1
|
Sheng Z, Zhang R, Ji Z, Liu Z, Zhou Y. Identification of mitophagy-related key genes and their correlation with immune cell infiltration in acute myocardial infarction via bioinformatics analysis. Front Cardiovasc Med 2025; 11:1501608. [PMID: 39872885 PMCID: PMC11770045 DOI: 10.3389/fcvm.2024.1501608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/06/2024] [Indexed: 01/30/2025] Open
Abstract
Background Acute myocardial infarction (AMI), a subset of acute coronary syndrome, remains the major cause of mortality worldwide. Mitochondrial dysfunction is critically involved in AMI progression, and mitophagy plays a vital role in eliminating damaged mitochondria. This study aimed to explore mitophagy-related biomarkers and their potential molecular basis in AMI. Methods AMI datasets (GSE24519 and GSE34198) from the Gene Expression Omnibus database were combined and the batch effects were removed. Differentially expressed genes (DEGs) in AMI were selected, intersected with mitophagy-related genes for mitophagy-related DEGs (MRDEGs), and then subjected to enrichment analyses. Next, the MRDEGs were screened using machine learning methods (logistic regression analysis, RandomForest, least absolute shrinkage and selection operator) to construct a diagnostic risk model and select the key genes in AMI. The diagnostic efficacy of the model was evaluated using a nomogram. Moreover, the infiltration patterns of different immune cells in two risk groups were compared. We also explored the interactions between the key genes themselves or with miRNAs/transcription factors (TFs) and drug compounds and visualized the protein structure of the key genes. Finally, we explored and validated the expression of key genes in plasma samples of patients with an AMI and healthy individuals. Results We screened 28 MRDEGs in AMI. Based on machine learning methods, 12 key genes were screened for the diagnostic risk model, including AGPS, CA2, CAT, LTA4H, MYO9B, PRDX6, PYGB, SIRT3, TFEB, TOM1, UBA52, and UBB. The nomogram further revealed the accuracy of the model for AMI diagnosis. Moreover, we found a lower abundance of immune cells such as gamma delta T and natural killer cells in the high-risk group, and the expression of key genes showed a significant correlation with immune infiltration levels in both groups. Finally, 64 miRNA-mRNA pairs, 75 TF-mRNA pairs, 119 RNA-binding protein-mRNA pairs, and 32 drug-mRNA pairs were obtained in the interaction networks. Conclusions In total, 12 key MRDEGs were identified and a risk model was constructed for AMI diagnosis. The findings of this study might provide novel biomarkers for improving the detection of AMI.
Collapse
Affiliation(s)
- Zulong Sheng
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | | | | | | | | |
Collapse
|
2
|
Ju F, Zhang X, Zhao Z, Cao Y, Xie A, Xia L, Zhou D. Aloperine Regulates Inflammation, Apoptosis, and Autophagy in H9C2 Rat Cardiomyoblast Cells After Excessive Hypoxia. Int Heart J 2025; 66:157-163. [PMID: 39894544 DOI: 10.1536/ihj.24-454] [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] [Indexed: 02/04/2025]
Abstract
Myocardial infarction (MI) is a cardiovascular condition that leads to increased morbidity and mortality, impacting the quality of life of individuals. Aloperine (ALO), derived from Sophora alopecuroides L, has been recognized for its beneficial effects in treating various diseases by showcasing therapeutic properties. However, the precise protective mechanisms of ALO on hypoxia/reoxygenation (H/R) -induced damage in cardiomyocytes in vitro remain unclear. In this study, it was manifested that cell proliferation was weakened after H/R treatment, but this impact was offset after ALO treatment. Furthermore, cell apoptosis was heightened after H/R treatment, but this phenomenon was neutralized after ALO treatment. ALO relieved inflammation in H/R-treated H9C2 rat cardiomyoblast cells. Moreover, ALO strengthened autophagy in H/R-triggered H9C2 rat cardiomyoblast cells through enhancing the LC3II/LC3I level and the LC3B fluorescence intensity. Lastly, it was testified that ALO can rescue the weakened autophagy, the heightened cell apoptosis, and the augmented inflammation after CC treatment in H/R-mediated H9C2 rat cardiomyoblast cells. In conclusion, ALO regulated inflammation, apoptosis, and autophagy through AMPK/Nrf2 pathway in H9C2 rat cardiomyoblast cells after excessive hypoxia. This study suggested that ALO may be an underlying drug for MI therapy.
Collapse
Affiliation(s)
- Feng Ju
- Department of Anesthesiology, People's Hospital of Deyang City
| | - Xianjie Zhang
- Department of Anesthesiology, People's Hospital of Deyang City
| | - Zhifu Zhao
- Department of Anesthesiology, People's Hospital of Deyang City
| | - Yuansheng Cao
- Department of Anesthesiology, People's Hospital of Deyang City
| | - An Xie
- Department of Anesthesiology, People's Hospital of Deyang City
| | - Leqiang Xia
- Department of Anesthesiology, People's Hospital of Deyang City
| | - Dan Zhou
- Department of Anesthesiology, People's Hospital of Deyang City
| |
Collapse
|
3
|
Donia T, Ali EMM, Kalantan AA, Alzahrani FA, Eid TM, Khamis AA. Synergistic anticancer efficacy of polydatin and sorafenib against the MCF-7 breast cancer cell line via inhibiting of PI3K/AKT/mTOR pathway and reducing resistance to treatment. Biochem Biophys Res Commun 2024; 739:150972. [PMID: 39541924 DOI: 10.1016/j.bbrc.2024.150972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/25/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Polydatin (PD), a glucoside derivative of resveratrol, has been investigated for its potential to mitigate sorafenib (SOF) side effects and combat multidrug resistance in cancer treatment. The study evaluated its mechanism of action for inhibiting the protein kinase B/mTOR pathway in promoting breast cancer proliferation. The combined PD and SOF have synergistic effects with a combination index (CI) < 1 in the liver (HepG2) and breast (MCF-7) cancer cell lines. Molecular docking studies were conducted to analyze interactions of PD& SOF with protein kinases as well as apoptotic and multidrug resistance proteins, including AKT1, PI3K, mTOR, Apaf-1, and ABCB1 in MCF-7 cells. Experimental validation through real-time PCR confirmed. PD has a strong binding affinity, particularly with AKT1 (-56 kcal/mol) and ABCB1 (-27.16 kcal/mol), a gene associated with multidrug resistance. These interactions were linked to anti-proliferative anti-angiogenic effects and reduced resistance to treatment, demonstrating PD has potential therapeutic benefits. Furthermore, PD combined with SOF induced apoptosis, inhibited cell growth, and arrested MCF-7 cells in the sub-G1 phase with increased intracellular ROS. This was accompanied by reduced expression of AKT1 and ABCB1 genes, reinforcing the anticancer efficacy of PD/SOF combination therapy. In conclusion, the findings suggest that PD/SOF could serve as a promising anticancer treatment strategy, warranting further investigation for potential clinical applications and mechanistic studies in vivo.
Collapse
Affiliation(s)
- Thoria Donia
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt.
| | - Ehab M M Ali
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt; Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.
| | - Abdulaziz A Kalantan
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.
| | - Faisal Ay Alzahrani
- Department of Chemistry, College of Sciences & Arts, King Abdulaziz University, 21911 Rabigh, Saudi Arabia.
| | - Thamir M Eid
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.
| | - Abeer A Khamis
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, Egypt.
| |
Collapse
|
4
|
Emili M, Stagni F, Russo C, Angelozzi L, Guidi S, Bartesaghi R. Reversal of neurodevelopmental impairment and cognitive enhancement by pharmacological intervention with the polyphenol polydatin in a Down syndrome model. Neuropharmacology 2024; 261:110170. [PMID: 39341334 DOI: 10.1016/j.neuropharm.2024.110170] [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: 04/23/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Intellectual disability (ID) is the unavoidable hallmark of Down syndrome (DS), a genetic condition due to triplication of chromosome 21. ID in DS is largely attributable to neurogenesis and dendritogenesis alterations taking place in the prenatal/neonatal period, the most critical time window for brain development. There are currently no treatments for ID in DS. Considering the timeline of brain development, treatment aimed at improving the neurological phenotypes of DS should be initiated as early as possible and use safe agents. The goal of this study was to establish whether it is possible to improve DS-linked neurodevelopmental defects through early treatment with polydatin, a natural polyphenol. We used the Ts65Dn mouse model of DS and focused on the hippocampus, a brain region fundamental for long-term memory. We found that in Ts65Dn mice of both sexes treated with polydatin from postnatal (P) day 3 to P15 there was full restoration of neurogenesis, neuron number, and dendritic development. These effects were accompanied by normalization of Cyclin D1 and DSCAM levels, which may account for the rescue of neurogenesis and dendritogenesis, respectively. Importantly, in Ts65Dn mice treated with polydatin from P3 to adolescence (∼P50) there was full restoration of hippocampus-dependent memory, indicating a pro-cognitive outcome of treatment. No adverse effects were observed on the body and brain weight. The efficacy and safety of polydatin in a model of DS prospect the possibility of its use during early life stages for amelioration of DS-linked neurodevelopmental alterations.
Collapse
Affiliation(s)
- Marco Emili
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Carla Russo
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Laura Angelozzi
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
5
|
Reyna-Bolaños I, Solís-García EP, Vargas-Vargas MA, Peña-Montes DJ, Saavedra-Molina A, Cortés-Rojo C, Calderón-Cortés E. Polydatin Prevents Electron Transport Chain Dysfunction and ROS Overproduction Paralleled by an Improvement in Lipid Peroxidation and Cardiolipin Levels in Iron-Overloaded Rat Liver Mitochondria. Int J Mol Sci 2024; 25:11104. [PMID: 39456885 PMCID: PMC11508176 DOI: 10.3390/ijms252011104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/28/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Increased intramitochondrial free iron is a key feature of various liver diseases, leading to oxidative stress, mitochondrial dysfunction, and liver damage. Polydatin is a polyphenol with a hepatoprotective effect, which has been attributed to its ability to enhance mitochondrial oxidative metabolism and antioxidant defenses, thereby inhibiting reactive oxygen species (ROS) dependent cellular damage processes and liver diseases. However, it has not been explored whether polydatin is able to exert its effects by protecting the phospholipid cardiolipin against damage from excess iron. Cardiolipin maintains the integrity and function of electron transport chain (ETC) complexes and keeps cytochrome c bound to mitochondria, avoiding uncontrolled apoptosis. Therefore, the effect of polydatin on oxidative lipid damage, ETC activity, cytochrome levels, and ROS production was explored in iron-exposed rat liver mitochondria. Fe2+ increased lipid peroxidation, decreased cardiolipin and cytochromes c + c1 and aa3 levels, inhibited ETC complex activities, and dramatically increased ROS production. Preincubation with polydatin prevented all these effects to a variable degree. These results suggest that the hepatoprotective mechanism of polydatin involves the attenuation of free radical production by iron, which enhances cardiolipin levels by counteracting membrane lipid peroxidation. This prevents the loss of cytochromes, improves ETC function, and decreases mitochondrial ROS production.
Collapse
Affiliation(s)
- Itzel Reyna-Bolaños
- Instituto Tecnológico Superior de Ciudad Hidalgo, Tecnológico Nacional de México, Ciudad Hidalgo 61100, Michoacán, Mexico; (I.R.-B.); (E.P.S.-G.)
| | - Elsa Paola Solís-García
- Instituto Tecnológico Superior de Ciudad Hidalgo, Tecnológico Nacional de México, Ciudad Hidalgo 61100, Michoacán, Mexico; (I.R.-B.); (E.P.S.-G.)
| | - Manuel Alejando Vargas-Vargas
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (M.A.V.-V.); (D.J.P.-M.); (A.S.-M.)
| | - Donovan J. Peña-Montes
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (M.A.V.-V.); (D.J.P.-M.); (A.S.-M.)
| | - Alfredo Saavedra-Molina
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (M.A.V.-V.); (D.J.P.-M.); (A.S.-M.)
| | - Christian Cortés-Rojo
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, Michoacán, Mexico; (M.A.V.-V.); (D.J.P.-M.); (A.S.-M.)
| | - Elizabeth Calderón-Cortés
- Facultad de Enfermería, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58260, Michoacán, Mexico
| |
Collapse
|
6
|
Sai Priya T, Ramalingam V, Suresh Babu K. Natural products: A potential immunomodulators against inflammatory-related diseases. Inflammopharmacology 2024:10.1007/s10787-024-01562-4. [PMID: 39196458 DOI: 10.1007/s10787-024-01562-4] [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: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
The incidence and prevalence of inflammatory-related diseases (IRDs) are increasing worldwide. Current approved treatments for IRDs in the clinic are combat against inhibiting the pro-inflammatory cytokines. Though significant development in the treatment in the IRDs has been achieved, the severe side effects and inefficiency of currently practicing treatments are endless challenge. Drug discovery from natural sources is efficacious over a resurgence and also natural products are leading than the synthetic molecules in both clinical trials and market. The use of natural products against IRDs is a conventional therapeutic approach since it is a reservoir of unique structural chemistry, accessibility and bioactivities with reduced side effects and low toxicity. In this review, we discuss the cause of IRDs, treatment of options for IRDs and the impact and adverse effects of currently practicing clinical drugs. As well, the significant role of natural products against various IRDs, the limitations in the clinical development of natural products and thus pave the way for development of natural products as immunomodulators against IRDs are also discussed.
