001), and WAIS-R general IQ (P ≤ 0.001), picture completion (P ≤ 0.001), vocabulary (P = 0.024), and digit span (P = 0.01). On the other hand, no significant differences were detected in the WAIS-R block design, ToL total time delay, ToL total lab, and ToL total result scores. Conclusion The MMSE and WAIS-R tests revealed that melatonin might have prophylactic effects against postoperative cognitive disturbance in patients undergoing elective CABG.
Collapse
|
12
|
ECG Markers of Acute Melatonin Treatment in a Porcine Model of Acute Myocardial Ischemia. Int J Mol Sci 2022; 23:ijms231911800. [PMID: 36233101 PMCID: PMC9570319 DOI: 10.3390/ijms231911800] [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: 08/20/2022] [Revised: 09/23/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
In myocardial ischemia, melatonin confers antiarrhythmic action, but its electrocardiographic expression is unclear. We aimed to evaluate the effects of melatonin treatment on electrocardiogram (ECG) parameters reflecting major arrhythmogenic factors and to test the association of these parameters with ventricular fibrillation (VF) incidence. Myocardial ischemia was induced by 40 min coronary artery occlusion in 25 anesthetized pigs. After induction of ischemia, 12 and 13 animals were given melatonin or placebo, respectively. Twelve-lead ECGs were recorded and durations of QRS, QT, Tpeak-Tend intervals and extrasystolic burden were measured at baseline and during occlusion. During ischemia, VF episodes clustered into early and delayed phases (<10 and >20 min, respectively), and QRS duration was associated with VF incidence. QT interval and extrasystolic burden did not differ between the groups. The Tpeak-Tend interval was progressively prolonged, and the prolongation was less pronounced in the treated animals. QRS duration increased, demonstrating two maxima (5−10 and 25 min, respectively). In the melatonin group, the earlier maximum was blunted, and VF development in this period was prevented. Thus, acute melatonin treatment prevented excessive prolongation of the QRS and Tpeak-Tend intervals in the porcine myocardial infarction model, and QRS duration can be used for the assessment of antiarrhythmic action of melatonin.
Collapse
|
13
|
Chen S, Li Y, Fu S, Li Y, Wang C, Sun P, Li H, Tian J, Du GQ. Melatonin alleviates arginine vasopressin-induced cardiomyocyte apoptosis via increasing Mst1-Nrf2 pathway activity to reduce oxidative stress. Biochem Pharmacol 2022; 206:115265. [DOI: 10.1016/j.bcp.2022.115265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 09/08/2022] [Accepted: 09/23/2022] [Indexed: 11/27/2022]
|
14
|
Zhang Y, Yang N, Huang X, Zhu Y, Gao S, Liu Z, Cao F, Wang Y. Melatonin Engineered Adipose-Derived Biomimetic Nanovesicles Regulate Mitochondrial Functions and Promote Myocardial Repair in Myocardial Infarction. Front Cardiovasc Med 2022; 9:789203. [PMID: 35402545 PMCID: PMC8985816 DOI: 10.3389/fcvm.2022.789203] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/31/2022] [Indexed: 12/22/2022] Open
Abstract
Myocardial infarction (MI), one type of ischemic heart disease, is a major cause of disability and mortality worldwide. Currently, extracellular vesicles (EVs) derived from adipose-derived stem cells (ADSC) have been proven to be a potentially promising therapeutic treatment for MI. However, the inconvenience of isolation, the low productivity, and the high cost of EVs greatly limits their application in clinic. In this study, we constructed novel biomimetic ADSC-derived nanovesicles (ADSC NVs) to achieve cell-free therapy for MI. Here, we firstly developed a novel Mel@NVs delivery system consisting of engineered ADSC NVs with melatonin (Mel). Then, the characterization and properties of Mel@NVs were performed. The effect of Mel@NVs on cellular oxidative stress and myocardial infarction repair was conducted. The results showed that Mel@NVs treatment under ischemia mimic condition reduced cell apoptosis from 42.59 ± 2.69% to 13.88 ± 1.77%. Moreover, this novel engineered Mel@NVs could ameliorate excessive ROS generation, promote microvessel formation, and attenuate cardiac fibrosis, which further alleviates mitochondrial dysfunction and finally enhance myocardial repair. Hence, the engineered NVs show a potential strategy for MI therapy.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Cardiology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital and Medical School of Chinese People's Liberation Army, Beijing, China
| | - Ning Yang
- Department of Cardiology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital and Medical School of Chinese People's Liberation Army, Beijing, China
| | - Xu Huang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yan Zhu
- Department of Cardiology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital and Medical School of Chinese People's Liberation Army, Beijing, China
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Shan Gao
- Department of Cardiology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital and Medical School of Chinese People's Liberation Army, Beijing, China
| | - Zhongyang Liu
- Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Feng Cao
- Department of Cardiology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital and Medical School of Chinese People's Liberation Army, Beijing, China
- Feng Cao
| | - Yabin Wang
- Department of Cardiology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital and Medical School of Chinese People's Liberation Army, Beijing, China
- *Correspondence: Yabin Wang
| |
Collapse
|
15
|
Pongkan W, Piamsiri C, Dechvongya S, Punyapornwitthaya V, Boonyapakorn C. Short-term melatonin supplementation decreases oxidative stress but does not affect left ventricular structure and function in myxomatous mitral valve degenerative dogs. BMC Vet Res 2022; 18:24. [PMID: 34996468 PMCID: PMC8740462 DOI: 10.1186/s12917-021-03125-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 12/20/2021] [Indexed: 11/27/2022] Open
Abstract
Background Cardiac wall stress and high oxidative stress are often found in cases of myxomatous mitral valve degenerative (MMVD) disease and can lead to myocardial injuries and cardiac dysfunction. Melatonin, an antioxidant, has been shown to exert cardioprotection in laboratory animal models. However, its effect on metabolic parameters and left ventricular (LV) adaptation in MMVD dogs has rarely been investigated. This clinical trial hypothesized that a melatonin supplement for 4 weeks would improve metabolic parameters, LV structure (diameters and wall thickness), and LV function in MMVD dogs. Blood profiles, echocardiograms, and oxidative stress levels were obtained from 18 dogs with MMVD stage B2 and C at baseline and after prescribed Melatonin (2 mg/kg) for 4 weeks. Eleven dogs with MMVD stage B2 and C, which received a placebo, were evaluated as a control group. Results In this clinical trial, the baseline plasma malondialdehyde (MDA) was no different between the treatment and placebo groups. The post-treatment plasma MDA levels (4.50 ± 0.63 mg/mL) in the treatment group was significantly decreased after 4 weeks of melatonin supplementation compared to pre-treatment levels (7.51 ± 1.11 mg/mL) (P = 0.038). However, blood profiles and LV structure and function investigated using echocardiography were found not to different between pre-and post-treatment in each group. No adverse effects were observed following melatonin supplementation. Conclusions This clinical trial demonstrated that a melatonin supplement for 4 weeks can attenuate oxidative stress levels in MMVD dogs, especially in MMVD stage C, but does not result in LV structural changes or LV function in MMVD dogs of either stage B2 or stage C.
Collapse
Affiliation(s)
- Wanpitak Pongkan
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand.,Integrative Research Center for Veterinary Circulatory Sciences, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand.,Veterinary Cardiopulmonary Clinic, Small Animal Hospital, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Chanon Piamsiri
- Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Sirada Dechvongya
- Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Verasak Punyapornwitthaya
- Department of Food Animal Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand
| | - Chavalit Boonyapakorn
- Integrative Research Center for Veterinary Circulatory Sciences, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand. .,Veterinary Cardiopulmonary Clinic, Small Animal Hospital, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand. .,Department of Companion Animal and Wildlife Clinic, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, 50100, Thailand.
| |
Collapse
|
16
|
Xie Y, Lou D, Zhang D. Melatonin Alleviates Age-Associated Endothelial Injury of Atherosclerosis via Regulating Telomere Function. J Inflamm Res 2021; 14:6799-6812. [PMID: 34924765 PMCID: PMC8674670 DOI: 10.2147/jir.s329020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background Atherosclerosis is an aging-related disease, partly attributed to telomerase dysfunction. This study aims to investigate whether telomere dysfunction-related vascular aging is involved in the protection mechanism of melatonin (MLT) in atherosclerosis. Methods Young and aged ApoE−/− mice were used to establish atherosclerotic mice model. H&E staining and immunofluorescence assay were performed to detect endothelial cell injury and apoptosis. Inflammatory cytokines and oxidative stress-related factors were determined using corresponding commercial assay kits. Telomerase activity was detected by TRAP assay, and SA-β-gal staining was conducted to evaluate cellular senescence. HUVECs were treated with H2O2 for 1 h to induce senescence. Western blot was performed to measure protein expression. Results An obvious vascular endothelial injury, reflected by excessive production of inflammatory cytokines, elevated ROS, MDA and SOD levels, and more apoptotic endothelial cells, was found in atherosclerotic mice, especially in aged mice, which were then greatly suppressed by MLT. In addition, telomere dysfunction and senescence occurred in atherosclerosis, especially in aged mice, while MLT significantly alleviated the conditions. CYP1A1, one of the targeted genes of MLT, was verified to be upregulated in atherosclerotic mice but downregulated by MLT. Furthermore, H2O2 induced a senescence model in HUVECs, which was accompanied with a remarkably increased cell viability loss and apoptosis rate, and a downregulated telomerase activity of HUVECs, and this phenomenon was strengthened by RHPS4, an inhibitor of telomerase activity. However, MLT could partly abolish these changes in H2O2- and RHPS4-treated HUVECs, demonstrating that MLT alleviated vascular endothelial injury by regulating senescence and telomerase activity. Conclusions Collectively, this study provided evidence for the protective role of MLT in atherosclerosis through regulating telomere dysfunction-related vascular aging.
Collapse
Affiliation(s)
- Yinghua Xie
- Department of Geriatrics, Fuzhou NO.1 Hospital Affiliated with Fujian Medical University, Fuzhou, Fujian, 350009, People's Republic of China
| | - Danfei Lou
- Department of Geriatrics, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, People's Republic of China
| | - Daimin Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210006, People's Republic of China
| |
Collapse
|
17
|
miR-190-5p Alleviates Myocardial Ischemia-Reperfusion Injury by Targeting PHLPP1. DISEASE MARKERS 2021; 2021:8709298. [PMID: 34868398 PMCID: PMC8639278 DOI: 10.1155/2021/8709298] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022]
Abstract
Objective Myocardial ischemia-reperfusion (I/R) injury (MIRI) refers to the more serious myocardial injury after blood flow recovery, which seriously affects the prognosis of patients with ischemic cardiomyopathy. This study explored the new targets for MIRI treatment by investigating the effects of miR-190-5p and its downstream target on the structure and function of myocardial cells. Methods We injected agomir miR-190-5p into the tail vein of rats to increase the expression of miR-190-5p in rat myocardial cells and made an I/R rat model by coronary artery occlusion. We used 2,3,5-triphenyl tetrazolium chloride staining, lactate dehydrogenase (LDH) detection, echocardiography, and hematoxylin-eosin (HE) staining to determine the degree of myocardial injury in I/R rats. In addition, we detected the expression of inflammatory factors and apoptosis-related molecules in rat serum and myocardial tissue to determine the level of inflammation and apoptosis in rat myocardium. Finally, we determined the downstream target of miR-190-5p by Targetscan system and dual luciferase reporter assay. Results The expression of miR-190-5p in an I/R rat myocardium was significantly lower than that in normal rats. After treatment of I/R rats with agomir miR-190-5p, the ischemic area of rat myocardium and the concentration of LDH decreased. The results of echocardiography and HE staining also found that overexpression of miR-190-5p improved the structure and function of rat myocardium. miR-190-5p was also found to improve the viability of H9c2 cells in vitro and reduce the level of apoptosis of H9c2 cells. The results of Targetscan system and dual luciferase reporter assay found that miR-190-5p targeted to inhibit pleckstrin homology domain leucine-rich repeat protein phosphatase 1 (PHLPP1). In addition, inhibition of PHLPP1 was found to improve the viability of H9c2 cells. Conclusion Therefore, miR-190-5p can reduce the inflammation and apoptosis of myocardium by targeting PHLPP1, thereby alleviating MIRI.
Collapse
|
18
|
Potentiating the Benefits of Melatonin through Chemical Functionalization: Possible Impact on Multifactorial Neurodegenerative Disorders. Int J Mol Sci 2021; 22:ijms222111584. [PMID: 34769013 PMCID: PMC8583879 DOI: 10.3390/ijms222111584] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022] Open
Abstract
Although melatonin is an astonishing molecule, it is possible that chemistry will help in the discovery of new compounds derived from it that may exceed our expectations regarding antioxidant protection and perhaps even neuroprotection. This review briefly summarizes the significant amount of data gathered to date regarding the multiple health benefits of melatonin and related compounds. This review also highlights some of the most recent directions in the discovery of multifunctional pharmaceuticals intended to act as one-molecule multiple-target drugs with potential use in multifactorial diseases, including neurodegenerative disorders. Herein, we discuss the beneficial activities of melatonin derivatives reported to date, in addition to computational strategies to rationally design new derivatives by functionalization of the melatonin molecular framework. It is hoped that this review will promote more investigations on the subject from both experimental and theoretical perspectives.
Collapse
|
19
|
Demir M, Altinoz E, Elbe H, Bicer Y, Yigitturk G, Karayakali M, Ballur AFH. Effects of pinealectomy and crocin treatment on rats with isoproterenol-induced myocardial infarction. Drug Chem Toxicol 2021; 45:2576-2585. [PMID: 34538161 DOI: 10.1080/01480545.2021.1977025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The present study aimed to analyze the effects of pinealectomy and crocin treatment in isoproterenol-induced myocardial damage. Seventy rats were divided into seven groups: control, sham control, pinealectomy (PNX), isoproterenol (ISO; 85 mg/kg on the 29th and 30th days of the experiment, subcutaneous injection), PNX + ISO, PNX + crocin (50 mg/kg/day for 30 days, intragastric administration), and PNX + ISO + crocin. PNX procedure was performed on the first day of the study. A significant increase was observed in serum cardiac damage markers (CK-MB, Troponin I) after ISO administration. ISO administration led to a significant increase in cardiac oxidative stress parameters, such as malondialdehyde (MDA) and total oxidant status (TOS), while it led to a decrease in antioxidant defense system parameters, such as reduced glutathione (GSH), superoxide dismutase (SOD), catalase (CAT) and total antioxidant status (TAS) when compared to control groups. Elevated MDA and TOS levels were observed, while reduced SOD and CAT activities, and decreased GSH and TAS levels were observed in the group that underwent PNX and ISO administration when compared to the PNX group. Furthermore, in the PNX + ISO + Crocin group, SOD and CAT activities, and GSH and TAS levels ameliorated and MDA and TOS levels were reduced with the crocin treatment when compared to the PNX + ISO group. Also, marked increases were observed in serum cardiac markers, histopathological and immunohistochemical findings after the crocin treatment. All findings demonstrated that crocin could be employed as a cardioprotective agent due to its antioxidant, anti-inflammatory, and anti-apoptotic properties.
Collapse
Affiliation(s)
- Mehmet Demir
- Department of Physiology, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - Eyup Altinoz
- Department of Medical Biochemistry, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - Hulya Elbe
- Department of Histology and Embryology, Faculty of Medicine, Mugla Sıtkı Kocman University, Mugla, Turkey
| | - Yasemin Bicer
- Department of Medical Biochemistry, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - Gurkan Yigitturk
- Department of Histology and Embryology, Faculty of Medicine, Mugla Sıtkı Kocman University, Mugla, Turkey
| | - Melike Karayakali
- Department of Medical Biochemistry, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - Arwa Fadıl Haqi Ballur
- Department of Medical Biochemistry, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| |
Collapse
|
20
|
Sedighi M, Ghorbanzadeh V, Abaszadeh S, Karimi A, Cheraghi M, Rafieian-Kopaei M, Moghimian M, Mohammadi A, Veiskarami S, Mokhayeri Y, Nazari A. Up-regulation of chemokine receptor type 4 expression in the ischemic reperfused heart by alcoholic extract of Cichorium intybus rescue the heart from ischemia injury in male rat. J Pharm Pharmacol 2021; 73:1351-1360. [PMID: 34076244 DOI: 10.1093/jpp/rgab076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 05/11/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Cichorium intybus is used in traditional medicine for various diseases including heart disease. This study aimed at evaluating the chemokine receptor type 4 up-regulation and cardioprotective effects of hydroalcoholic extract of C. intybus in a rat model of ischemic reperfusion. METHODS Animals in four groups of eight rats each received vehicle or one of three doses of C. intybus (50, 100 or 200 mg/kg/d) for 14 days. Then they were subjected to 30 min of ischemia followed by 7 days of reperfusion. At the end of the experiment, blood specimens were prepared for serum assays. The level of myocardium chemokine receptor type 4 was also measured using RT-PCR. KEY FINDINGS Cichorium intybus (CI-50) improved infarct size, episodes of the ventricular ectopic beat, ventricular tachycardia, and duration of ventricular tachycardia, QTc shortening. It also stabilized the ST segment changes and increased heart rate during ischemia. The blood pressure decreased in CI-50 group in comparison to the control and CI-200 group. C. intybus increased serum superoxide dismutase and reduced lactate dehydrogenase activity, Cardiac Troponin I and malondialdehyde levels. C. intybus led to an increase in the expression of chemokine receptor type 4. CONCLUSIONS These findings suggest that C. intybus administration before ischemia is able to induce cardioprotective effect against ischemic reperfusion injury, probably through chemokine receptor type 4 over-expression and antioxidant activity.