Collapse
Affiliation(s)
- Telukuntla Sai Priya
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vaikundamoorthy Ramalingam
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Katragadda Suresh Babu
- Department of Natural Products & Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
7
|
Wang J, Hashimoto Y, Hiemori-Kondo M, Nakamoto A, Sakai T, Ye W, Abe-Kanoh N. Resveratrol and piceid enhance efferocytosis by increasing the secretion of MFG-E8 in human THP-1 macrophages. Biosci Biotechnol Biochem 2024; 88:1090-1101. [PMID: 38830798 DOI: 10.1093/bbb/zbae079] [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: 03/19/2024] [Accepted: 05/26/2024] [Indexed: 06/05/2024]
Abstract
The process of apoptotic cell clearance by phagocytes, known as efferocytosis, plays an essential role in maintaining homeostasis. Defects in efferocytosis can lead to inflammatory diseases such as atherosclerosis and autoimmune disorders. Therefore, the maintenance and promotion of efferocytosis are considered crucial for preventing these diseases. In this study, we observed that resveratrol, a representative functional food ingredient, and its glycoside, piceid, promoted efferocytosis in both human THP-1 macrophages differentiated with phorbol 12-myristate 13-acetate and peritoneal macrophages from thioglycolate-elicited mice. Resveratrol and piceid significantly increased mRNA expression and protein secretion of MFG-E8 in THP-1 macrophages. Furthermore, the activation of efferocytosis and the increment in MFG-E8 protein secretion caused by resveratrol or piceid treatment were canceled by MFG-E8 knockdown in THP-1 macrophages. In conclusion, we have demonstrated for the first time that resveratrol and piceid promote efferocytosis through the upregulation of MFG-E8 excretion in human THP-1 macrophages.
Collapse
Affiliation(s)
- Jing Wang
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang Key Laboratory of Grapevine Improvement and Utilization, Weifang, Shandong, China
| | - Yuki Hashimoto
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Miki Hiemori-Kondo
- Department of Nutrition, Faculty of Nutrition, University of Kochi, Kochi, Japan
| | - Akiko Nakamoto
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Tohru Sakai
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Wenxiu Ye
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang Key Laboratory of Grapevine Improvement and Utilization, Weifang, Shandong, China
| | - Naomi Abe-Kanoh
- Department of Food, Life and Environmental Science, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata, Japan
| |
Collapse
|
8
|
Trinh D, Al Halabi L, Brar H, Kametani M, Nash JE. The role of SIRT3 in homeostasis and cellular health. Front Cell Neurosci 2024; 18:1434459. [PMID: 39157755 PMCID: PMC11327144 DOI: 10.3389/fncel.2024.1434459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 06/25/2024] [Indexed: 08/20/2024] Open
Abstract
Mitochondria are responsible for maintaining cellular energy levels, and play a major role in regulating homeostasis, which ensures physiological function from the molecular to whole animal. Sirtuin 3 (SIRT3) is the major protein deacetylase of mitochondria. SIRT3 serves as a nutrient sensor; under conditions of mild metabolic stress, SIRT3 activity is increased. Within the mitochondria, SIRT3 regulates every complex of the electron transport chain, the tricarboxylic acid (TCA) and urea cycles, as well as the mitochondria membrane potential, and other free radical scavengers. This article reviews the role of SIRT3 in regulating homeostasis, and thus physiological function. We discuss the role of SIRT3 in regulating reactive oxygen species (ROS), ATP, immunological function and mitochondria dynamics.
Collapse
Affiliation(s)
- Dennison Trinh
- Department of Biological Sciences, University of Toronto, Toronto, ON, Canada
| | - Lina Al Halabi
- Department of Biological Sciences, University of Toronto, Toronto, ON, Canada
| | - Harsimar Brar
- Department of Biological Sciences, University of Toronto, Toronto, ON, Canada
| | - Marie Kametani
- Department of Biological Sciences, University of Toronto, Toronto, ON, Canada
| | - Joanne E. Nash
- Department of Biological Sciences, University of Toronto Scarborough Graduate Department of Cells Systems Biology, University of Toronto Cross-Appointment with Department of Psychology, University of Toronto Scarborough Scientist – KITE, Toronto, ON, Canada
| |
Collapse
|
9
|
Cai K, Jiang H, Zou Y, Song C, Cao K, Chen S, Wu Y, Zhang Z, Geng D, Zhang N, Liu B, Sun G, Tang M, Li Z, Zhang Y, Sun Y, Zhang Y. Programmed death of cardiomyocytes in cardiovascular disease and new therapeutic approaches. Pharmacol Res 2024; 206:107281. [PMID: 38942341 DOI: 10.1016/j.phrs.2024.107281] [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: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Cardiovascular diseases (CVDs) have a complex pathogenesis and pose a major threat to human health. Cardiomyocytes have a low regenerative capacity, and their death is a key factor in the morbidity and mortality of many CVDs. Cardiomyocyte death can be regulated by specific signaling pathways known as programmed cell death (PCD), including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, etc. Abnormalities in PCD can lead to the development of a variety of cardiovascular diseases, and there are also molecular-level interconnections between different PCD pathways under the same cardiovascular disease model. Currently, the link between programmed cell death in cardiomyocytes and cardiovascular disease is not fully understood. This review describes the molecular mechanisms of programmed death and the impact of cardiomyocyte death on cardiovascular disease development. Emphasis is placed on a summary of drugs and potential therapeutic approaches that can be used to treat cardiovascular disease by targeting and blocking programmed cell death in cardiomyocytes.
Collapse
Affiliation(s)
- Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Haoyue Jiang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China
| | - Bo Liu
- The first hospital of China Medical University, Department of cardiac surgery, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Man Tang
- Department of clinical pharmacology, College of Pharmacy, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| |
Collapse
|
10
|
Cilleros-Holgado P, Gómez-Fernández D, Piñero-Pérez R, Romero Domínguez JM, Talaverón-Rey M, Reche-López D, Suárez-Rivero JM, Álvarez-Córdoba M, Romero-González A, López-Cabrera A, Oliveira MCD, Rodríguez-Sacristan A, Sánchez-Alcázar JA. Polydatin and Nicotinamide Rescue the Cellular Phenotype of Mitochondrial Diseases by Mitochondrial Unfolded Protein Response (mtUPR) Activation. Biomolecules 2024; 14:598. [PMID: 38786005 PMCID: PMC11118892 DOI: 10.3390/biom14050598] [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: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
Primary mitochondrial diseases result from mutations in nuclear DNA (nDNA) or mitochondrial DNA (mtDNA) genes, encoding proteins crucial for mitochondrial structure or function. Given that few disease-specific therapies are available for mitochondrial diseases, novel treatments to reverse mitochondrial dysfunction are necessary. In this work, we explored new therapeutic options in mitochondrial diseases using fibroblasts and induced neurons derived from patients with mutations in the GFM1 gene. This gene encodes the essential mitochondrial translation elongation factor G1 involved in mitochondrial protein synthesis. Due to the severe mitochondrial defect, mutant GFM1 fibroblasts cannot survive in galactose medium, making them an ideal screening model to test the effectiveness of pharmacological compounds. We found that the combination of polydatin and nicotinamide enabled the survival of mutant GFM1 fibroblasts in stress medium. We also demonstrated that polydatin and nicotinamide upregulated the mitochondrial Unfolded Protein Response (mtUPR), especially the SIRT3 pathway. Activation of mtUPR partially restored mitochondrial protein synthesis and expression, as well as improved cellular bioenergetics. Furthermore, we confirmed the positive effect of the treatment in GFM1 mutant induced neurons obtained by direct reprogramming from patient fibroblasts. Overall, we provide compelling evidence that mtUPR activation is a promising therapeutic strategy for GFM1 mutations.
Collapse
Affiliation(s)
- Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - David Gómez-Fernández
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - José Manuel Romero Domínguez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Marta Talaverón-Rey
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Juan Miguel Suárez-Rivero
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Ana Romero-González
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Alejandra López-Cabrera
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Marta Castro De Oliveira
- Neuropediatria, Neurolinkia, C. Jardín de la Isla, 8, Local 4 y 5, 41014 Sevilla, Spain;
- FEA Pediatría, Centro Universitario Hospitalar de Faro, R. Leão Penedo, 8000-386 Faro, Portugal
| | - Andrés Rodríguez-Sacristan
- Neuropediatría, Servicio de Pediatría, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain;
- Departamento de Farmacología, Radiología y Pediatría, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - José Antonio Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.D.); (M.T.-R.); (D.R.-L.); (J.M.S.-R.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| |
Collapse
|
11
|
Ning Y, Dou X, Wang Z, Shi K, Wang Z, Ding C, Sang X, Zhong X, Shao M, Han X, Cao G. SIRT3: A potential therapeutic target for liver fibrosis. Pharmacol Ther 2024; 257:108639. [PMID: 38561088 DOI: 10.1016/j.pharmthera.2024.108639] [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: 12/16/2023] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
Sirtuin3 (SIRT3) is a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase located in the mitochondria, which mainly regulates the acetylation of mitochondrial proteins. In addition, SIRT3 is involved in critical biological processes, including oxidative stress, inflammation, DNA damage, and apoptosis, all of which are closely related to the progression of liver disease. Liver fibrosis characterized by the deposition of extracellular matrix is a result of long termed or repeated liver damage, frequently accompanied by damaged hepatocytes, the recruitment of inflammatory cells, and the activation of hepatic stellate cells. Based on the functions and pharmacology of SIRT3, we will review its roles in liver fibrosis from three aspects: First, the main functions and pharmacological effects of SIRT3 were investigated based on its structure. Second, the roles of SIRT3 in major cells in the liver were summarized to reveal its mechanism in developing liver fibrosis. Last, drugs that regulate SIRT3 to prevent and treat liver fibrosis were discussed. In conclusion, exploring the pharmacological effects of SIRT3, especially in the liver, may be a potential strategy for treating liver fibrosis.
Collapse
Affiliation(s)
- Yan Ning
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyue Dou
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhichao Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kao Shi
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zeping Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Ding
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang Zhong
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meiyu Shao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China; The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Lambona C, Zwergel C, Valente S, Mai A. SIRT3 Activation a Promise in Drug Development? New Insights into SIRT3 Biology and Its Implications on the Drug Discovery Process. J Med Chem 2024; 67:1662-1689. [PMID: 38261767 PMCID: PMC10859967 DOI: 10.1021/acs.jmedchem.3c01979] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024]
Abstract
Sirtuins catalyze deacetylation of lysine residues with a NAD+-dependent mechanism. In mammals, the sirtuin family is composed of seven members, divided into four subclasses that differ in substrate specificity, subcellular localization, regulation, as well as interactions with other proteins, both within and outside the epigenetic field. Recently, much interest has been growing in SIRT3, which is mainly involved in regulating mitochondrial metabolism. Moreover, SIRT3 seems to be protective in diseases such as age-related, neurodegenerative, liver, kidney, heart, and metabolic ones, as well as in cancer. In most cases, activating SIRT3 could be a promising strategy to tackle these health problems. Here, we summarize the main biological functions, substrates, and interactors of SIRT3, as well as several molecules reported in the literature that are able to modulate SIRT3 activity. Among the activators, some derive from natural products, others from library screening, and others from the classical medicinal chemistry approach.