Collapse
Affiliation(s)
- Mehrnoosh Sedighi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Vajihe Ghorbanzadeh
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Saber Abaszadeh
- Department of clinical biochemistry, Lorestan University of Medical Science, Khorramabad, Iran
| | - Arash Karimi
- Department of Anesthesiology, Anesthesiologist, Faculty of Medicine, Lorestan University of Medical Science, Khorramabad, Iran
| | - Mostafa Cheraghi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Mahmoud Rafieian-Kopaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Siences, Shahrekord, Iran
| | - Maryam Moghimian
- Department of Physiology, Gonabad University of Medical Science, Gonabad, Iran
| | - Asghar Mohammadi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Saeid Veiskarami
- Lorestan Agricultural and Natural Resources Research and Education Center, Department of animal science, Iran
| | - Yaser Mokhayeri
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Afshin Nazari
- Department of Physiology, Lorestan University of Medical Science, Khorramabad, Iran
| |
Collapse
|
21
|
Guo Y, Gao J, Liu Y, Zhang X, An X, Zhou J, Su P. miR-451 on Myocardial Ischemia-Reperfusion in Rats by Regulating AMPK Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9933998. [PMID: 34307674 PMCID: PMC8279856 DOI: 10.1155/2021/9933998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/28/2021] [Indexed: 11/20/2022]
Abstract
Myocardial infarction is the main cause of death in patients with coronary heart disease. At present, the main method to treat cardiovascular disease is perfusion therapy. Myocardial ischemia-reperfusion will inevitably lead to reperfusion injury, which is also a major problem in the treatment of cardiovascular diseases. It has been reported that mir-451 in microRNA family participates in the protection of myocardial ischemia-reperfusion by regulating AMPK. The aim of this study was to investigate the effect of mir-451 on myocardial ischemia-reperfusion in rats by regulating AMPK signaling pathway. Sixty adult male rats were selected to establish myocardial ischemia-reperfusion animal model by ligating and loosening coronary artery. The expression level of mir-451 was regulated by injection of mir-451 virus vector and antibody, and the effect of increased or decreased mir-451 expression level on the activity of AMPK signaling pathway was detected. The myocardial infarct area and apoptosis rate of myocardial tissue were detected after 75 min ischemia-reperfusion. The results showed that when the expression level of mir-451 decreased by 15.7%, the activity index of AMPK signaling pathway was increased by 18.3%, the infarct area was reduced by 22.4%, and the apoptosis rate of myocardial cells was decreased by 25.2%. At the same time, the pathological structure of myocardial tissue was improved. Therefore, mir-451 is an inhibitor gene of AMPK signaling pathway. Reducing the expression of mir-451 can enhance the activity of AMPK signal pathway, and the increase of AMPK signal pathway activity is beneficial to reduce myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yulin Guo
- Department of Cardiac Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jie Gao
- Department of Cardiac Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yan Liu
- Department of Cardiac Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xitao Zhang
- Department of Cardiac Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiangguang An
- Department of Cardiac Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jian Zhou
- Department of Cardiac Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Pixiong Su
- Department of Cardiac Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
22
|
Huang X, Hou J, Huang S, Feng K, Yue Y, Li H, Huang S, Liang M, Chen G, Wu Z. Melatonin ameliorates myocardial injury by reducing apoptosis and autophagy of cardiomyocytes in a rat cardiopulmonary bypass model. PeerJ 2021; 9:e11264. [PMID: 33954056 PMCID: PMC8053380 DOI: 10.7717/peerj.11264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Background Myocardial injury is a frequent complication after cardiac surgery with cardiopulmonary bypass (CPB). This study aimed to test the hypothesis that melatonin could attenuate myocardial injury in a rat CPB model. Methods Eighteen male Sprague-Dawley rats were randomly divided into three groups, n = 6 for each group: the sham operation (SO) group, CPB group and melatonin group. Rats in the SO group underwent cannulation without CPB, rats in CPB group intraperitoneal injected an equal volume of vehicle daily for 7 days before being subjected to CPB and rats in melatonin group intraperitoneal injected 20 mg/kg of melatonin solution daily for 7 days before being subjected to CPB. After 120 min for CPB, the expression levels of plasma interleukin (IL) -6, IL-1β, superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), malondialdehyde (MDA), creatine kinase (CK) -MB and cardiac troponin T (cTnT) were measured. Reactive oxygen species (ROS) were detected by dihydroethidium (DHE). Apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining. Mitochondrial damage and autophagosomes were detected by electron microscopy. Apoptosis inducing factor (AIF) was detected by immunofluorescence. The expression of B cell lymphoma/leukemia2 associated X (Bax), B cell lymphoma/leukemia 2 (Bcl-2), cytochrome C (Cyto-C), cleaved caspase-9, AKT, p-AKT, signal transducer and activator of transcription 3 (STAT3), p-STAT3, LC3, P62, mechanistic target of rapamycin kinase (mTOR), p-mTOR and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were determined using western blotting. Results Melatonin significantly decreased the levels of IL-1β, IL-6, MDA, CK-MB and cTnT and increased the levels of SOD and GSH-Px, all of which were altered by CPB. Melatonin reduced cardiomyocyte superoxide production, the apoptosis index and autophagy in cardiomyocytes induced by CPB. The AKT, STAT3 and mTOR signaling pathways were activated by melatonin during CPB. Conclusion Melatonin may serve as a cardioprotective factor in CPB by inhibiting oxidative damage, apoptosis and autophagy. The AKT, STAT3 and mTOR signaling pathways were involved in this process.
Collapse
Affiliation(s)
- Xiaolin Huang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Jian Hou
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Suiqing Huang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Kangni Feng
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuan Yue
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Huayang Li
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Shaojie Huang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Mengya Liang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Guangxian Chen
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Zhongkai Wu
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Baburina Y, Lomovsky A, Krestinina O. Melatonin as a Potential Multitherapeutic Agent. J Pers Med 2021; 11:jpm11040274. [PMID: 33917344 PMCID: PMC8067360 DOI: 10.3390/jpm11040274] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/19/2022] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine, MEL) is a hormone produced by the pineal gland that was discovered many years ago. The physiological roles of this hormone in the body are varied. The beneficial effects of MEL administration may be related to its influence on mitochondrial physiology. Mitochondrial dysfunction is considered an important factor in various physiological and pathological processes, such as the development of neurodegenerative and cardiovascular diseases, diabetes, various forms of liver disease, skeletal muscle disorders, and aging. Mitochondrial dysfunction induces an increase in the permeability of the inner membrane, which leads to the formation of a permeability transition pore (mPTP) in the mitochondria. The long-term administration of MEL has been shown to improve the functional state of mitochondria and inhibit the opening of the mPTP during aging. It is known that MEL is able to suppress the initiation, progression, angiogenesis, and metastasis of cancer as well as the sensitization of malignant cells to conventional chemotherapy and radiation therapy. This review summarizes the studies carried out by our group on the combined effect of MEL with chemotherapeutic agents (retinoic acid, cytarabine, and navitoclax) on the HL-60 cells used as a model of acute promyelocytic leukemia. Data on the effects of MEL on oxidative stress, aging, and heart failure are also reported.
Collapse
|
24
|
Activation of Paraventricular Melatonin Receptor 2 Mediates Melatonin-Conferred Cardioprotection Against Myocardial Ischemia/Reperfusion Injury. J Cardiovasc Pharmacol 2021; 76:197-206. [PMID: 32433359 DOI: 10.1097/fjc.0000000000000851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Previous studies have shown that melatonin (Mel) can effectively ameliorate myocardial ischemia/reperfusion (MI/R) injury, but the mechanism is yet to be fully elucidated. Mel receptors are expressed in the paraventricular nucleus (PVN), which is also involved in regulating cardiac sympathetic nerve activity. The aim of this study was to examine whether Mel receptors in the PVN are involved in the protective effects of Mel against MI/R injury. The results of quantitative polymerase chain reaction, western blot, and immunofluorescence assays indicated that Mel receptor 2 (MT2) expression in the PVN was upregulated after MI/R. Intraperitoneal administration of Mel significantly improved post-MI/R cardiac function and reduced the infarct size, whereas shRNA silencing of MT2 in the PVN partially blocked this effect. Intraperitoneal administration of Mel reduced sympathetic nerve overexcitation caused by MI/R, whereas shRNA silencing of MT2 in the PVN partially diminished this effect. Furthermore, enzyme-linked immunosorbent assay and western blot results indicated that intraperitoneal administration of Mel lowered the levels of inflammatory cytokines in the PVN after MI/R injury, whereas the application of sh-MT2 in the PVN reduced this effect of Mel. Mel significantly reduced the levels of NF-κB after astrocyte oxygen and glucose deprivation/reoxygenation injury, and this effect was offset when MT2 was silenced. The above experimental results suggest that MT2 in the PVN partially mediated the protective effects of Mel against MI/R injury, and its underlying mechanisms may be related to postactivation amelioration of PVN inflammation and reduction of cardiac sympathetic nerve overexcitation.