Collapse
Affiliation(s)
- Chiara Lambona
- Department
of Drug Chemistry and Technologies, Sapienza
University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Clemens Zwergel
- Department
of Drug Chemistry and Technologies, Sapienza
University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Sergio Valente
- Department
of Drug Chemistry and Technologies, Sapienza
University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Antonello Mai
- Department
of Drug Chemistry and Technologies, Sapienza
University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
- Pasteur
Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
13
|
Lee H, Yoon H. Mitochondrial sirtuins: Energy dynamics and cancer metabolism. Mol Cells 2024; 47:100029. [PMID: 38331199 PMCID: PMC10960136 DOI: 10.1016/j.mocell.2024.100029] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Mitochondria are pivotal for energy regulation and are linked to cancer. Mitochondrial sirtuins, (Sirtuin) SIRT3, SIRT4, and SIRT5, play crucial roles in cancer metabolism. This review explores their impact on cellular processes, with a focus on the NAD+ interplay and the modulation of their enzymatic activities. The varied roles of SIRT3, SIRT4, and SIRT5 in metabolic adaptation and cancer are outlined, emphasizing their tumor suppressor or oncogenic nature. We propose new insights into sirtuin biology, and cancer therapeutics, suggesting an integrated proteomics and metabolomics approach for a comprehensive understanding of mitochondrial sirtuins in cancer.
Collapse
Affiliation(s)
- Hojun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Haejin Yoon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| |
Collapse
|
14
|
Wang Z, Lin D, Cui B, Zhang D, Wu J, Ma J. Melatonin protects against myocardial ischemia-reperfusion injury by inhibiting excessive mitophagy through the Apelin/SIRT3 signaling axis. Eur J Pharmacol 2024; 963:176292. [PMID: 38128867 DOI: 10.1016/j.ejphar.2023.176292] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Excessive or uncontrolled mitophagy may result in a drastic shortage of healthy mitochondrial for ATP supply after reperfusion, leading to irreversible myocardial damage. Melatonin, a hormone produced by the pineal gland, has been proven to ameliorate myocardial ischemia-reperfusion (I/R) injury via regulating mitophagy. However, its underlying mechanism has not been fully elucidated. The present study focused on the role of mitophagy in the cardioprotective effects of melatonin by using the myocardial I/R rat model. The rats were pretreated with or without the apelin inhibitor ML221, the sirtuin 3 (SIRT3) inhibitor 3-TYP and then subjected to I/R injury, with melatonin administrated 10 min before reperfusion. The effects of melatonin on myocardial infarct size, biomarkers of myocardial injury, oxidative stress, and mitochondrial function were detected, and the expression of apelin, SIRT3, and mitophagy-related proteins were also measured. Excessive mitophagy was activated after I/R injury and was correlated with oxidative stress and mitochondrial dysfunction. Melatonin pretreatment ameliorated myocardial injury by decreasing oxidative stress, restoring mitochondrial function, and inhibiting excessive mitophagy. However, ML221 or 3-TYP disrupted these beneficial effects of melatonin on I/R injury. Taken together, these results suggest that melatonin pretreatment ameliorates myocardial I/R injury through regulating the apelin/SIRT3 pathway to inhibit excessive mitophagy.
Collapse
Affiliation(s)
- Zhaoqi Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Duomao Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Boqun Cui
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Dongni Zhang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jinjing Wu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
15
|
Liu Y, Wei H, Li J. A review on SIRT3 and its natural small molecule activators as a potential Preventive and therapeutic target. Eur J Pharmacol 2024; 963:176155. [PMID: 37914065 DOI: 10.1016/j.ejphar.2023.176155] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
Sirtuins (SIRTs) were originally characterized by yeast Sir2 as a lifespan regulator that is conserved in all three structural domains of bacteria, archaea and eukaryotes and belong to histone deacetylases consisting of seven members (SIRT1-SIRT7). Surprisingly, SIRTs have been shown to play important regulatory roles in almost all cellular functions, including mitochondrial biogenesis, oxidative stress, inflammation, cell growth, energy metabolism, neural function, and stress resistance. Among the SIRT members, sirtuin 3 (SIRT3) is one of the most important deacetylases that regulates the mitochondrial acetylation and plays a role in pathological processes, such as metabolism, DNA repair, oxidative stress, apoptosis and ferroptosis. Therefore, SIRT3 is considered as a potential target for the treatment of a variety of pathological diseases, including metabolic diseases, neurodegenerative diseases, age-related diseases and others. Furthermore, the isolation, screening, and development of SIRT3 signaling agonists, especially from natural products, have become a widely investigated objective. This paper describes the structure of SIRT3 protein, discusses the pathological process of SIRT3-mediated acetylation modification, and reviews the role of SIRT3 in diseases, SIRT3 activators and its related disease studies.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China
| | - Haidong Wei
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, 150030, China.
| |
Collapse
|
16
|
Zhang Q, Siyuan Z, Xing C, Ruxiu L. SIRT3 regulates mitochondrial function: A promising star target for cardiovascular disease therapy. Biomed Pharmacother 2024; 170:116004. [PMID: 38086147 DOI: 10.1016/j.biopha.2023.116004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Dysregulation of mitochondrial homeostasis is common to all types of cardiovascular diseases. SIRT3 regulates apoptosis and autophagy, material and energy metabolism, mitochondrial oxidative stress, inflammation, and fibrosis. As an important mediator and node in the network of mechanisms, SIRT3 is essential to many activities. This review explains how SIRT3 regulates mitochondrial homeostasis and the tricarboxylic acid cycle to treat common cardiovascular diseases. A novel description of the impact of lifestyle factors on SIRT3 expression from the angles of nutrition, exercise, and temperature is provided.
Collapse
Affiliation(s)
- Qin Zhang
- Guang'anmen Hospital, Chinese Academy of traditional Chinese medicine, Beijing, China
| | - Zhou Siyuan
- Guang'anmen Hospital, Chinese Academy of traditional Chinese medicine, Beijing, China
| | - Chang Xing
- Guang'anmen Hospital, Chinese Academy of traditional Chinese medicine, Beijing, China
| | - Liu Ruxiu
- Guang'anmen Hospital, Chinese Academy of traditional Chinese medicine, Beijing, China.
| |
Collapse
|
17
|
Foglio E, D’Avorio E, Vitiello L, Masuelli L, Bei R, Pacifici F, Della-Morte D, Mirabilii S, Ricciardi MR, Tafuri A, Garaci E, Russo MA, Tafani M, Limana F. Doxorubicin-Induced Cardiac Senescence Is Alleviated Following Treatment with Combined Polyphenols and Micronutrients through Enhancement in Mitophagy. Cells 2023; 12:2605. [PMID: 37998340 PMCID: PMC10670650 DOI: 10.3390/cells12222605] [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: 06/28/2023] [Revised: 10/29/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Oxidative stress and impaired mitophagy are the hallmarks of cardiomyocyte senescence. Specifically, a decrease in mitophagic flux leads to the accumulation of damaged mitochondria and the development of senescence through increased ROS and other mediators. In this study, we describe the preventive role of A5+, a mix of polyphenols and other micronutrients, in doxorubicin (DOXO)-induced senescence of H9C2 cells. Specifically, H9C2 cells exposed to DOXO showed an increase in the protein expression proteins of senescence-associated genes, p21 and p16, and a decrease in the telomere binding factors TRF1 and TRF2, indicative of senescence induction. Nevertheless, A5+ pre-treatment attenuated the senescent-like cell phenotype, as evidenced by inhibition of all senescent markers and a decrease in SA-β-gal staining in DOXO-treated H9C2 cells. Importantly, A5+ restored the LC3 II/LC3 I ratio, Parkin and BNIP3 expression, therefore rescuing mitophagy, and decreased ROS production. Further, A5+ pre-treatment determined a ripolarization of the mitochondrial membrane and improved basal respiration. A5+-mediated protective effects might be related to its ability to activate mitochondrial SIRT3 in synergy with other micronutrients, but in contrast with SIRT4 activation. Accordingly, SIRT4 knockdown in H9C2 cells further increased MnSOD activity, enhanced mitophagy, and reduced ROS generation following A5+ pre-treatment and DOXO exposure compared to WT cells. Indeed, we demonstrated that A5+ protects H9C2 cells from DOXO-induced senescence, establishing a new specific role for A5+ in controlling mitochondrial quality control by restoring SIRT3 activity and mitophagy, which provided a molecular basis for the development of therapeutic strategies against cardiomyocyte senescence.
Collapse
Affiliation(s)
- Eleonora Foglio
- Technoscience, Parco Scientifico e Tecnologico Pontino, 04100 Latina, Italy
| | - Erica D’Avorio
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
| | | | - Laura Masuelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (L.M.); (M.T.)
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Francesca Pacifici
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - David Della-Morte
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
- Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Simone Mirabilii
- Hematology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Maria Rosaria Ricciardi
- Hematology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Agostino Tafuri
- Hematology, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (M.R.R.); (A.T.)
| | - Enrico Garaci
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
| | - Matteo Antonio Russo
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
- IRCCS San Raffaele Roma, 00166 Rome, Italy;
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (L.M.); (M.T.)
| | - Federica Limana
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, 00166 Rome, Italy (F.P.); (D.D.-M.); (E.G.); (M.A.R.)
- Laboratory of Cellular and Molecular Pathology, IRCCS San Raffaele Roma, 00166 Rome, Italy
| |
Collapse
|
18
|
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
|
19
|
Mishra Y, Kumar Kaundal R. Role of SIRT3 in mitochondrial biology and its therapeutic implications in neurodegenerative disorders. Drug Discov Today 2023; 28:103583. [PMID: 37028501 DOI: 10.1016/j.drudis.2023.103583] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/19/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023]
Abstract
Sirtuin 3 (SIRT3), a mitochondrial deacetylase expressed preferentially in high-metabolic-demand tissues including the brain, requires NAD+ as a cofactor for catalytic activity. It regulates various processes such as energy homeostasis, redox balance, mitochondrial quality control, mitochondrial unfolded protein response (UPRmt), biogenesis, dynamics and mitophagy by altering protein acetylation status. Reduced SIRT3 expression or activity causes hyperacetylation of hundreds of mitochondrial proteins, which has been linked with neurological abnormalities, neuro-excitotoxicity and neuronal cell death. A body of evidence has suggested, SIRT3 activation as a potential therapeutic modality for age-related brain abnormalities and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yogesh Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (UP)-226002, India
| | - Ravinder Kumar Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow (UP)-226002, India.
| |
Collapse
|
20
|
Jiang B, Zhou X, Yang T, Wang L, Feng L, Wang Z, Xu J, Jing W, Wang T, Su H, Yang G, Zhang Z. The role of autophagy in cardiovascular disease: Cross-interference of signaling pathways and underlying therapeutic targets. Front Cardiovasc Med 2023; 10:1088575. [PMID: 37063954 PMCID: PMC10090687 DOI: 10.3389/fcvm.2023.1088575] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
Autophagy is a conserved lysosomal pathway for the degradation of cytoplasmic proteins and organelles, which realizes the metabolic needs of cells and the renewal of organelles. Autophagy-related genes (ATGs) are the main molecular mechanisms controlling autophagy, and their functions can coordinate the whole autophagic process. Autophagy can also play a role in cardiovascular disease through several key signaling pathways, including PI3K/Akt/mTOR, IGF/EGF, AMPK/mTOR, MAPKs, p53, Nrf2/p62, Wnt/β-catenin and NF-κB pathways. In this paper, we reviewed the signaling pathway of cross-interference between autophagy and cardiovascular diseases, and analyzed the development status of novel cardiovascular disease treatment by targeting the core molecular mechanism of autophagy as well as the critical signaling pathway. Induction or inhibition of autophagy through molecular mechanisms and signaling pathways can provide therapeutic benefits for patients. Meanwhile, we hope to provide a unique insight into cardiovascular treatment strategies by understanding the molecular mechanism and signaling pathway of crosstalk between autophagy and cardiovascular diseases.
Collapse
Affiliation(s)
- Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xuan Zhou
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Yang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linlin Wang
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Longfei Feng
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zheng Wang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jin Xu
- Department of First Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Wang
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Haixiang Su
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - GuoWei Yang
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| | - Zheng Zhang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
21
|
Cherian S, Hacisayidli KM, Kurian R, Mathews A. Therapeutically important bioactive compounds of the genus Polygonum L. and their possible interventions in clinical medicine. J Pharm Pharmacol 2023; 75:301-327. [PMID: 36757388 DOI: 10.1093/jpp/rgac105] [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: 06/28/2022] [Accepted: 12/26/2022] [Indexed: 02/10/2023]
Abstract
OBJECTIVES Increasing literature data have suggested that the genus Polygonum L. possesses pharmacologically important plant secondary metabolites. These bioactive compounds are implicated as effective agents in preclinical and clinical practice due to their pharmacological effects such as anti-inflammatory, anticancer, antidiabetic, antiaging, neuroprotective or immunomodulatory properties among many others. However, elaborate pharmacological and clinical data concerning the bioavailability, tissue distribution pattern, dosage and pharmacokinetic profiles of these compounds are still scanty. KEY FINDINGS The major bioactive compounds implicated in the therapeutic effects of Polygonum genus include phenolic and flavonoid compounds, anthraquinones and stilbenes, such as quercetin, resveratrol, polydatin and others, and could serve as potential drug leads or as adjuvant agents. Data from in-silico network pharmacology and computational molecular docking studies are also highly helpful in identifying the possible drug target of pathogens or host cell machinery. SUMMARY We provide an up-to-date overview of the data from pharmacodynamic, pharmacokinetic profiles and preclinical (in-vitro and in-vivo) investigations and the available clinical data on some of the therapeutically important compounds of genus Polygonum L. and their medical interventions, including combating the outbreak of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Sam Cherian
- Indian Society for Plant Physiology, New Delhi, India
| | - Kushvar Mammadova Hacisayidli
- Department of Hygiene and Food Safety, Veterinary Medicine Faculty, Azerbaijan State Agricultural University, Ganja City, Azerbaijan
| | - Renju Kurian
- Department of Pathology, Manipal University College, Melaka, Malaysia
| | - Allan Mathews
- Faculty of Pharmacy, Quest International University Perak, Ipoh, Malaysia
| |
Collapse
|
22
|
Li ZY, Lu GQ, Lu J, Wang PX, Zhang XL, Zou Y, Liu PQ. SZC-6, a small-molecule activator of SIRT3, attenuates cardiac hypertrophy in mice. Acta Pharmacol Sin 2023; 44:546-560. [PMID: 36042291 PMCID: PMC9958013 DOI: 10.1038/s41401-022-00966-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/24/2022] [Indexed: 11/09/2022]
Abstract
Sirtuin3 (SIRT3), a class III histone deacetylase, is implicated in various cardiovascular diseases as a novel therapeutic target. SIRT3 has been proven to be cardioprotective in a model of Ang II-induced cardiac hypertrophy. However, a few small-molecule compounds targeting deacetylases could activate SIRT3. In this study, we generated a novel SIRT3 activator, 3-(2-bromo-4-hydroxyphenyl)-7-hydroxy-2H-chromen-2-one (SZC-6), through structural optimization of the first SIRT3 agonist C12. We demonstrated that SZC-6 directly bound to SIRT3 with Kd value of 15 μM, and increased SIRT3 deacetylation activity with EC50 value of 23.2 ± 3.3 µM. In neonatal rat cardiomyocytes (NRCMs), pretreatment with SZC-6 (10, 20, 40 µM) dose-dependently attenuated isoproterenol (ISO)-induced hypertrophic responses. Administration of SZC-6 (20, 40 and 60 mg·kg-1·d-1, s.c.) for 2 weeks starting from one week prior ISO treatment dose-dependently reversed ISO-induced impairment of diastolic and systolic cardiac function in wild-type mice, but not in SIRT3 knockdown mice. We showed that SZC-6 (10, 20, 40 µM) dose-dependently inhibited cardiac fibroblast proliferation and differentiation into myofibroblasts, which was abolished in SIRT3-knockdown mice. We further revealed that activation of SIRT3 by SZC-6 increased ATP production and rate of mitochondrial oxygen consumption, and reduced ROS, improving mitochondrial function in ISO-treated NRCMs. We also found that SZC-6 dose-dependently enhanced LKB1 phosphorylation, thereby promoting AMPK activation to inhibit Drp1-dependent mitochondrial fragmentation. Taken together, these results demonstrate that SZC-6 is a novel SIRT3 agonist with potential value in the treatment of cardiac hypertrophy partly through activation of the LKB1-AMPK pathway.
Collapse
Affiliation(s)
- Ze-Yu Li
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guo-Qing Lu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Lu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Pan-Xia Wang
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xiao-Lei Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yong Zou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Pei-Qing Liu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratoty for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
23
|
Farooq U, Wang H, Hu J, Li G, Jehan S, Shi J, Li D, Sui G. Polydatin Inhibits Hepatocellular Carcinoma Cell Proliferation and Sensitizes Doxorubicin and Cisplatin through Targeting Cell Mitotic Machinery. Cells 2023; 12:cells12020222. [PMID: 36672157 PMCID: PMC9856937 DOI: 10.3390/cells12020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/22/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Polydatin (PD) is a natural compound with anticancer activities, but the underlying mechanisms remain largely unclear. To understand how PD inhibited hepatocellular carcinoma (HCC), we studied PD treatments in HCC HepG2 and SK-HEP1 cells, and normal liver HL-7702 cells. PD selectively blocked the proliferation of HCC cells but showed low toxicity in normal cells, while the effects of doxorubicin (DOX) and cisplatin (DDP) on HCC and normal liver cells were opposite. In the cotreatment studies, PD synergistically improved the inhibitory activities of DOX and DDP in HCC cells but alleviated their toxicity in HL-7702 cells. Furthermore, RNA-seq studies of PD-treated HepG2 cells revealed multiple altered signaling pathways. We identified 1679 Differentially Expressed Genes (DEGs) with over a 2.0-fold change in response to PD treatment. Integrative analyses using the DEGs in PD-treated HepG2 cells and DEGs in a TCGA dataset of HCC patients revealed five PD-repressed DEGs regulating mitotic spindle midzone formation. The expression of these genes showed significantly positive correlation with poor clinical outcomes of HCC patients, suggesting that mitotic machinery was likely a primary target of PD. Our findings improve the understanding of PD's anticancer mechanisms and provide insights into developing effective clinical approaches in HCC therapies.
Collapse
Affiliation(s)
- Umar Farooq
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Hao Wang
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Jingru Hu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Guangyue Li
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Shah Jehan
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Jinming Shi
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
| | - Dangdang Li
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
- Correspondence: (D.L.); (G.S.)
| | - Guangchao Sui
- College of Life Sciences, Northeast Forestry University, 26 Hexing Road, Harbin 150040, China
- Correspondence: (D.L.); (G.S.)
| |
Collapse
|
24
|
Liu Y, Huang Y, Xu C, An P, Luo Y, Jiao L, Luo J, Li Y. Mitochondrial Dysfunction and Therapeutic Perspectives in Cardiovascular Diseases. Int J Mol Sci 2022; 23:16053. [PMID: 36555691 PMCID: PMC9788331 DOI: 10.3390/ijms232416053] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/21/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
High mortality rates due to cardiovascular diseases (CVDs) have attracted worldwide attention. It has been reported that mitochondrial dysfunction is one of the most important mechanisms affecting the pathogenesis of CVDs. Mitochondrial DNA (mtDNA) mutations may result in impaired oxidative phosphorylation (OXPHOS), abnormal respiratory chains, and ATP production. In dysfunctional mitochondria, the electron transport chain (ETC) is uncoupled and the energy supply is reduced, while reactive oxygen species (ROS) production is increased. Here, we discussed and analyzed the relationship between mtDNA mutations, impaired mitophagy, decreased OXPHOS, elevated ROS, and CVDs from the perspective of mitochondrial dysfunction. Furthermore, we explored current potential therapeutic strategies for CVDs by eliminating mtDNA mutations (e.g., mtDNA editing and mitochondrial replacement), enhancing mitophagy, improving OXPHOS capacity (e.g., supplement with NAD+, nicotinamide riboside (NR), nicotinamide mononucleotide (NMN), and nano-drug delivery), and reducing ROS (e.g., supplement with Coenzyme Q10 and other antioxidants), and dissected their respective advantages and limitations. In fact, some therapeutic strategies are still a long way from achieving safe and effective clinical treatment. Although establishing effective and safe therapeutic strategies for CVDs remains challenging, starting from a mitochondrial perspective holds bright prospects.
Collapse
Affiliation(s)
- Yu Liu
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Yuejia Huang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Chong Xu
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Peng An
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yongting Luo
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Lei Jiao
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Junjie Luo
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yongzhi Li
- China Astronaut Research and Training Center, Beijing 100094, China
| |
Collapse
|
25
|
Karami A, Fakhri S, Kooshki L, Khan H. Polydatin: Pharmacological Mechanisms, Therapeutic Targets, Biological Activities, and Health Benefits. Molecules 2022; 27:6474. [PMID: 36235012 PMCID: PMC9572446 DOI: 10.3390/molecules27196474] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
Polydatin is a natural potent stilbenoid polyphenol and a resveratrol derivative with improved bioavailability. Polydatin possesses potential biological activities predominantly through the modulation of pivotal signaling pathways involved in inflammation, oxidative stress, and apoptosis. Various imperative biological activities have been suggested for polydatin towards promising therapeutic effects, including anticancer, cardioprotective, anti-diabetic, gastroprotective, hepatoprotective, neuroprotective, anti-microbial, as well as health-promoting roles on the renal system, the respiratory system, rheumatoid diseases, the skeletal system, and women's health. In the present study, the therapeutic targets, biological activities, pharmacological mechanisms, and health benefits of polydatin are reviewed to provide new insights to researchers. The need to develop further clinical trials and novel delivery systems of polydatin is also considered to reveal new insights to researchers.
Collapse
Affiliation(s)
- Ahmad Karami
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Leila Kooshki
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| |
Collapse
|
26
|
Şöhretoğlu D, Barut B, Sari S, Özel A, Kuruüzüm-Uz A, Arroo R. In Vitro and in Silico Investigation of DNA Interaction, Topoisomerase I and II Inhibitory Properties of Polydatin. Chem Biodivers 2022; 19:e202200352. [PMID: 36149030 DOI: 10.1002/cbdv.202200352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 09/05/2022] [Indexed: 11/11/2022]
Abstract
Polydatin or piceid, is the 3-O-glucoside of resveratrol and is found abundantly in grapes, peanuts, wine, beer, and cacao products. Although anticancer activity of polydatin was reported before, and potential antiproliferative mechanisms of polydatin have been proposed, its direct effects on DNA and inhibitory potential against topoisomerase enzymes have remained unknown. In this study we aimed to reveal the link between polydatin's effects on DNA and DNA-topoisomerases and its antiproliferative promise. For this purpose, we evaluated the effects of polydatin on DNA and DNA topoisomerase using in vitro and in silico techniques. Polydatin was found to protect DNA against Fenton reaction-induced damage while not showing any hydrolytic nuclease effect. Further, polydatin inhibited topoisomerase II but not topoisomerase I. According to molecular docking studies, polydatin preferably showed minor groove binding to DNA where the stilbene moiety was important for binding to the DNA-topoisomerase II complex. As a result, topoisomerase II inhibition might be another anticancer mechanism of polydatin.