Collapse
|
25
|
Rahbarghazi A, Siahkouhian M, Rahbarghazi R, Ahmadi M, Bolboli L, Mahdipour M, Haghighi L, Hassanpour M, Sokouti Nasimi F, Keyhanmanesh R. Melatonin and prolonged physical activity attenuated the detrimental effects of diabetic condition on murine cardiac tissue. Tissue Cell 2021; 69:101486. [PMID: 33453677 DOI: 10.1016/j.tice.2021.101486] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/04/2021] [Accepted: 01/04/2021] [Indexed: 12/22/2022]
Abstract
In this study, the combined effects of four-week swimming training and melatonin were examined on the oxidative response, inflammation, apoptosis, and angiogenesis capacity of cardiac tissue in the mouse model of diabetes. The mice were randomly allocated into five groups (n = 10 per group) as follows: Control; Diabetic group; Diabetic + Melatonin group; Diabetic + Exercise group; and Diabetic + Exercise + Melatonin group. 50 mg/kg streptozotocin was intraperitoneally administrated. In melatonin-treated groups, melatonin was injected intraperitoneally at 3 mg/kg body weight for four weeks and twice weekly. Swimming exercises were performed for four weeks. We measured cardiac superoxide dismutase, glutathione peroxidase enzymes, malondialdehyde, and total antioxidant capacity. The expression of tumor necrosis factor-α, Caspase‑3, Sirtuin1, and Connexin-43 was measured using real-time PCR analysis. The vascular density was analyzed by immunohistochemistry using CD31 and α-smooth muscle actin antibodies. The combination of melatonin and exercise elevated cardiac superoxide dismutase, glutathione peroxidase coincided with the reduction of malondialdehyde and increase of total antioxidant capacity as compared to the diabetic mice (p < 0.05). In Diabetic + Exercise + Melatonin mice, tumor necrosis factor-α, Caspase‑3 was significantly down-regulated compared to the Diabetic group (p < 0.05). Melatonin and exercise suppressed the expression of Connexin-43 and Sirtuin1 in diabetic mice in comparison with the control mice (p < 0.05). H & E staining showed necrosis and focal hyperemia reduction in the Diabetic + Exercise + Melatonin group compared to the Diabetic group. Data showed a decrease of CD31+ and α-smooth muscle actin+ vessels in the Diabetic group as compared to the normal samples (p < 0.05). The number of CD31+ vessels, but not α-smooth muscle actin+ type, increased in the Diabetic + Exercise + Melatonin group compared to the Diabetic mice. These data demonstrated that exercise along with melatonin administration could diminish the detrimental effects of diabetes on cardiac tissue via using different mechanisms.
Collapse
Affiliation(s)
- Afshin Rahbarghazi
- Department of Physical Education and Sports Sciences, Faculty of Educational Science and Psychology, University of Mohaghegh Ardabil, Ardabil, Iran; Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marefat Siahkouhian
- Department of Physical Education and Sports Sciences, Faculty of Educational Science and Psychology, University of Mohaghegh Ardabil, Ardabil, Iran.
| | - Reza Rahbarghazi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahdi Ahmadi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Lotfali Bolboli
- Department of Physical Education and Sports Sciences, Faculty of Educational Science and Psychology, University of Mohaghegh Ardabil, Ardabil, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Haghighi
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Hassanpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Rana Keyhanmanesh
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
Ishtiaq A, Ali T, Bakhtiar A, Bibi R, Bibi K, Mushtaq I, Li S, Khan W, Khan U, Anis RA, Anees M, Sultan A, Murtaza I. Melatonin abated Bisphenol A-induced neurotoxicity via p53/PUMA/Drp-1 signaling. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:17789-17801. [PMID: 33398767 DOI: 10.1007/s11356-020-12129-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA), an endocrine disruptor, is widely used in the manufacture of different daily life products. Accumulating evidence supports the association between the increasing incidence of neurodegenerative diseases and the BPA level in the environment. In the present study, we aimed to evaluate the neuroprotective role of melatonin against BPA-induced mitochondrial dysfunction-mediated apoptosis in the brain. Herein, adult Sprague Dawley rats were administrated (subcutaneously) with BPA (100 μg/kg BW, 1 mg/kg BW, and 10 mg/kg BW) and melatonin (4 mg/kg BW) for 16 days. Our results showed BPA exposure significantly increased the oxidative stress as demonstrated by increased free radicals (ROS), TBARs level, disrupted cellular architecture, and decreased antioxidant enzymes including SOD, CAT, APX, POD, and GSH levels. Additionally, BPA treatment increased the expression of PUMA, p53, and Drp-1 resulting in apoptosis in the brain tissue of rats. However, melatonin treatment significantly attenuated BPA-induced toxic effects by scavenging ROS, boosting antioxidant enzyme activities, and interestingly enervated brain apoptosis by normalizing p53, PUMA, and Drp-1 expressions at both transcriptional and translational level. Moreover, the brain tissue histology also revealed the therapeutic potential of melatonin by normalizing the cellular architecture. Conclusively, our finding suggests that melatonin could alleviate oxidative stress and mitochondrial dysfunction-linked apoptosis, rendering its neuroprotective potential against BPA-induced toxicity.
Collapse
Affiliation(s)
- Ayesha Ishtiaq
- Signal Transduction Laboratory, Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Tahir Ali
- Signal Transduction Laboratory, Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Attia Bakhtiar
- Signal Transduction Laboratory, Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Robina Bibi
- Signal Transduction Laboratory, Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Kinza Bibi
- Signal Transduction Laboratory, Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Iram Mushtaq
- Signal Transduction Laboratory, Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Wajiha Khan
- Department of Biotechnology, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Uzma Khan
- Faculty of Biological Sciences, Hazara University, Mansehra, KPK, Pakistan
| | - Riffat Aysha Anis
- Institute of Diet and Nutritional Sciences, The University of Lahore, Islamabad Campus, Islamabad, Pakistan
| | - Mariam Anees
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Aneesa Sultan
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Iram Murtaza
- Signal Transduction Laboratory, Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| |
Collapse
|
27
|
Overexpression of TGR5 alleviates myocardial ischemia/reperfusion injury via AKT/GSK-3β mediated inflammation and mitochondrial pathway. Biosci Rep 2021; 40:221795. [PMID: 31909787 PMCID: PMC6981096 DOI: 10.1042/bsr20193482] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/26/2019] [Accepted: 12/28/2019] [Indexed: 12/31/2022] Open
Abstract
Ischemia/reperfusion (I/R) injury reduces cell proliferation, triggers inflammation, promotes cell apoptosis and necrosis, which are the leading reasons of morbidity and mortality in patients with cardiac disease. TGR5 is shown to express in hearts, but its functional role in I/R-induced myocardial injury is unclear. In the present study, we aimed to explore the underlying molecular mechanism of TGR5 in hypoxia/reoxygenation (H/R)-induced cardiomyocyte injury in vitro. The results showed that TGR5 was significantly up-regulated in H9C2 (rat cardiomyocyte cells) and human cardiomyocytes (HCMs) after H/R. Overexpression of TGR5 significantly improved cell proliferation, alleviated apoptosis rate, the activities of caspase-3, cleaved caspases-3 and Bax protein expression levels, and increased Bcl-2 level. Overexpression of TGR5 significantly up-regulated ROS generation, stabilized the mitochondrial membrane potential (MMP), and reduced the concentration of intracellular Ca2+ as well as cytosolic translocation of mitochondrial cytochrome c (cyto-c). Meanwhile, overexpressed TGR5 also enhanced the mRNA and protein levels of interleukin (IL)-10, and decreased the mRNA and protein levels of IL-6 and tumor necrosis factor α (TNF-α). The shTGR5+H/R group followed opposite trends. In addition, overexpressed TGR5 induced an increase in the levels of p-AKT and p-GSK-3β. The protective effects of TGR5 were partially reversed by AKT inhibitor MK-2206. Taken together, these results suggest that TGR5 attenuates I/R-induced mitochondrial dysfunction and cell apoptosis as well as inflammation, and these protections may through AKT/GSK-3β pathway.
Collapse
|
28
|
Su ZDZ, Wei XB, Fu YB, Xu J, Wang ZH, Wang Y, Cao JF, Huang JL, Yu DQ. Melatonin alleviates lipopolysaccharide-induced myocardial injury by inhibiting inflammation and pyroptosis in cardiomyocytes. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:413. [PMID: 33842634 PMCID: PMC8033388 DOI: 10.21037/atm-20-8196] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Melatonin (MT) has been shown to protect against various cardiovascular diseases. However, the effect of MT on lipopolysaccharide (LPS)-induced myocardial injury is poorly understood. This study aims to evaluate the effects of MT on LPS-induced myocardial injury in vitro. Methods H9C2 cells were divided into a control group, MT group, LPS group, and MT + LPS group. The control group was treated with sterile saline solution, the LPS group received 8 µg/mL LPS for 24 h, MT + LPS cells were pretreated with 200 µmol/L MT for 2 h then with 8 µg/mL LPS for 24 h, and the MT group received only 200 µmol/L MT for 2 h. The CCK-8 assay and lactate dehydrogenase (LDH) activity assay were used to analyze cell viability and LDH release, respectively. Intracellular reactive oxygen species (ROS) and the rate of pyroptosis were measured using the fluorescent probe dichloro-dihydro-fluorescein diacetate (DCFH-DA) and propidium iodide (PI) staining, respectively. The cell supernatants were used to measure the levels of inflammatory cytokines, including IL-6, TNF-α, and IL-1β by enzyme-linked immunosorbent assay (ELISA). The protein levels of iNOS, COX-2, NF-κB, p-NF-κB, NLRP3, caspase-1, and GSDMD were detected by western blot. Results MT pretreatment significantly improved LPS-induced myocardial injury by inhibiting inflammation and pyroptosis in H9C2 cells. Moreover, MT inhibited the activation of the NF-κB pathway, and reduced the expression of inflammation-related proteins (iNOS and COX-2), and pyroptosis-related proteins (NLRP3, caspase-1, and GSDMD). Conclusions Our data suggests that MT can alleviate LPS-induced myocardial injury, providing novel insights into the treatment of sepsis-induced myocardial dysfunction.