Collapse
Affiliation(s)
- Didem Şöhretoğlu
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacognosy, Sıhhiye, Ankara, TR-06100, Ankara, Turkey
| | - Burak Barut
- Karadeniz Technical University, Faculty of Pharmacy, Department of Biochemistry, Trabzon, Turkey
| | - Suat Sari
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Sıhhiye, Ankara, TR-06100, Ankara, Turkey
| | - Arzu Özel
- Karadeniz Technical University, Faculty of Pharmacy, Department of Biochemistry, Trabzon, Turkey.,Karadeniz Technical University, Drug and Pharmaceutical Technology Application and Research Center, Trabzon, Turkey
| | - Ayşe Kuruüzüm-Uz
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacognosy, Sıhhiye, Ankara, TR-06100, Ankara, Turkey
| | - Randolph Arroo
- De Montfort University, Leicester School of Pharmacy, The Gateway, Leicester, LE1 9BH, United Kingdom
| |
Collapse
|
27
|
Yu LM, Dong X, Li N, Jiang H, Zhao JK, Xu YL, Xu DY, Xue XD, Zhou ZJ, Huang YT, Zhao QS, Wang ZS, Yin ZT, Wang HS. Polydatin attenuates chronic alcohol consumption-induced cardiomyopathy through a SIRT6-dependent mechanism. Food Funct 2022; 13:7302-7319. [PMID: 35726783 DOI: 10.1039/d2fo00966h] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Polydatin has attracted much attention as a potential cardioprotective agent against ischemic heart disease and diabetic cardiomyopathy. However, the effect and mechanism of polydatin supplementation on alcoholic cardiomyopathy (ACM) are still unknown. This study aimed to determine the therapeutic effect of polydatin against ACM and to explore the molecular mechanisms with a focus on SIRT6-AMP-activated protein kinase (AMPK) signaling and mitochondrial function. The ACM model was established by feeding C57/BL6 mice with an ethanol Lieber-DeCarli diet for 12 weeks. The mice received polydatin (20 mg kg-1) or vehicle treatment. We showed that polydatin treatment not only improved cardiac function but also reduced myocardial fibrosis and dynamin-related protein 1 (Drp-1)-mediated mitochondrial fission, and enhanced PTEN-induced putative kinase 1 (PINK1)-Parkin-dependent mitophagy in alcohol-treated myocardium. Importantly, these beneficial effects were mimicked by SIRT6 overexpression but abolished by the infection of recombinant serotype 9 adeno-associated virus (AAV9) carrying SIRT6-specific small hairpin RNA. Mechanistically, alcohol consumption induced a gradual decrease in the myocardial SIRT6 level, while polydatin effectively activated SIRT6-AMPK signaling and modulated mitochondrial dynamics and mitophagy, thus reducing oxidative stress damage and preserving mitochondrial function. In summary, these data present new information regarding the therapeutic actions of polydatin, suggesting that the activation of SIRT6 signaling may represent a new approach for tackling ACM-related cardiac dysfunction.
Collapse
Affiliation(s)
- Li-Ming Yu
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Xue Dong
- The Third Outpatient Department, General Hospital of Northern Theater Command, 49 Beiling Road, Shenyang, Liaoning 110032, P. R. China
| | - Ning Li
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Hui Jiang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Ji-Kai Zhao
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Yin-Li Xu
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Deng-Yue Xu
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Xiao-Dong Xue
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Zi-Jun Zhou
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Yu-Ting Huang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Qiu-Sheng Zhao
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Zhi-Shang Wang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Zong-Tao Yin
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| | - Hui-Shan Wang
- Department of Cardiovascular Surgery, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, P. R. China.
| |
Collapse
|
28
|
Zeng X, Zhang YD, Ma RY, Chen YJ, Xiang XM, Hou DY, Li XH, Huang H, Li T, Duan CY. Activated Drp1 regulates p62-mediated autophagic flux and aggravates inflammation in cerebral ischemia-reperfusion via the ROS-RIP1/RIP3-exosome axis. Mil Med Res 2022; 9:25. [PMID: 35624495 PMCID: PMC9137164 DOI: 10.1186/s40779-022-00383-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/06/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CIRI) refers to a secondary brain injury that can occur when the blood supply to the ischemic brain tissue is restored. However, the mechanism underlying such injury remains elusive. METHODS The 150 male C57 mice underwent middle cerebral artery occlusion (MCAO) for 1 h and reperfusion for 24 h, Among them, 50 MCAO mice were further treated with Mitochondrial division inhibitor 1 (Mdivi-1) and 50 MCAO mice were further treated with N-acetylcysteine (NAC). SH-SY5Y cells were cultured in a low-glucose culture medium for 4 h under hypoxic conditions and then transferred to normal conditions for 12 h. Then, cerebral blood flow, mitochondrial structure, mitochondrial DNA (mtDNA) copy number, intracellular and mitochondrial reactive oxygen species (ROS), autophagic flux, aggresome and exosome expression profiles, cardiac tissue structure, mitochondrial length and cristae density, mtDNA and ROS content, as well as the expression of Drp1-Ser616/Drp1, RIP1/RIP3, LC3 II/LC3 I, TNF-α, IL-1β, etc., were detected under normal or Drp1 interference conditions. RESULTS The mtDNA content, ROS levels, and Drp1-Ser616/Drp1 were elevated by 2.2, 1.7 and 2.7 times after CIRI (P < 0.05). However, the high cytoplasmic LC3 II/I ratio and increased aggregation of p62 could be reversed by 44% and 88% by Drp1 short hairpin RNA (shRNA) (P < 0.05). The low fluorescence intensity of autophagic flux and the increased phosphorylation of RIP3 induced by CIRI could be attenuated by ROS scavenger, NAC (P < 0.05). RIP1/RIP3 inhibitor Necrostatin-1 (Nec-1) restored 75% to a low LC3 II/LC3 I ratio and enhanced 2 times to a high RFP-LC3 after Drp1 activation (P < 0.05). In addition, although CIRI-induced ROS production caused no considerable accumulation of autophagosomes (P > 0.05), it increased the packaging and extracellular secretion of exosomes containing p62 by 4 - 5 times, which could be decreased by Mdivi-1, Drp1 shRNA, and Nec-1 (P < 0.05). Furthermore, TNF-α and IL-1β increased in CIRI-derived exosomes could increase RIP3 phosphorylation in normal or oxygen-glucose deprivation/reoxygenation (OGD/R) conditions (P < 0.05). CONCLUSIONS CIRI activated Drp1 and accelerated the p62-mediated formation of autophagosomes while inhibiting the transition of autophagosomes to autolysosomes via the RIP1/RIP3 pathway activation. Undegraded autophagosomes were secreted extracellularly in the form of exosomes, leading to inflammatory cascades that further damaged mitochondria, resulting in excessive ROS generation and the blockage of autophagosome degradation, triggering a vicious cycle.
Collapse
Affiliation(s)
- Xue Zeng
- Department of Anaesthesiology, the Second Affiliated Hospital of Chongqing Medical University, 400010, Chongqing, China.,Department of Neurology, the Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yun-Dong Zhang
- Department of Neurology, the Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Rui-Yan Ma
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yuan-Jing Chen
- Department of Anaesthesiology, the Second Affiliated Hospital of Chongqing Medical University, 400010, Chongqing, China
| | - Xin-Ming Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dong-Yao Hou
- Department of Anaesthesiology, the Second Affiliated Hospital of Chongqing Medical University, 400010, Chongqing, China
| | - Xue-Han Li
- Department of Anaesthesiology, the Second Affiliated Hospital of Chongqing Medical University, 400010, Chongqing, China
| | - He Huang
- Department of Anaesthesiology, the Second Affiliated Hospital of Chongqing Medical University, 400010, Chongqing, China.
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Chen-Yang Duan
- Department of Anaesthesiology, the Second Affiliated Hospital of Chongqing Medical University, 400010, Chongqing, China.
| |
Collapse
|
29
|
Ouyang S, Zhang Q, Lou L, Zhu K, Li Z, Liu P, Zhang X. The Double-Edged Sword of SIRT3 in Cancer and Its Therapeutic Applications. Front Pharmacol 2022; 13:871560. [PMID: 35571098 PMCID: PMC9092499 DOI: 10.3389/fphar.2022.871560] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Reprogramming of cellular energy metabolism is considered an emerging feature of cancer. Mitochondrial metabolism plays a crucial role in cancer cell proliferation, survival, and metastasis. As a major mitochondrial NAD+-dependent deacetylase, sirtuin3 (SIRT3) deacetylates and regulates the enzymes involved in regulating mitochondrial energy metabolism, including fatty acid oxidation, the Krebs cycle, and the respiratory chain to maintain metabolic homeostasis. In this article, we review the multiple roles of SIRT3 in various cancers, and systematically summarize the recent advances in the discovery of its activators and inhibitors. The roles of SIRT3 vary in different cancers and have cell- and tumor-type specificity. SIRT3 plays a unique function by mediating interactions between mitochondria and intracellular signaling. The critical functions of SIRT3 have renewed interest in the development of small molecule modulators that regulate its activity. Delineation of the underlying mechanism of SIRT3 as a critical regulator of cell metabolism and further characterization of the mitochondrial substrates of SIRT3 will deepen our understanding of the role of SIRT3 in tumorigenesis and progression and may provide novel therapeutic strategies for cancer targeting SIRT3.
Collapse
Affiliation(s)
- Shumin Ouyang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Qiyi Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Linlin Lou
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Kai Zhu
- Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, China
| | - Zeyu Li
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Peiqing Liu
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Xiaolei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
30
|
Polydatin Attenuates Intra-Uterine Growth Retardation-Induced Liver Injury and Mitochondrial Dysfunction in Weanling Piglets by Improving Energy Metabolism and Redox Balance. Antioxidants (Basel) 2022; 11:antiox11040666. [PMID: 35453351 PMCID: PMC9028342 DOI: 10.3390/antiox11040666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 02/08/2023] Open
Abstract
The present study investigated the potential of polydatin to protect against liver injury and the mitochondrial dysfunction of weanling piglets suffering from intra-uterine growth retardation (IUGR). Thirty-six normal birth weight weanling piglets and an equal number of IUGR littermates were given a basal diet with or without polydatin (250 mg/kg) from 21 to 35 d of age. Plasma and liver samples were collected to measure biochemistry parameters at 35 d of age. IUGR caused hepatic apoptosis, mitochondrial dysfunction, and oxidative damage, along with a lower efficiency of energy metabolism and inferior antioxidant ability. Polydatin decreased apoptotic rate, improved the features of mitochondrial damage, inhibited mitochondrial swelling and superoxide anion formation, and preserved mitochondrial membrane potential in the liver. Concurrently, polydatin promoted mitochondrial biogenesis, increased sirtuin 1 activity, and upregulated the expression levels of several genes related to mitochondrial function and fitness. Polydatin also facilitated mitochondrial oxidative metabolism with a beneficial outcome of increased energy production. Furthermore, polydatin mitigated the IUGR-induced reduction in manganese superoxide dismutase activity and prevented the excessive accumulation of oxidative damaging products in the liver. These findings indicate that polydatin confers protection against hepatic injury and mitochondrial dysfunction in the IUGR piglets by improving energy metabolism and redox balance.
Collapse
|
31
|
Murugasamy K, Munjal A, Sundaresan NR. Emerging Roles of SIRT3 in Cardiac Metabolism. Front Cardiovasc Med 2022; 9:850340. [PMID: 35369299 PMCID: PMC8971545 DOI: 10.3389/fcvm.2022.850340] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/31/2022] [Indexed: 12/17/2022] Open
Abstract
The heart is a highly metabolically active organ that predominantly utilizes fatty acids as an energy substrate. The heart also derives some part of its energy by oxidation of other substrates, including glucose, lactose, amino acids and ketones. The critical feature of cardiac pathology is metabolic remodeling and loss of metabolic flexibility. Sirtuin 3 (SIRT3) is one of the seven mammalian sirtuins (SIRT1 to SIRT7), with NAD+ dependent deacetylase activity. SIRT3 is expressed in high levels in healthy hearts but downregulated in the aged or diseased hearts. Experimental evidence shows that increasing SIRT3 levels or activity can ameliorate several cardiac pathologies. The primary deacetylation targets of SIRT3 are mitochondrial proteins, most of which are involved in energy metabolism. Thus, SIRT3 improves cardiac health by modulating cardiac energetics. In this review, we discuss the essential role of SIRT3 in regulating cardiac metabolism in the context of physiology and pathology. Specifically, we summarize the recent advancements that emphasize the critical role of SIRT3 as a master regulator of cardiac metabolism. We also present a comprehensive view of all known activators of SIRT3, and elaborate on their therapeutic potential to ameliorate energetic abnormalities in various cardiac pathologies.
Collapse
|
32
|
Cao M, Zhao Q, Sun X, Qian H, Lyu S, Chen R, Xia H, Yuan W. Sirtuin 3: Emerging therapeutic target for cardiovascular diseases. Free Radic Biol Med 2022; 180:63-74. [PMID: 35031448 DOI: 10.1016/j.freeradbiomed.2022.01.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 12/26/2022]
Abstract
Acetylation is one of the most important methods of modification that lead to a change in the function of proteins. In humans, metabolic enzymes commonly undergo acetylation, which regulates the activities of metabolic enzymes and metabolic pathways. Sirtuin 3 (SIRT3) is a prominent deacetylase that participates in mitochondrial metabolism, redox balance, and mitochondrial dynamics by regulating mitochondrial protein acetylation, thereby protecting mitochondria from damage. Normal mitochondrial function is essential for maintaining the metabolism and function of the heart. Therefore, mitochondrial dysfunction caused by SIRT3 consumption and defects leads to the development of a variety of cardiovascular diseases. A comprehensive understanding of the role of SIRT3 in cardiovascular disease is critical for developing new therapeutic strategies. Herein, we summarize the function of SIRT3 in mitochondria, the complex mechanisms mediating cardiovascular diseases, and the potential value of SIRT3 small-molecule agonists in future clinical treatments.