Collapse
Affiliation(s)
- Ze-Da-Zhong Su
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xue-Biao Wei
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yan-Bin Fu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jia Xu
- Department of Emergency, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhong-Hua Wang
- Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yu Wang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jian-Feng Cao
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jie-Leng Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dan-Qing Yu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
29
|
Veeradittakit J, Jumrus S, Sringarm K, Prom‐u‐thai C. Improving nutritional values in purple rice through germination and parboiling processes. J FOOD PROCESS PRES 2021. [DOI: 10.1111/jfpp.14979] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Jeeraporn Veeradittakit
- Division of Agronomy, Department of Plant and Soil Sciences, Faculty of Agriculture Chiang Mai University Chiang Mai Thailand
| | - Suchada Jumrus
- Division of Agronomy, Department of Plant and Soil Sciences, Faculty of Agriculture Chiang Mai University Chiang Mai Thailand
| | - Korawan Sringarm
- Department of Animal and Aquatic Science, Faculty of Agriculture Chiang Mai University Chiang Mai Thailand
| | - Chanakan Prom‐u‐thai
- Division of Agronomy, Department of Plant and Soil Sciences, Faculty of Agriculture Chiang Mai University Chiang Mai Thailand
- Lanna Rice Research Center Chiang Mai University Chiang Mai Thailand
| |
Collapse
|
30
|
Peng CL, Jiang N, Zhao JF, Liu K, Jiang W, Cao PG. Metformin relieves H/R-induced cardiomyocyte injury through miR-19a/ACSL axis - possible therapeutic target for myocardial I/R injury. Toxicol Appl Pharmacol 2021; 414:115408. [PMID: 33476677 DOI: 10.1016/j.taap.2021.115408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/10/2021] [Accepted: 01/13/2021] [Indexed: 10/22/2022]
Abstract
This study proposed to investigate the function of miR-19a/ACSL axis in hypoxia/reoxygenation (H/R)-induced myocardial injury and determine whether metformin exerts its protective effect via miR-19a/ACSL axis. Firstly, bioinformatics analysis of data from Gene Expression Omnibus (GEO) database indicated that miR-19a was downregulated in patients with myocardial infarction (MI) compared to that in control group. H/R model was constructed with AC16 cells in vitro. qRT-PCR assay revealed that miR-19a was downregulated in H/R-treated AC16 cells. Then, CCK-8 assay demonstrated that upregulation of miR-19a significantly alleviated H/R-induced decline of cell viability. Moreover, bioinformatics prediction, western blotting and dual-luciferase reporter assays were performed to check the target genes of miR-19a, and ACSL1 was determined as a downstream target gene of miR-19a. Besides, the analysis based on Comparative Toxicogenomics Database (CTD) suggested that metformin targeting ACSL1 can be used as a potential drug for further research. Biological function experiments in vitro revealed that H/R markedly declined the viability and elevated the apoptosis of AC16 cells, while metformin can significantly mitigate these effects. Furthermore, overexpression of miR-19a significantly strengthened the beneficial effect of metformin on H/R-induced AC16 cells injury, which can be reversed by upregulation of ACSL1. In conclusion, metformin can alleviate H/R-induced cells injury via regulating miR-19a/ACSL axis, which lays a foundation for identifying novel targets for myocardial I/R injury therapy.
Collapse
Affiliation(s)
- Cai-Liang Peng
- Department of Cardiovascular Diseases, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Ning Jiang
- Department of Cardiovascular Diseases, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Jian-Fei Zhao
- Department of Cardiovascular Diseases, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Kun Liu
- Department of Cardiovascular Diseases, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Wei Jiang
- Department of Cardiovascular Diseases, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Pei-Gang Cao
- Department of Cardiology, General Hospital of Heilongjiang Agricultural Reclamation Bureau, Harbin, PR China.
| |
Collapse
|
31
|
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: 5] [Impact Index Per Article: 1.3] [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
|
32
|
Melatonin prevents doxorubicin-induced cardiotoxicity through suppression of AMPKα2-dependent mitochondrial damage. Exp Mol Med 2020; 52:2055-2068. [PMID: 33339952 PMCID: PMC8080573 DOI: 10.1038/s12276-020-00541-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/09/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
The clinical application of doxorubicin, one of the most effective anticancer drugs, has been limited due to its adverse effects, including cardiotoxicity. One of the hallmarks of doxorubicin-induced cytotoxicity is mitochondrial dysfunction. Despite intensive research over recent decades, there are no effective approaches for alleviating doxorubicin-induced cytotoxicity. Melatonin, a natural hormone that is primarily secreted by the pineal gland, is emerging as a promising adjuvant that protects against doxorubicin-induced cytotoxicity owing to its pharmaceutical effect of preserving mitochondrial integrity. However, the underlying mechanisms are far from completely understood. Here, we provide novel evidence that treatment of H9c2 cardiomyoblasts with doxorubicin strongly induced AMP-activated protein kinase α2 (AMPKα2), which translocated to mitochondria and interfered with their function and integrity, ultimately leading to cellular apoptosis. These phenomena were significantly blocked by melatonin treatment. The levels of AMPKα2 in murine hearts were tightly associated with cardiotoxicity in the context of doxorubicin and melatonin treatment. Therefore, our study suggests that the maintenance of mitochondrial integrity is a key factor in reducing doxorubicin-induced cytotoxicity and indicates that AMPKα2 may serve as a novel target in the design of cytoprotective combination therapies that include doxorubicin. The hormone melatonin reduces heart damage caused by a commonly used chemotherapeutic agent, opening the way towards safer cancer treatment. Doxorubicin is a potent killer of tumor cells, but also has toxic effects on cardiac muscle cells, where it severely damages the mitochondria. Melatonin is best known as a regulator of circadian rhythms, but Joohun Ha and colleagues at Kyung Hee University in Seoul, South Korea, have determined that it can also counteract doxorubicin toxicity. Working with cultured heart cells, the researchers showed that doxorubicin stimulates production of a signaling protein called AMPKα2. This protein subsequently enters the mitochondria and disrupts their structural integrity, leading to cell death. However, adding melatonin to treatment with doxorubicin prevents induction of AMPKα2, thereby increasing heart cell survival.
Collapse
|
33
|
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
|
34
|
Ferlazzo N, Andolina G, Cannata A, Costanzo MG, Rizzo V, Currò M, Ientile R, Caccamo D. Is Melatonin the Cornucopia of the 21st Century? Antioxidants (Basel) 2020; 9:antiox9111088. [PMID: 33167396 PMCID: PMC7694322 DOI: 10.3390/antiox9111088] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
Abstract
Melatonin, an indoleamine hormone produced and secreted at night by pinealocytes and extra-pineal cells, plays an important role in timing circadian rhythms (24-h internal clock) and regulating the sleep/wake cycle in humans. However, in recent years melatonin has gained much attention mainly because of its demonstrated powerful lipophilic antioxidant and free radical scavenging action. Melatonin has been proven to be twice as active as vitamin E, believed to be the most effective lipophilic antioxidant. Melatonin-induced signal transduction through melatonin receptors promotes the expression of antioxidant enzymes as well as inflammation-related genes. Melatonin also exerts an immunomodulatory action through the stimulation of high-affinity receptors expressed in immunocompetent cells. Here, we reviewed the efficacy, safety and side effects of melatonin supplementation in treating oxidative stress- and/or inflammation-related disorders, such as obesity, cardiovascular diseases, immune disorders, infectious diseases, cancer, neurodegenerative diseases, as well as osteoporosis and infertility.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Daniela Caccamo
- Correspondence: ; Tel.: +39-090-221-3386 or +39-090-221-3389
| |
Collapse
|
35
|
Genario R, Cipolla-Neto J, Bueno AA, Santos HO. Melatonin supplementation in the management of obesity and obesity-associated disorders: A review of physiological mechanisms and clinical applications. Pharmacol Res 2020; 163:105254. [PMID: 33080320 DOI: 10.1016/j.phrs.2020.105254] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/11/2020] [Accepted: 10/11/2020] [Indexed: 02/08/2023]
Abstract
Despite the evolving advances in clinical approaches to obesity and its inherent comorbidities, the therapeutic challenge persists. Among several pharmacological tools already investigated, recent studies suggest that melatonin supplementation could be an efficient therapeutic approach in the context of obesity. In the present review, we have amalgamated the evidence so far available on physiological effects of melatonin supplementation in obesity therapies, addressing its effects upon neuroendocrine systems, cardiometabolic biomarkers and body composition. Most studies herein appraised employed melatonin supplementation at dosages ranging from 1 to 20 mg/day, and most studies followed up participants for periods from 3 weeks to 12 months. Overall, it was observed that melatonin plays an important role in glycaemic homeostasis, in addition to modulation of white adipose tissue activity and lipid metabolism, and mitochondrial activity. Additionally, melatonin increases brown adipose tissue volume and activity, and its antioxidant and anti-inflammatory properties have also been demonstrated. There appears to be a role for melatonin in adiposity reduction; however, several questions remain unanswered, for example melatonin baseline levels in obesity, and whether any seeming hypomelatonaemia or melatonin irresponsiveness could be clarifying factors. Supplementation dosage studies and more thorough clinical trials are needed to ascertain not only the relevance of such findings but also the efficacy of melatonin supplementation.
Collapse
Affiliation(s)
- Rafael Genario
- School of Medicine, University of Sao Paulo (USP), São Paulo, Brazil.
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Worcester, United Kingdom
| | - Heitor O Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil.
| |
Collapse
|
36
|
Singhanat K, Apaijai N, Jaiwongkam T, Kerdphoo S, Chattipakorn SC, Chattipakorn N. Melatonin as a therapy in cardiac ischemia-reperfusion injury: Potential mechanisms by which MT2 activation mediates cardioprotection. J Adv Res 2020; 29:33-44. [PMID: 33842003 PMCID: PMC8020169 DOI: 10.1016/j.jare.2020.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/13/2020] [Accepted: 09/22/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction Previous studies reported the beneficial effects of pretreatment with melatonin on the heart during cardiac ischemia/reperfusion (I/R) injury. However, the effects of melatonin given after cardiac ischemia, as well as its comparative temporal effects are unknown. These include pretreatment, during ischemia, and at the onset of reperfusion. Also, the association between melatonin receptors and cardiac arrhythmias, mitochondrial function and dynamics, autophagy, and mitophagy during cardiac I/R have not been investigated. Objectives We tested two major hypotheses in this study. Firstly, the temporal effect of melatonin administration exerts different cardioprotective efficacy during cardiac I/R. Secondly, melatonin provides cardioprotective effects via MT2 activation, leading to improvement in cardiac mitochondrial function and dynamics, reduced excessive mitophagy and autophagy, and decreased cardiac arrhythmias, resulting in improved LV function. Methods Male rats were subjected to cardiac I/R, and divided into 4 intervention groups: vehicle, pretreatment with melatonin, melatonin given during ischemia, and melatonin given at the onset of reperfusion. In addition, either a non-specific melatonin receptor (MT) blocker or specific MT2 blocker was given to rats. Results Treatment with melatonin at all time points alleviated cardiac I/R injury to a similar extent, quantified by reduction in infarct size, arrhythmia score, LV dysfunction, cardiac mitochondrial dysfunction, imbalance of mitochondrial dynamics, excessive mitophagy, and a decreased Bax/Bcl2 ratio. In H9C2 cells, melatonin increased %cell viability by reducing mitochondrial dynamic imbalance and a decrease in Bax protein expression. The cardioprotective effects of melatonin were dependent on MT2 activation. Conclusion Melatonin given before or after ischemia exerted equal levels of cardioprotection on the heart with I/R injury, and its beneficial effects on cardiac arrhythmias, cardiac mitochondrial function and dynamics were dependent upon the activation of MT2.
Collapse
Affiliation(s)
- Kodchanan Singhanat
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thidarat Jaiwongkam
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
37
|
Tanaka K, Okada Y, Maiko H, Mori H, Tanaka Y. Associations between urinary 6-sulfatoxymelatonin excretion and diabetic vascular complications or arteriosclerosis in patients with type 2 diabetes. J Diabetes Investig 2020; 12:601-609. [PMID: 33460308 PMCID: PMC8015816 DOI: 10.1111/jdi.13374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 11/29/2022] Open
Abstract
AIMS/INTRODUCTION There are limited reports on the association between melatonin levels and vascular complications in patients with type 2 diabetes. The aim of this study was to determine the association between urinary 6-sulfatoxymelatonin, which is a urinary metabolite of melatonin, and diabetic vascular complications or arteriosclerosis in patients with type 2 diabetes. MATERIALS AND METHODS This retrospective study included patients (167 patients with type 2 diabetes and 27 patients without diabetes adjusted for age and sex) admitted to the hospital who underwent measurement of urinary 6-sulfatoxymelatonin. The urinary 6-sulfatoxymelatonin/creatinine ratio (6-SMT) was calculated. RESULTS The natural logarithmically scaled 6-SMT level (Ln 6-SMT) was significantly lower in type 2 diabetes patients (1.9 ± 1.1) compared with patients without diabetes (2.8 ± 1.0, P < 0.001). Multivariate linear regression analysis identified duration of diabetes, smoking status, urinary albumin-to-creatinine ratio, retinopathy and coronary heart disease as factors that could influence Ln 6-SMT levels in type 2 diabetes patients (R2 = 0.232, P < 0.001). Ln 6-SMT was associated with decreased odds of diabetic retinopathy, even after adjustment for various confounding factors (odds ratio 0.559, 95% confidence interval 0.369-0.846, P = 0.006). Similarly, Ln 6-SMT was associated with decreased odds of coronary heart disease (odds ratio 0.442, P = 0.030). CONCLUSIONS Our results showed the presence of low levels of Ln 6-SMT in type 2 diabetes patients relative to patients without diabetes. Furthermore, Ln 6-SMT is an independent risk factor of diabetic retinopathy and coronary heart diseases. These findings suggest that 6-SMT could be a useful biomarker for the prediction of micro- and macrovasculopathies in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Kenichi Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yosuke Okada
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hajime Maiko
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hiroko Mori
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
38
|
Chitimus DM, Popescu MR, Voiculescu SE, Panaitescu AM, Pavel B, Zagrean L, Zagrean AM. Melatonin's Impact on Antioxidative and Anti-Inflammatory Reprogramming in Homeostasis and Disease. Biomolecules 2020; 10:biom10091211. [PMID: 32825327 PMCID: PMC7563541 DOI: 10.3390/biom10091211] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/30/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
There is a growing consensus that the antioxidant and anti-inflammatory properties of melatonin are of great importance in preserving the body functions and homeostasis, with great impact in the peripartum period and adult life. Melatonin promotes adaptation through allostasis and stands out as an endogenous, dietary, and therapeutic molecule with important health benefits. The anti-inflammatory and antioxidant effects of melatonin are intertwined and are exerted throughout pregnancy and later during development and aging. Melatonin supplementation during pregnancy can reduce ischemia-induced oxidative damage in the fetal brain, increase offspring survival in inflammatory states, and reduce blood pressure in the adult offspring. In adulthood, disturbances in melatonin production negatively impact the progression of cardiovascular risk factors and promote cardiovascular and neurodegenerative diseases. The most studied cardiovascular effects of melatonin are linked to hypertension and myocardial ischemia/reperfusion injury, while the most promising ones are linked to regaining control of metabolic syndrome components. In addition, there might be an emerging role for melatonin as an adjuvant in treating coronavirus disease 2019 (COVID 19). The present review summarizes and comments on important data regarding the roles exerted by melatonin in homeostasis and oxidative stress and inflammation related pathologies.
Collapse
Affiliation(s)
- Diana Maria Chitimus
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Mihaela Roxana Popescu
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, Elias University Hospital, 010164 Bucharest, Romania;
| | - Suzana Elena Voiculescu
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Anca Maria Panaitescu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, Filantropia Clinical Hospital, 010164 Bucharest, Romania;
| | - Bogdan Pavel
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Leon Zagrean
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
| | - Ana-Maria Zagrean
- Division of Physiology and Neuroscience, Department of Functional Sciences, “Carol Davila” University of Medicine and Pharmacy, 010164 Bucharest, Romania; (D.M.C.); (S.E.V.); (B.P.); (L.Z.)
- Correspondence:
| |
Collapse
|
39
|
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
|
40
|
Infusion of Melatonin Into the Paraventricular Nucleus Ameliorates Myocardial Ischemia-Reperfusion Injury by Regulating Oxidative Stress and Inflammatory Cytokines. J Cardiovasc Pharmacol 2020; 74:336-347. [PMID: 31356536 PMCID: PMC6791501 DOI: 10.1097/fjc.0000000000000711] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Melatonin, the receptors for which are abundant in the hypothalamic paraventricular nucleus (PVN), can protect the heart from myocardial ischemia–reperfusion (MI/R) injury. The aim of this study was to determine whether the infusion of melatonin into the PVN protects the heart from MI/R injury by suppressing oxidative stress or regulating the balance between proinflammatory cytokines and anti-inflammatory cytokines in MI/R rats. Male Sprague–Dawley rats were treated with a bilateral PVN infusion of melatonin. MI/R operation was performed 1 week after infusion. At the end of the third week after the infusion, all the rats were euthanized. This was followed by immunohistochemistry and immunofluorescence studies of the rats. MI/R rats showed larger infarct size, increased left ventricular (LV) end-diastolic volume, and decreased LV ejection fraction and LV fractional shortening. Moreover, MI/R rats had a higher level of norepinephrine in the plasma, heart, and PVN; higher PVN levels of reactive oxygen species, NOX2, NOX4, IL-1β, and NF-κB activity; and lower PVN levels of copper/zinc superoxide dismutase (Cu/Zn-SOD) and IL-10 compared with the sham group. Melatonin infusion in PVN reduced LV end-diastolic volume, norepinephrine, reactive oxygen species, NOX2, NOX4, IL-1β, and NF-κB activity, and increased LV ejection fraction, LV fractional shortening, Cu/Zn-SOD, and IL-10. Overall, these results suggest that the infusion of melatonin ameliorates sympathetic nerve activity and MI/R injury by attenuating oxidative stress and inflammatory cytokines in the PVN of MI/R rats.