Collapse
Affiliation(s)
- Mengfei Cao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Qianru Zhao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Xia Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Han Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Shumei Lyu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Rui Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Hao Xia
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212000, China.
| |
Collapse
|
33
|
Tang D, Zhang Q, Duan H, Ye X, Liu J, Peng W, Wu C. Polydatin: A Critical Promising Natural Agent for Liver Protection via Antioxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9218738. [PMID: 35186191 PMCID: PMC8853764 DOI: 10.1155/2022/9218738] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 01/18/2022] [Indexed: 12/11/2022]
Abstract
Polydatin, one of the natural active small molecules, was commonly applied in protecting and treating liver disorders in preclinical studies. Oxidative stress plays vital roles in liver injury caused by various factors, such as alcohol, viral infections, dietary components, drugs, and other chemical reagents. It is reported that oxidative stress might be one of the main reasons in the progressive development of alcohol liver diseases (ALDs), nonalcoholic liver diseases (NAFLDs), liver injury, fibrosis, hepatic failure (HF), and hepatocellular carcinoma (HCC). In this paper, we comprehensively summarized the pharmacological effects and potential molecular mechanisms of polydatin for protecting and treating liver disorders via regulation of oxidative stress. According to the previous studies, polydatin is a versatile natural compound and exerts significantly protective and curative effects on oxidative stress-associated liver diseases via various molecular mechanisms, including amelioration of liver function and insulin resistance, inhibition of proinflammatory cytokines, lipid accumulation, endoplasmic reticulum stress and autophagy, regulation of PI3K/Akt/mTOR, and activation of hepatic stellate cells (HSCs), as well as increase of antioxidant enzymes (such as catalase (CAT), glutathione peroxidase (GPx), glutathione (GSH), superoxide dismutase (SOD), glutathione reductase (GR), and heme oxygenase-1 (HO-1)). In addition, polydatin acts as a free radical scavenger against reactive oxygen species (ROS) by its phenolic and ethylenic bond structure. However, further clinical investigations are still needed to explore the comprehensive molecular mechanisms and confirm the clinical treatment effect of polydatin in liver diseases related to regulation of oxidative stress.
Collapse
Affiliation(s)
- Dandan Tang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Qing Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Huxinyue Duan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Xun Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Jia Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Wei Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Chunjie Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| |
Collapse
|
34
|
Figarola-Centurión I, Escoto-Delgadillo M, González-Enríquez GV, Gutiérrez-Sevilla JE, Vázquez-Valls E, Torres-Mendoza BM. Sirtuins Modulation: A Promising Strategy for HIV-Associated Neurocognitive Impairments. Int J Mol Sci 2022; 23:643. [PMID: 35054829 PMCID: PMC8775450 DOI: 10.3390/ijms23020643] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 02/01/2023] Open
Abstract
HIV-Associated neurocognitive disorder (HAND) is one of the major concerns since it persists in 40% of this population. Nowadays, HAND neuropathogenesis is considered to be caused by the infected cells that cross the brain-blood barrier and produce viral proteins that can be secreted and internalized into neurons leading to disruption of cellular processes. The evidence points to viral proteins such as Tat as the causal agent for neuronal alteration and thus HAND. The hallmarks in Tat-induced neurodegeneration are endoplasmic reticulum stress and mitochondrial dysfunction. Sirtuins (SIRTs) are NAD+-dependent deacetylases involved in mitochondria biogenesis, unfolded protein response, and intrinsic apoptosis pathway. Tat interaction with these deacetylases causes inhibition of SIRT1 and SIRT3. Studies revealed that SIRTs activation promotes neuroprotection in neurodegenerative diseases such Alzheimer's and Parkinson's disease. Therefore, this review focuses on Tat-induced neurotoxicity mechanisms that involve SIRTs as key regulators and their modulation as a therapeutic strategy for tackling HAND and thereby improving the quality of life of people living with HIV.
Collapse
Affiliation(s)
- Izchel Figarola-Centurión
- Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Mexico;
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (M.E.-D.); (J.E.G.-S.)
| | - Martha Escoto-Delgadillo
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (M.E.-D.); (J.E.G.-S.)
- Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Guadalajara 44600, Mexico
| | - Gracia Viviana González-Enríquez
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Juan Ernesto Gutiérrez-Sevilla
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (M.E.-D.); (J.E.G.-S.)
- Microbiología Médica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Eduardo Vázquez-Valls
- Generación de Recursos Profesionales, Investigación y Desarrollo, Secretaria de Salud, Jalisco, Guadalajara 44100, Mexico;
| | - Blanca Miriam Torres-Mendoza
- Laboratorio de Inmunodeficiencias y Retrovirus Humanos, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (M.E.-D.); (J.E.G.-S.)
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| |
Collapse
|
35
|
Videla LA, Marimán A, Ramos B, José Silva M, Del Campo A. Standpoints in mitochondrial dysfunction: Underlying mechanisms in search of therapeutic strategies. Mitochondrion 2022; 63:9-22. [PMID: 34990812 DOI: 10.1016/j.mito.2021.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023]
Abstract
Mitochondrial dysfunction has been defined as a reduced efficiency of mitochondria to produce ATP given by a loss of mitochondrial membrane potential, alterations in the electron transport chain (ETC) function, with increase in reactive oxygen species (ROS) generation and decrease in oxygen consumption. During the last decades, mitochondrial dysfunction has been the focus of many researchers as a convergent point for the pathophysiology of several diseases. Numerous investigations have demonstrated that mitochondrial dysfunction is detrimental to cells, tissues and organisms, nevertheless, dysfunctional mitochondria can signal in a particular way in response to stress, a characteristic that may be useful to search for new therapeutic strategies with a common feature. The aim of this review addresses mitochondrial dysfunction and stress signaling as a promising target for future drug development.
Collapse
Affiliation(s)
- Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago 8380453, Chile.
| | - Andrea Marimán
- Laboratorio de Fisiología y Bioenergética Celular, Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
| | - Bastián Ramos
- Laboratorio de Fisiología y Bioenergética Celular, Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
| | - María José Silva
- Laboratorio de Fisiología y Bioenergética Celular, Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
| | - Andrea Del Campo
- Laboratorio de Fisiología y Bioenergética Celular, Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile.
| |
Collapse
|
36
|
Ye P, Wu H, Jiang Y, Xiao X, Song D, Xu N, Ma X, Zeng J, Guo Y. Old dog, new tricks: Polydatin as a multitarget agent for current diseases. Phytother Res 2021; 36:214-230. [PMID: 34936712 DOI: 10.1002/ptr.7306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/19/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022]
Abstract
Polydatin (PD) is a natural single-crystal product that is primarily extracted from the traditional plant Polygonum cuspidatum Sieb. et Zucc. Early research showed that PD exhibited a variety of biological activities. PD has attracted increasing research interest since 2014, but no review comprehensively summarized the new findings. A great gap between its biological activities and drug development remains. It is necessary to summarize new findings on the pharmacological effects of PD on current diseases. We propose that PD will most likely be used in cardiac and cerebral ischaemia/reperfusion-related diseases and atherosclerosis in the future. The present work classified these new findings according to diseases and summarized the main effects of PD via specific mechanisms of action. In summary, we found that PD played a therapeutic role in a variety of diseases, primarily via five mechanisms: antioxidative effects, antiinflammatory effects, regulation of autophagy and apoptosis, maintenance of mitochondrial function, and lipid regulation.
Collapse
Affiliation(s)
- Penghui Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hefei Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yinxiao Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolin Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nuo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaoguang Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
37
|
Dong F, Pu Y, Lv Y, Liu X, Cao Y. Protective effect of Pulsatilla saponin A on acute myocardial infarction via miR-24-3p/p16. Toxicol Mech Methods 2021; 32:27-36. [PMID: 34412561 DOI: 10.1080/15376516.2021.1963364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The effect of Pulsatilla saponin A (PsA) on acute myocardial infarction (AMI) was unknown. This study targeted to examine the roles of PsA on hypoxia-triggered toxicity to H9c2 cells and reveal the potential mechanism. H9c2 cells were maintained under a hypoxic environment for 12 h to construct the AMI cell model and the cells were pretreated by PsA. Hypoxia triggered toxicity to H9c2 cells and the anti-toxicity effect of PsA was evaluated by CCK8, TUNEL, and Western blot. The levels of miR-24-3p and p16 in H9c2 cells, AMI group tissues, and their respective controls were assessed using qRT-PCR. The dual-luciferase assay was applied to verify the targeting mechanism of miR-24-3p on p16. Then the effects of miR-24-3p inhibitor or/and si-p16 on H9c2 cells treated with PsA under hypoxia were detected by CCK8, TUNEL, and Western blot. Flow cytometry was executed to determine the cell cycle. Hypoxia decreased viability and proliferation and increased apoptosis of H9c2 cells, which were ameliorated by PsA pretreatment. The level of miR-24-3p was diminished, but p16 expression was elevated in hypoxia-treated cells and AMI group tissues. MiR-24-3p could sponge p16 in hypoxia-treated cells. Furthermore, the impact of applying miR-24-3p inhibitor on PsA and hypoxia-treated cells could be reversed by si-p16. PsA relieved hypoxia-triggered cell toxicity via miR-24-3p/p16 axis. These findings provided some fresh insights into the potential therapeutic effects of the application of PsA in AMI.
Collapse
Affiliation(s)
- Feng Dong
- Department of General Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan City, China
| | - Yanhua Pu
- Department of General Family Medicine No.1, The Fourth Hospital of Jinan, Jinan City, China
| | - Yanfei Lv
- Department of Rehabilitation Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan City, China
| | - Xiujuan Liu
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan City, China
| | - Yimin Cao
- Department of Emergency, Jinan Traditional Chinese Medicine Hospital, Jinan City, China
| |
Collapse
|
38
|
Bai Y, Yang Y, Gao Y, Lin D, Wang Z, Ma J. Melatonin postconditioning ameliorates anoxia/reoxygenation injury by regulating mitophagy and mitochondrial dynamics in a SIRT3-dependent manner. Eur J Pharmacol 2021; 904:174157. [PMID: 33971181 DOI: 10.1016/j.ejphar.2021.174157] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/17/2021] [Accepted: 05/04/2021] [Indexed: 12/17/2022]
Abstract
Ischaemia/reperfusion (I/R) injury is accompanied by excessive mitochondrial autophagy (mitophagy) and an imbalance in mitochondrial dynamics. Melatonin has been reported to alleviate I/R injury by regulating mitophagy and mitochondrial dynamics. However, the underlying mechanism associated with this activity is not fully understood. The goal of the present study was to investigate whether and how melatonin administration at the beginning of reoxygenation exerts protective effects by regulating mitophagy and mitochondrial dynamics. H9c2 cells were transfected with sirtuin 3 (SIRT3)-targeting siRNA and then subjected to anoxia/reoxygenation (A/R) injury, with melatonin (150 μM) administered at the onset of reoxygenation. Biomarkers related to cellular apoptosis, oxidative stress, mitochondrial function, mitophagy and mitochondrial dynamics were assessed, and the expression and activity of SIRT3 was also measured. Mitochondrial fission and mitophagy were activated after A/R injury and were accompanied by cellular apoptosis, oxidative stress, and mitochondrial dysfunction. However, melatonin postconditioning inhibited excessive mitochondrial fission and mitophagy, promoted mitochondrial fusion, restored mitochondrial function and reduced cellular apoptosis, and the mitophagy inhibitor 3-methyladenine (3-MA) also attenuated A/R-induced apoptosis. Moreover, the A/R-induced decreases in SIRT3 and manganese superoxide dismutase (SOD2) activities were ameliorated by melatonin. However, SIRT3 silencing abolished the beneficial effects of melatonin, eliminated the inhibitory effects of melatonin on mitochondrial fission and mitophagy, and reversed the melatonin-induced increase in SOD2 activity. These results indicate that melatonin postconditioning protects H9c2 cells from A/R injury by inhibiting excessive mitophagy and maintaining the balance of mitochondrial fission and fusion in a SIRT3-dependent manner.