Collapse
|
41
|
Melatonin enhances atherosclerotic plaque stability by inducing prolyl-4-hydroxylase α1 expression. J Hypertens 2020; 37:964-971. [PMID: 30335670 DOI: 10.1097/hjh.0000000000001979] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Melatonin, an endogenous neurohormone secreted predominately by the pineal gland, has a variety of physiological functions. However, its protective role in atherosclerosis is not clear. In this study, we sought to investigate the potential effects of melatonin in modulating atherosclerotic plaque stability in apolipoprotein E knockout (ApoE) mice. METHOD AND RESULTS Smooth muscle cells were treated with melatonin, which significantly increased mRNA and protein levels of a key intracellular enzyme essential for collagen maturation and secretion, prolyl-4-hydroxylase α1 (P4Hα1). Mechanistically, melatonin increased Akt phosphorylation and transcriptional activation of specificity protein 1 (Sp1), which bound with the P4Hα1 promoter and then induced P4Hα1 expression. Pretreatment with either Akt inhibitor LY294002 or Sp1 inhibitor mithramycin A (MTM) could inhibit melatonin-induced P4Hα1 expression. Finally, atherosclerotic lesions were induced by placing a perivascular collar on the right common carotid artery of ApoE mice, which were received with or without different doses of melatonin or MTM. High-dose melatonin enhanced atherosclerotic plaque stability in ApoE mice in vivo by inducing the expression of P4Hα1, which was reversed by MTM. CONCLUSION We propose that melatonin supplementation may provide a novel and promising approach to atherosclerosis treatment.
Collapse
|
42
|
Song YJ, Zhong CB, Wu W. Cardioprotective effects of melatonin: Focusing on its roles against diabetic cardiomyopathy. Biomed Pharmacother 2020; 128:110260. [PMID: 32447213 DOI: 10.1016/j.biopha.2020.110260] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/01/2020] [Accepted: 05/10/2020] [Indexed: 02/06/2023] Open
Abstract
Melatonin is a pineal-produced indole known for its anti-aging, antiapoptotic and antioxidant properties. In past decades, the protective potentials of melatonin for cardiovascular diseases, such as atherosclerosis and myocardial infarction, have been widely revealed, triggering more investigations focused on other cardioprotective effects of melatonin. Recently, the roles of melatonin in diabetic cardiomyopathy (DCM) have attracted increased attention. In this regard, researchers found that melatonin attenuated cardiac fibrosis and hypertrophy, thus interrupting the development of DCM. Retinoid-related orphan receptor α is a key melatonin receptor that contributed to the cardioprotective effect of melatonin in hearts with DCM. For the downstream mechanisms, the inhibition of mammalian STE20-like kinase 1 plays a pivotal role, which exerts antiapoptotic and proautophagic effects, thus enhancing cardiac tolerance in high-glucose conditions. In addition, other signalling mechanisms, such as sirtuin-1/peroxisome proliferator-activated receptor gamma-coactivator alpha and endoplasmic reticulum-related signalling, are also involved in the protective effects of melatonin on cardiomyocytes under diabetic conditions. This review will focus on the protective signalling mechanisms regulated by melatonin and provide a better understanding of the therapeutic applications of melatonin signalling in DCM.
Collapse
Affiliation(s)
- Yan-Jun Song
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, 1 Shuai Fu Yuan, Beijing, 100730, PR China.
| | - Chong-Bin Zhong
- Department of Cardiology, Heart Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, PR China.
| | - Wei Wu
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, 1 Shuai Fu Yuan, Beijing, 100730, PR China.
| |
Collapse
|
43
|
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: 43] [Impact Index Per Article: 8.6] [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
|
44
|
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: 98] [Impact Index Per Article: 19.6] [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
|
45
|
Zhi W, Li K, Wang H, Lei M, Guo Y. Melatonin elicits protective effects on OGD/R‑insulted H9c2 cells by activating PGC‑1α/Nrf2 signaling. Int J Mol Med 2020; 45:1294-1304. [PMID: 32323734 PMCID: PMC7138270 DOI: 10.3892/ijmm.2020.4514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
Melatonin (Mel) elicits beneficial effects on myocardial ischemia/reperfusion injury. However, the underlying mechanism of Mel against oxygen-glucose deprivation/ reperfusion (OGD/R)-induced H9c2 cardiomyocyte damage remains largely unknown. The aim of the present study was to investigate the biological roles and the potential mechanisms of Mel in OGD/R-exposed H9c2 cardiomyocytes. The results of the present study demonstrated that Mel significantly elevated the viability and reduced the activity of lactate dehydrogenase and creatine kinase myocardial band in a doseand time-dependent manner in OGD/R-insulted H9c2 cells. In addition, Mel suppressed OGD/R-induced oxidative stress in H9c2 cells, as demonstrated by the decreased reactive oxygen species and malondialdehyde levels, as well as the increased activities of superoxide dismutase, catalase and glutathione peroxidase. Mel exerted an antioxidant effect by activating the peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Mel reduced the expression of OGD/R-enhanced pro-inflammatory tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-1β, IL-8 and monocyte chemotactic protein-1. Mel also abolished the OGD/R-induced increase in H9c2 apoptosis, as evidenced by mitochondrial membrane potential restoration and caspase-3 and caspase-9 inactivation, as well as the upregulation of Bcl-2 and down-regulation of cleaved caspase-3 and Bax. The Mel-induced antiapoptotic effects were dependent on PGC-1α/TNF-α signaling. Overall, the results of the present study demonstrated that Mel alleviated OGD/R-induced H9c2 cell injury via the inhibition of oxidative stress and inflammation by regulating the PGC-1α/Nrf2 and PGC-1α/TNF-α signaling pathways, suggesting a promising role for Mel in the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Weiwei Zhi
- Department of Cardiology, Xi'an No. 3 Hospital, Xi'an, Shaanxi 710018, P.R. China
| | - Kai Li
- Department of Cardiology, Xi'an No. 3 Hospital, Xi'an, Shaanxi 710018, P.R. China
| | - Hongbing Wang
- Department of Cardiology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Ming Lei
- Department of Cardiology, Xi'an No. 3 Hospital, Xi'an, Shaanxi 710018, P.R. China
| | - Yingqiang Guo
- Department of Cardiology, Xi'an No. 3 Hospital, Xi'an, Shaanxi 710018, P.R. China
| |
Collapse
|
46
|
Li HR, Wang C, Sun P, Liu DD, Du GQ, Tian JW. Melatonin attenuates doxorubicin-induced cardiotoxicity through preservation of YAP expression. J Cell Mol Med 2020; 24:3634-3646. [PMID: 32068341 PMCID: PMC7131936 DOI: 10.1111/jcmm.15057] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/10/2020] [Accepted: 01/22/2020] [Indexed: 12/21/2022] Open
Abstract
There are increasing concerns related to the cardiotoxicity of doxorubicin in the clinical setting. Recently, melatonin has been shown to exert a cardioprotective effect in various cardiovascular diseases, including cardiotoxic conditions. In this study, we examined the possible protective effects of melatonin on doxorubicin‐induced cardiotoxicity and explored the underlying mechanisms related to this process. We found that in vitro doxorubicin treatment significantly decreased H9c2 cell viability and induced apoptosis as manifested by increased TUNEL‐positive cells, down‐regulation of anti‐apoptotic protein Bcl‐2, as well as up‐regulation of pro‐apoptotic protein Bax. This was associated with increased reactive oxygen species (ROS) levels and decreased mitochondrial membrane potentials (MMP). In vivo, five weeks of doxorubicin treatment significantly decreased cardiac function, as evaluated by echocardiography. TUNEL staining results confirmed the increased apoptosis caused by doxorubicin. On the other hand, combinational treatment of doxorubicin with melatonin decreased cardiomyocyte ROS and apoptosis levels, along with increasing MMP. Such doxorubicin‐melatonin co‐treatment alleviated in vivo doxorubicin‐induced cardiac injury. Western Blots, along with in vitro immunofluorescence and in vivo immunohistochemical staining confirmed that doxorubicin treatment significantly down‐regulated Yes‐associated protein (YAP) expression, while YAP levels were maintained under co‐treatment of doxorubicin and melatonin. YAP inhibition by siRNA abolished the protective effects of melatonin on doxorubicin‐treated cardiomyocytes, with reversed ROS level and apoptosis. Our findings suggested that melatonin treatment attenuated doxorubicin‐induced cardiotoxicity through preserving YAP levels, which in turn decreases oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Hai-Ru Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Chao Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Ping Sun
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Dan-Dan Liu
- Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guo-Qing Du
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Jia-Wei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
47
|
Di S, Wang Z, Hu W, Yan X, Ma Z, Li X, Li W, Gao J. The Protective Effects of Melatonin Against LPS-Induced Septic Myocardial Injury: A Potential Role of AMPK-Mediated Autophagy. Front Endocrinol (Lausanne) 2020; 11:162. [PMID: 32373063 PMCID: PMC7176935 DOI: 10.3389/fendo.2020.00162] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
Aim: Melatonin is an indolamine secreted by the pineal gland, as well as most of the organs and tissues. In addition to regulating circadian biology, studies have confirmed the multiple pharmacological effects of melatonin. Melatonin provides a strong defense against septic myocardial injury. However, the underlying mechanism has not been fully described. In this study, we investigated the protective effects of melatonin against lipopolysaccharide (LPS)-induced myocardial injury as well as the mechanisms involved. Methods: Mice were intraperitoneally injected with LPS to induce a septic myocardial injury model or an LPS shock model, depending on the dose of LPS. Melatonin was given (20 mg/kg/day, via intraperitoneal injection) for a week prior to LPS insult. 6 h after LPS injection, echocardiographic analysis, TUNEL staining, transmission electron microscopy (TEM), western blot, quantitative real-time PCR and ELISA were used to investigate the protective effects of melatonin against LPS induced myocardial injury. AMPK inhibitor, autophagy activator and inhibitor, siRNAs were used for further validation. Results: Survival test showed that melatonin significantly increased the survival rate after LPS-induced shock. In the sepsis model, melatonin markedly ameliorated myocardial dysfunction, decreased the release of inflammatory cytokines, activated AMP-activated protein kinase (AMPK), improved mitochondrial function, and activated autophagy. To confirm whether the protection of melatonin was mediated by AMPK and autophagy, Compound C, an AMPK inhibitor; 3-MA, an autophagy inhibitor; and Rapamycin (Rapa), an autophagy activator, were used in this study. AMPK inhibition down-regulated autophagy, abolished protection of melatonin, as indicated by significantly decreased cardiac function, increased inflammation and damaged mitochondrial function. Furthermore, autophagy inhibition by 3-MA significantly impaired the protective effects of melatonin, whereas autophagy activation by Rapa reversed LPS + Compound C induced myocardial injury. In addition, in vitro studies further confirmed the protection of melatonin against LPS-induced myocardial injury and the mechanisms involving AMPK-mediated autophagy signaling. Conclusions: In summary, our results demonstrated that melatonin protects against LPS-induced septic myocardial injury by activating AMPK mediated autophagy pathway.