Collapse
Affiliation(s)
- Yang Bai
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Yanli Yang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Yafen Gao
- Department of Anesthesiology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Duomao Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Zhaoqi Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China.
| |
Collapse
|
39
|
Junior AG, de Almeida TL, Tolouei SEL, Dos Santos AF, Dos Reis Lívero FA. Predictive Value of Sirtuins in Acute Myocardial Infarction - Bridging the Bench to the Clinical Practice. Curr Pharm Des 2021; 27:206-216. [PMID: 33019924 DOI: 10.2174/1381612826666201005153848] [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] [Received: 03/29/2020] [Accepted: 08/09/2020] [Indexed: 11/22/2022]
Abstract
Acute myocardial infarction (AMI) is a non-transmissible condition with high prevalence, morbidity, and mortality. Different strategies for the management of AMI are employed worldwide, but its early diagnosis remains a major challenge. Many molecules have been proposed in recent years as predictive agents in the early detection of AMI, including troponin (C, T, and I), creatine kinase MB isoenzyme, myoglobin, heart-type fatty acid-binding protein, and a family of histone deacetylases with enzymatic activities named sirtuins. Sirtuins may be used as predictive or complementary treatment strategies and the results of recent preclinical studies are promising. However, human clinical trials and data are scarce, and many issues have been raised regarding the predictive values of sirtuins. The present review summarizes research on the predictive value of sirtuins in AMI. We also briefly summarize relevant clinical trials and discuss future perspectives and possible clinical applications.
Collapse
Affiliation(s)
- Arquimedes G Junior
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, MS, Brazil
| | - Thiago L de Almeida
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, MS, Brazil
| | - Sara E L Tolouei
- Laboratory of Reproductive Toxicology, Department of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - Andreia F Dos Santos
- Laboratory of Preclinical Research of Natural Products, Post-Graduate Program in Animal Science with Emphasis on Bioactive Products, Paranaense University, Umuarama, PR, Brazil
| | - Francislaine A Dos Reis Lívero
- Laboratory of Preclinical Research of Natural Products, Post-Graduate Program in Animal Science with Emphasis on Bioactive Products, Paranaense University, Umuarama, PR, Brazil
| |
Collapse
|
40
|
Soluble Epoxide Hydrolase in Aged Female Mice and Human Explanted Hearts Following Ischemic Injury. Int J Mol Sci 2021; 22:ijms22041691. [PMID: 33567578 PMCID: PMC7915306 DOI: 10.3390/ijms22041691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI) accounts for a significant proportion of death and morbidity in aged individuals. The risk for MI in females increases as they enter the peri-menopausal period, generally occurring in middle-age. Cytochrome (CYP) 450 metabolizes N-3 and N-6 polyunsaturated fatty acids (PUFA) into numerous lipid mediators, oxylipids, which are further metabolised by soluble epoxide hydrolase (sEH), reducing their activity. The objective of this study was to characterize oxylipid metabolism in the left ventricle (LV) following ischemic injury in females. Human LV specimens were procured from female patients with ischemic cardiomyopathy (ICM) or non-failing controls (NFC). Female C57BL6 (WT) and sEH null mice averaging 13–16 months old underwent permanent occlusion of the left anterior descending coronary artery (LAD) to induce myocardial infarction. WT (wild type) mice received vehicle or sEH inhibitor, trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (tAUCB), in their drinking water ad libitum for 28 days. Cardiac function was assessed using echocardiography and electrocardiogram. Protein expression was determined using immunoblotting, mitochondrial activity by spectrophotometry, and cardiac fibre respiration was measured using a Clark-type electrode. A full metabolite profile was determined by LC–MS/MS. sEH was significantly elevated in ischemic LV specimens from patients, associated with fundamental changes in oxylipid metabolite formation and significant decreases in mitochondrial enzymatic function. In mice, pre-treatment with tAUCB or genetic deletion of sEH significantly improved survival, preserved cardiac function, and maintained mitochondrial quality following MI in female mice. These data indicate that sEH may be a relevant pharmacologic target for women with MI. Although future studies are needed to determine the mechanisms, in this pilot study we suggest targeting sEH may be an effective strategy for reducing ischemic injury and mortality in middle-aged females.
Collapse
|
41
|
Chen J, Chen S, Zhang B, Liu J. SIRT3 as a potential therapeutic target for heart failure. Pharmacol Res 2021; 165:105432. [PMID: 33508434 DOI: 10.1016/j.phrs.2021.105432] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/12/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
Heart failure causes significant morbidity and mortality worldwide. The underlying mechanisms and pathological changes associated with heart failure are exceptionally complex. Despite recent advances in heart failure research, treatment outcomes remain poor. The sirtuin family member sirtuin-3 (SIRT3) is involved in several key biological processes, including ATP production, catabolism, and reactive oxygen species detoxification. In addition to its role in metabolism, SIRT3 regulates cell death and survival and has been implicated in the pathogenesis of cardiovascular diseases. Emerging evidence also shows that SIRT3 can protect cardiomyocytes from hypertrophy, ischemia-reperfusion injury, cardiac fibrosis, and impaired angiogenesis. In this review article, we summarize the recent advances in SIRT3 research and discuss the role of SIRT3 in heart failure. We also discuss the potential use of SIRT3 as a therapeutic target in heart failure.
Collapse
Affiliation(s)
- Jie Chen
- Cardiovascular Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, 430071, People's Republic of China
| | - Shiqi Chen
- Cardiovascular Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, 430071, People's Republic of China
| | - Bingxia Zhang
- Cardiovascular Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, 430071, People's Republic of China
| | - Junwei Liu
- Cardiovascular Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, 430071, People's Republic of China.
| |
Collapse
|
42
|
Lee C, Hong WJ, Jung KH, Hong HC, Kim DY, Ok HC, Choi MS, Park SK, Kim J, Koh HJ. Arachis hypogaea resveratrol synthase 3 alters the expression pattern of UDP-glycosyltransferase genes in developing rice seeds. PLoS One 2021; 16:e0245446. [PMID: 33444365 PMCID: PMC7808588 DOI: 10.1371/journal.pone.0245446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/31/2020] [Indexed: 12/19/2022] Open
Abstract
The resveratrol-producing rice (Oryza sativa L.) inbred lines, Iksan 515 (I.515) and Iksan 526 (I.526), developed by the expression of the groundnut (Arachis hypogaea) resveratrol synthase 3 (AhRS3) gene in the japonica rice cultivar Dongjin, accumulated both resveratrol and its glucoside, piceid, in seeds. Here, we investigated the effect of the AhRS3 transgene on the expression of endogenous piceid biosynthesis genes (UGTs) in the developing seeds of the resveratrol-producing rice inbred lines. Ultra-performance liquid chromatography (UPLC) analysis revealed that I.526 accumulates significantly higher resveratrol and piceid in seeds than those in I.515 seeds and, in I.526 seeds, the biosynthesis of resveratrol and piceid reached peak levels at 41 days after heading (DAH) and 20 DAH, respectively. Furthermore, RNA-seq analysis showed that the expression patterns of UGT genes differed significantly between the 20 DAH seeds of I.526 and those of Dongjin. Quantitative real-time PCR (RT-qPCR) analyses confirmed the data from RNA-seq analysis in seeds of Dongjin, I.515 and I.526, respectively, at 9 DAH, and in seeds of Dongjin and I.526, respectively, at 20 DAH. A total of 245 UGTs, classified into 31 UGT families, showed differential expression between Dongjin and I.526 seeds at 20 DAH. Of these, 43 UGTs showed more than 2-fold higher expression in I.526 seeds than in Dongjin seeds. In addition, the expression of resveratrol biosynthesis genes (PAL, C4H and 4CL) was also differentially expressed between Dongjin and I.526 developing seeds. Collectively, these data suggest that AhRS3 altered the expression pattern of UGT genes, and PAL, C4H and 4CL in developing rice seeds.
Collapse
Affiliation(s)
- Choonseok Lee
- Department of Plant Science, Research Institute for Agriculture and Life Sciences, and Plant Genomics and Breeding Institute, Seoul National University, Seoul, Republic of Korea
| | - Woo-Jong Hong
- Graduate School of Biotechnology, Kyung Hee University, Yongin, Gyeonggi-do, Republic of Korea
| | - Ki-Hong Jung
- Graduate School of Biotechnology, Kyung Hee University, Yongin, Gyeonggi-do, Republic of Korea
| | - Ha-Cheol Hong
- National Institute of Agricultural Sciences, Wanju, Jeollabuk-do, Republic of Korea
| | - Dool-Yi Kim
- National Institute of Crop Science, Wanju, Jeollabuk-do, Republic of Korea
| | - Hyun-Choong Ok
- Rural Development Administration, Jeonju, Jeollabuk-do, Republic of Korea
| | - Man-Soo Choi
- National Institute of Crop Science, Wanju, Jeollabuk-do, Republic of Korea
| | - Soo-Kwon Park
- Rural Development Administration, Jeonju, Jeollabuk-do, Republic of Korea
| | - Jaehyun Kim
- National Institute of Crop Science, Wanju, Jeollabuk-do, Republic of Korea
- * E-mail: (JK); (HJK)
| | - Hee-Jong Koh
- Department of Plant Science, Research Institute for Agriculture and Life Sciences, and Plant Genomics and Breeding Institute, Seoul National University, Seoul, Republic of Korea
- * E-mail: (JK); (HJK)
| |
Collapse
|
43
|
Gong N, Wang X, Wang Y, Yang S, Song J, Lu Y, Du G. Control over Polymorph Formation of Polydatin in Binary Solvent System and Structural Characterization. J Pharm Biomed Anal 2020; 190:113260. [PMID: 32846398 DOI: 10.1016/j.jpba.2020.113260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/05/2020] [Accepted: 03/14/2020] [Indexed: 11/26/2022]
Abstract
Polydatin is a natural product used for anti-oxidant, anti-inflammatory and anti-tumor purposes, and often added in medicine, nutraceutical, cosmetics, and dietary supplement. Polymorphism is a key feature of solid-state pharmaceutical products. Polymorphic modifications may exhibit different physical and chemical properties. Here we report two different polymorphs, and the amorphous form of Polydatin. Polymorphs were prepared in binary solvent system. The crystal structures of the two forms were revealed for the first time. The structure and 3D packing were determined with single crystal X-ray diffraction analysis. The batch consistency and stability were identified with Powder X-ray diffraction analysis. Various functional groups present in the polymorphs were analyzed with fourier transform infrared spectroscopic method. The thermal properties were investigated with DSC and TGA. HPLC-MS was used for the pharmacokinetic study. Results show that form B has the faster absorption, and can be maintained in animal bodies for a longer time than form A.
Collapse
Affiliation(s)
- Ningbo Gong
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Xue Wang
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ying Wang
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shiying Yang
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Junke Song
- Beijing City Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College. Beijing, 100050, China
| | - Yang Lu
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Guanhua Du
- Beijing City Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College. Beijing, 100050, China.
| |
Collapse
|
44
|
Protective Effect of Polydatin on Jejunal Mucosal Integrity, Redox Status, Inflammatory Response, and Mitochondrial Function in Intrauterine Growth-Retarded Weanling Piglets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7178123. [PMID: 33101591 PMCID: PMC7576365 DOI: 10.1155/2020/7178123] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/23/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022]
Abstract
Intrauterine growth retardation (IUGR) delays the gut development of neonates, but effective treatment strategies are still limited. This study used newborn piglets as a model to evaluate the protective effect of polydatin (PD) against IUGR-induced intestinal injury. In total, 36 IUGR piglets and an equal number of normal birth weight (NBW) littermates were fed either a basal diet or a PD-supplemented diet from 21 to 35 days of age. Compared with NBW, IUGR induced jejunal damage and barrier dysfunction of piglets, as indicated by observable bacterial translocation, enhanced apoptosis, oxidative and immunological damage, and mitochondrial dysfunction. PD treatment decreased bacterial translocation and inhibited the IUGR-induced increases in circulating diamine oxidase activity (P = 0.039) and D-lactate content (P = 0.004). The apoptotic rate (P = 0.024) was reduced by 35.2% in the PD-treated piglets, along with increases in villus height (P = 0.033) and in ratio of villus height to crypt depth (P = 0.049). PD treatment promoted superoxide dismutase (P = 0.026) and glutathione S-transferase activities (P = 0.006) and reduced malondialdehyde (P = 0.015) and 8-hydroxy-2′-deoxyguanosine accumulation (P = 0.034) in the jejunum. The PD-treated IUGR piglets showed decreased jejunal myeloperoxidase activity (P = 0.029) and tumor necrosis factor alpha content (P = 0.035) than those received a basal diet. PD stimulated nuclear sirtuin 1 (P = 0.028) and mitochondrial citrate synthase activities (P = 0.020) and facilitated adenosine triphosphate production (P = 0.009) in the jejunum of piglets. Furthermore, PD reversed the IUGR-induced declines in mitochondrial DNA content (P = 0.048), the phosphorylation of adenosine monophosphate-activated protein kinase alpha (P = 0.027), and proliferation-activated receptor gamma coactivator 1 alpha expression (P = 0.033). Altogether, the results indicate that PD may improve jejunal integrity, mitigate mucosal oxidative and immunological damage, and facilitate mitochondrial function in IUGR piglets.
Collapse
|
45
|
Wu X, Liu Z, Yu XY, Xu S, Luo J. Autophagy and cardiac diseases: Therapeutic potential of natural products. Med Res Rev 2020; 41:314-341. [PMID: 32969064 DOI: 10.1002/med.21733] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 08/28/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022]
Abstract
The global incidence of cardiac diseases is expected to increase in the coming years, imposing a substantial socioeconomic burden on healthcare systems. Autophagy is a tightly regulated lysosomal degradation mechanism important for cell survival, homeostasis, and function. Accumulating pieces of evidence have indicated a major role of autophagy in the regulation of cardiac homeostasis and function. It is well established that dysregulation of autophagy in cardiomyocytes is involved in cardiac hypertrophy, myocardial infarction, diabetic cardiomyopathy, and heart failure. In this sense, autophagy seems to be an attractive therapeutic target for cardiac diseases. Recently, multiple natural products/phytochemicals, such as resveratrol, berberine, and curcumin have been shown to regulate cardiomyocyte autophagy via different pathways. The autophagy-modifying capacity of these compounds should be taken into consideration for designing novel therapeutic agents. This review focuses on the role of autophagy in various cardiac diseases and the pharmacological basis and therapeutic potential of reported natural products in cardiac diseases by modifying autophagic processes.
Collapse
Affiliation(s)
- Xiaoqian Wu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zumei Liu
- Department of Central Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Suowen Xu
- Department of Endocrinology and Metabolism, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Jiandong Luo
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
46
|
Zhang S, Wang S, Shi X, Feng X. Polydatin alleviates parkinsonism in MPTP-model mice by enhancing glycolysis in dopaminergic neurons. Neurochem Int 2020; 139:104815. [PMID: 32758587 DOI: 10.1016/j.neuint.2020.104815] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/30/2020] [Accepted: 07/16/2020] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease. Damage to energy metabolism and reduced adenosine triphosphate (ATP) levels in dopaminergic neurons are common features of PD. Previous studies suggested that the occurrence of PD often affects glucose metabolism and ATP production in the brain, and increased glycolysis or ATP production protects dopaminergic neuronal degeneration in the brain of PD patients. These systems may provide new potential therapeutic targets for the prevention of PD. The present study investigated the inhibitory action of polydatin (PLD) on early dopaminergic neuronal degeneration induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The results showed that PLD protected against MPTP-induced early dopaminergic neuronal degeneration. PLD reduced the MPTP-induced loss of dopaminergic neurons in substantia nigra and striatum, inhibited the occurrence of neural apoptosis, and restored motor function in mice. PLD also increased the continuous activity duration and rhythm amplitude in mice during the circadian activity test. PLD improved glucose metabolism in the brain and restored ATP production levels. These observations suggest that PLD attenuates MPTP-induced early PD-like symptoms, and its mechanism of action may be associated with the promotion of glucose metabolism in neurons.
Collapse
Affiliation(s)
- Shaozhi Zhang
- College of Life Science, The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Sijie Wang
- College of Life Science, The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xingzhu Shi
- College of Life Sciences, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Xizeng Feng
- College of Life Science, The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
47
|
Cortés-Rojo C, Vargas-Vargas MA, Olmos-Orizaba BE, Rodríguez-Orozco AR, Calderón-Cortés E. Interplay between NADH oxidation by complex I, glutathione redox state and sirtuin-3, and its role in the development of insulin resistance. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165801. [PMID: 32305451 DOI: 10.1016/j.bbadis.2020.165801] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/16/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022]
Abstract
Metabolic diseases are characterized by high NADH/NAD+ ratios due to excessive electron supply, causing defective mitochondrial function and impaired sirtuin-3 (SIRT-3) activity, the latter driving to oxidative stress and altered fatty acid β-oxidation. NADH is oxidized by the complex I in the electron transport chain, thereby factors inhibiting complex I like acetylation, cardiolipin peroxidation, and glutathionylation by low GSH/GSSG ratios affects SIRT3 function by increasing the NADH/NAD+ ratio. In this review, we summarized the evidence supporting a role of the above events in the development of insulin resistance, which is relevant in the pathogenesis of obesity and diabetes. We propose that maintenance of proper NADH/NAD+ and GSH/GSSG ratios are central to ameliorate insulin resistance, as alterations in these redox couples lead to complex I dysfunction, disruption of SIRT-3 activity, ROS production and impaired β-oxidation, the latter two being key effectors of insulin resistance.
Collapse
Affiliation(s)
- Christian Cortés-Rojo
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mich 58030, México.
| | - Manuel Alejandro Vargas-Vargas
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mich 58030, México
| | - Berenice Eridani Olmos-Orizaba
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mich 58030, México
| | - Alain Raimundo Rodríguez-Orozco
- Facultad de Ciencias Médicas y Biológicas "Dr. Ignacio Chávez", Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mich 58020, México
| | - Elizabeth Calderón-Cortés
- Facultad de Enfermería, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mich 58260, México
| |
Collapse
|
48
|
Zhang J, Xiang H, Liu J, Chen Y, He RR, Liu B. Mitochondrial Sirtuin 3: New emerging biological function and therapeutic target. Theranostics 2020; 10:8315-8342. [PMID: 32724473 PMCID: PMC7381741 DOI: 10.7150/thno.45922] [Citation(s) in RCA: 282] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/08/2020] [Indexed: 02/05/2023] Open
Abstract
Sirtuin 3 (SIRT3) is one of the most prominent deacetylases that can regulate acetylation levels in mitochondria, which are essential for eukaryotic life and inextricably linked to the metabolism of multiple organs. Hitherto, SIRT3 has been substantiated to be involved in almost all aspects of mitochondrial metabolism and homeostasis, protecting mitochondria from a variety of damage. Accumulating evidence has recently documented that SIRT3 is associated with many types of human diseases, including age-related diseases, cancer, heart disease and metabolic diseases, indicating that SIRT3 can be a potential therapeutic target. Here we focus on summarizing the intricate mechanisms of SIRT3 in human diseases, and recent notable advances in the field of small-molecule activators or inhibitors targeting SIRT3 as well as their potential therapeutic applications for future drug discovery.
Collapse
|
49
|
Wu J, Yang Y, Gao Y, Wang Z, Ma J. Melatonin Attenuates Anoxia/Reoxygenation Injury by Inhibiting Excessive Mitophagy Through the MT2/SIRT3/FoxO3a Signaling Pathway in H9c2 Cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2047-2060. [PMID: 32546969 PMCID: PMC7260543 DOI: 10.2147/dddt.s248628] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
Purpose Autophagy caused by ischemia/reperfusion (I/R) increases the extent of cardiomyocyte damage. Melatonin (Mel) diminishes cardiac injury through regulating autophagy and mitochondrial dynamics. However, illustrating the specific role of mitophagy in the cardioprotective effects of melatonin remains a challenge. The aim of our research was to investigate the impact and underlying mechanisms of melatonin in connection with mitophagy during anoxia/reoxygenation (A/R) injury in H9c2 cells. Methods H9c2 cells were pretreated with melatonin with or without the melatonin membrane receptor 2 (MT2) antagonist 4-P-PDOT, the MT2 agonist IIK7 and the sirtuin 3 (SIRT3) inhibitor 3-TYP for 4 hours and then subjected to A/R injury. Cell viability, cellular apoptosis, necrosis levels and oxidative markers were assessed. The expression of SIRT3 and forkhead box O3a (FoxO3a), mitochondrial function and the levels of mitophagy-related proteins were also evaluated. Results A/R injury provoked enhanced mitophagy in H9c2 myocytes. In addition, increased mitophagy was correlated with decreased cellular viability, increased oxidative stress and mitochondrial dysfunction in H9c2 cells. However, melatonin pretreatment notably increased cell survival and decreased cell apoptosis and oxidative response after A/R injury, accompanied by restored mitochondrial function. The inhibition of excessive mitophagy is involved in the cardioprotective effects of melatonin, as shown by the decreased expression of the mitophagy-related molecules Parkin, Beclin1, and BCL2-interacting protein 3-like (BNIP3L, best known as NIX) and decreased light chain 3 II/light chain 3 I (LC3 II/LC3 I) ratio and upregulation of p62 expression. Moreover, the decreased expression of SIRT3 and FoxO3a in A/R-injured H9c2 cells was abrogated by melatonin, but these beneficial effects were attenuated by the MT2 antagonist 4-P-PDOT or the SIRT3 inhibitor 3-TYP and enhanced by the MT2 agonist IIK7. Conclusion These results indicate that melatonin protects H9c2 cells during A/R injury through suppressing excessive mitophagy by activating the MT2/SIRT3/FoxO3a pathway. Melatonin may be a useful candidate for alleviating myocardial ischemia/reperfusion (MI/R) injury in the future, and the MT2 receptor might become a therapeutic target.
Collapse
Affiliation(s)
- Jinjing Wu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Yanli Yang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Yafen Gao
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Zhaoqi Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, People's Republic of China
| |
Collapse
|
50
|
Dai J, Ma J, Liao Y, Luo X, Chen G. Polydatin protects H9c2 cells from hypoxia-induced injury via up-regulating long non-coding RNA DGCR5. ACTA ACUST UNITED AC 2019; 52:e8834. [PMID: 31826181 PMCID: PMC6903803 DOI: 10.1590/1414-431x20198834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
Polydatin (PD), a monocrystalline polyphenolic drug mainly found in the roots of Polygonum cuspidatum, has various pharmacological activities. Long non-coding RNAs (lncRNA) DiGeorge syndrome critical region gene 5 (DGCR5) was found to participate in the suppression of multiple cancers. Here, we proposed to study the effect of PD on myocardial infarction (MI) by inducing DGCR5. CCK-8 assay was performed to detect the viability of H9c2 cells. Flow cytometry was utilized to test apoptosis of H9c2 cells. These results determined the optimal concentration and effect time of hypoxia as well as PD. Si-DGCR5 was transfected into cells and the expression level was determined by qRT-PCR. Western blot was utilized to evaluate the expression of apoptosis-related proteins, Bcl-2, Bax, and cleaved-caspase-3, as well as autophagy-associated proteins including Beclin-1, p62, and LC3-II/LC3-I. As a result, PD efficiently attenuated hypoxia-induced apoptosis and autophagy in H9c2 cells. The expression of DGCR5 was down-regulated by hypoxia and up-regulated by PD. Besides, knocking-down the expression of DGCR5 inhibited the protection of PD in H9c2 cells. In addition, PD up-regulated the accumulation of DGCR5, DGCR5 decreased the expression of Bcl-2 and p62, raised the expression of Bax and cleaved-caspase-3, and the proportion of LC3-II/LC3-I. PD stimulated the PI3K/AKT/mTOR and MEK/ERK signaling pathways via up-regulating the expression of DGCR5. Our data demonstrated that PD reduced cell apoptosis and autophagy induced by hypoxia in cardiomyocytes. Moreover, PD activated PI3K/AKT/mTOR and MEK/ERK signaling pathways by up-regulating the expression of DGCR5.
Collapse
Affiliation(s)
- Jinhua Dai
- Department of Clinical Laboratory, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, Zhejiang, China
| | - Jianbo Ma
- Department of Clinical Laboratory, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, Zhejiang, China
| | - Yufeng Liao
- Department of Clinical Laboratory, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, Zhejiang, China
| | - Xianhai Luo
- Department of Clinical Laboratory, Ningbo Kangning Hospital, Ningbo Mental Health Center, Ningbo, Zhejiang, China
| | - Guofang Chen
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, Zhejiang, China
| |
Collapse
|