Collapse
Affiliation(s)
- Shouyin Di
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- Department of Thoracic Surgery, Sixth Medical Center of PLA General Hospital, Beijing, China
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Wei Hu
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Weimiao Li
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- *Correspondence: Weimiao Li
| | - Jianyuan Gao
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- Jianyuan Gao
| |
Collapse
|
48
|
Dube K, Dhanabalan K, Salie R, Blignaut M, Huisamen B, Lochner A. Melatonin has profound effects on mitochondrial dynamics in myocardial ischaemia/reperfusion. Heliyon 2019; 5:e02659. [PMID: 31720456 PMCID: PMC6838907 DOI: 10.1016/j.heliyon.2019.e02659] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/11/2019] [Accepted: 10/11/2019] [Indexed: 02/07/2023] Open
Abstract
Research focus recently shifted to mitochondrial dynamics and the role of fusion and fission in cardioprotection. The aim of this study was to evaluate (i) the function and dynamics of mitochondria isolated from hearts exposed to ischaemia/reperfusion (I/R) (ii) the effects of melatonin, a powerful cardioprotectant, on mitochondrial dynamics in I/R. Isolated perfused rat hearts were stabilized for 30 min, subjected to 20 min global ischaemia, followed by 30 min reperfusion. Tissue was collected, mitochondria isolated for measurement of mitochondrial oxidative function and lysates from mitochondrial and cytosolic fractions prepared for western blotting. Melatonin (0.3 or 50 μM) was administered for 10 min immediately before the onset of ischaemia and for 10 min at the onset of reperfusion. Infarct size was assessed after 35 min regional ischaemia/60 min reperfusion using triphenyltetrazolium staining. The results show that reperfusion significantly reduced mitochondrial QO2 (states 3 and 4), with minor effects by melatonin. Cytosolic Beclin 1 and the LC3 II/I ratio were reduced by ischaemia and increased by reperfusion. Both ischaemia and reperfusion reduced mitochondrial PINK1 and Parkin levels, while reperfusion increased p62. An alternative mitophagy pathway mediated by Rab9 is activated during myocardial ischaemia/reperfusion. Ischaemia reduced and reperfusion increased cytosolic ULK1 expression, associated with redistribution of Rab9 and Drp1 between the cytosol and mitochondria. Melatonin significantly reduced mitochondrial p62 expression upon reperfusion. Throughout the protocol, melatonin significantly (i) increased cytosolic total (t) and phospho (p) ULK1, and Rab9 levels (ii) increased the cytosolic and reduced the mitochondrial pDrp1 levels and p/t Drp1 ratio, suggesting inhibition of mitochondrial fission. Fusion was affected to a lesser extent. Cardioprotection by melatonin is associated with substantial effects on mitophagy, the significance thereof remains to be established.
Collapse
|
49
|
Prado NJ, Egan Beňová T, Diez ER, Knezl V, Lipták B, Ponce Zumino AZ, Llamedo-Soria M, Szeiffová Bačová B, Miatello RM, Tribulová N. Melatonin receptor activation protects against low potassium-induced ventricular fibrillation by preserving action potentials and connexin-43 topology in isolated rat hearts. J Pineal Res 2019; 67:e12605. [PMID: 31408542 DOI: 10.1111/jpi.12605] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/27/2019] [Accepted: 08/05/2019] [Indexed: 12/22/2022]
Abstract
Hypokalemia prolongs the QRS and QT intervals, deteriorates intercellular coupling, and increases the risk for arrhythmia. Melatonin preserves gap junctions and shortens action potential as potential antiarrhythmic mechanisms, but its properties under hypokalemia remain unknown. We hypothesized that melatonin protects against low potassium-induced arrhythmias through the activation of its receptors, resulting in action potential shortening and connexin-43 preservation. After stabilization in Krebs-Henseleit solution (4.5 mEq/L K+ ), isolated hearts from Wistar rats underwent perfusion with low-potassium (1 mEq/L) solution and melatonin (100 μmol/L), a melatonin receptor blocker (luzindole, 5 μmol/L), melatonin + luzindole or vehicle. The primary endpoint of the study was the prevention of ventricular fibrillation. Electrocardiography was used, and epicardial action potentials and heart function were measured and analyzed. The ventricular expression, dephosphorylation, and distribution of connexin-43 were examined. Melatonin reduced the incidence of low potassium-induced ventricular fibrillation from 100% to 59%, delayed the occurrence of ventricular fibrillation and induced a faster recovery of sinus rhythm during potassium restitution. Melatonin prevented QRS widening, action potential activation delay, and the prolongation of action potential duration at 50% of repolarization. Other ECG and action potential parameters, the left ventricular developed pressure, and nonsustained ventricular arrhythmias did not differ among groups. Melatonin prevented connexin-43 dephosphorylation and its abnormal topology (lateralization). Luzindole abrogated the protective effects of melatonin on electrophysiological properties and connexin-43 misdistribution. Our results indicate that melatonin receptor activation protects against low potassium-induced ventricular fibrillation, shortens action potential duration, preserves ventricular electrical activation, and prevents acute changes in connexin-43 distribution. All of these properties make melatonin a remarkable antifibrillatory agent.
Collapse
Affiliation(s)
- Natalia Jorgelina Prado
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
| | - Tamara Egan Beňová
- Center of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovakia
| | - Emiliano Raúl Diez
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
- Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Vladimír Knezl
- Center of Experimental Medicine, Slovak Academy of Sciences, Institute of Experimental Pharmacology and Toxicology, Bratislava, Slovakia
| | - Boris Lipták
- Center of Experimental Medicine, Slovak Academy of Sciences, Institute of Experimental Pharmacology and Toxicology, Bratislava, Slovakia
| | - Amira Zulma Ponce Zumino
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
- Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Mariano Llamedo-Soria
- Department of Electronic Engineering, Universidad Tecnológica Nacional, Buenos Aires, Argentina
| | - Barbara Szeiffová Bačová
- Center of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovakia
| | - Roberto Miguel Miatello
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
- Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Narcisa Tribulová
- Center of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovakia
| |
Collapse
|
50
|
Zahid JA, Isbrand A, Kleif J, Schou-Pedersen AMV, Lykkesfeldt J, Madsen MT, Gögenur I. The effect of melatonin on endothelial dysfunction in patients after acute coronary syndrome: The MEFACS randomized clinical trial. J Pineal Res 2019; 67:e12600. [PMID: 31355944 DOI: 10.1111/jpi.12600] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/13/2019] [Accepted: 07/21/2019] [Indexed: 12/23/2022]
Abstract
Endothelial dysfunction (ED) precedes acute coronary syndrome. Oxidative stress results in ED but is reversible. Melatonin is aside from being a circadian hormone, also an antioxidant. The aim of this study was to investigate whether 25 mg melatonin administered for twelve weeks following acute coronary syndrome (ACS) could improve ED. In this placebo-controlled randomized trial, ED was measured as reactive hyperemia index (RHI) at baseline, day 14, and day 84. The effect was assessed using a generalized estimating equation adjusted for the baseline RHI. As secondary outcome, the concentrations of three biomarkers were measured: l-arginine, asymmetric dimethylarginine, and uric acid. Thirty-one patients were included in the study. The intention-to-treat analysis of the primary outcome had 26 patients due to missing data. The estimated marginal mean difference in RHI at day 14 and day 84 between the groups was 0.15 (95% CI: 0.29-0.01, P = .039) in favor of the placebo group. No significant differences in the biomarker concentrations were found. Melatonin treatment after ACS did not improve but may have aggravated ED. The significant difference between groups was in favor of placebo, but this might be due to the effect of missing data or uneven distribution of comorbidities.
Collapse
Affiliation(s)
- Jawad Ahmad Zahid
- Department of Surgery, Centre for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Anders Isbrand
- Department of Clinical Physiology and Nuclear Medicine, Herlev Hospital, Herlev, Denmark
| | - Jakob Kleif
- Department of Surgery, Nordsjaellands Hospital, Hillerød, Denmark
| | | | - Jens Lykkesfeldt
- Faculty of Health & Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Tvilling Madsen
- Department of Surgery, Centre for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Ismail Gögenur
- Department of Surgery, Centre for Surgical Science, Zealand University Hospital, Køge, Denmark
| |
Collapse
|