1
|
Zheng S, Yang L, Dai Q, Li X, Masuoka T, Lv J. Role of sirtuin 1 in depression‑induced coronary heart disease: Molecular pathways and therapeutic potential (Review). Biomed Rep 2025; 22:46. [PMID: 39882335 PMCID: PMC11775641 DOI: 10.3892/br.2025.1924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Depression and coronary heart disease (CHD) are two interconnected diseases that profoundly impact global health. Depression is both a complex psychiatric disorder and an established risk factor for CHD. Sirtuin 1 (SIRT1) is an enzyme that requires the cofactor nicotinamide adenine dinucleotide (NAD+) to perform its deacetylation function, and its involvement is crucial in reducing cardiovascular risks that are associated with depression. SIRT1 exerts its cardioprotective effects via modulating oxidative stress, inflammation and metabolic processes, all of which are central to the pathogenesis of CHD in individuals with depression. Through influencing these pathways, SIRT1 helps to reduce endothelial dysfunction, prevent the formation of atherosclerotic plaques and stabilize existing plaques, thereby decreasing the overall risk of CHD. The present review underscores the important role of SIRT1 in serving as a therapeutic intervention molecule for tackling cardiovascular complications stemming from depression. Furthermore, it highlights the need for further studies to clarify how SIRT1 influences both depression and CHD at the molecular level. The ultimate goal of this research will be to translate these findings into practical clinical intervention strategies.
Collapse
Affiliation(s)
- Shijie Zheng
- Department of Cardiology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei 443001, P.R. China
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Linlin Yang
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei 443001, P.R. China
| | - Qiuting Dai
- Department of Cardiology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei 443001, P.R. China
| | - Xiangyan Li
- Department of Cardiology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei 443001, P.R. China
| | - Takayoshi Masuoka
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Jianfeng Lv
- Department of Cardiology, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei 443001, P.R. China
| |
Collapse
|
2
|
Chen H, Weng Z, Kalinowska M, Xiong L, Wang L, Song H, Xiao J, Wang F, Shen X. Anti-osteoporosis effect of bioactives in edible medicinal plants: a comprehensive review. Crit Rev Food Sci Nutr 2024:1-17. [PMID: 39093554 DOI: 10.1080/10408398.2024.2386449] [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: 08/04/2024]
Abstract
Current treatments for osteoporosis include a calcium-rich diet, adequate exercise, and medication. Many synthetic drugs, although fast-acting, can cause a range of side effects for patients when taken over a long period, such as irritation of the digestive tract and a burden on the kidneys. As the world's population ages, the prevalence of osteoporosis is increasing, and the development of safe and effective treatments is urgently needed. Active compounds in edible and medicinal homologous plants have been used for centuries to improve bone quality. It is possible to employ them as dietary supplements to prevent osteoporosis. In this review, we analyze the influencing factors of osteoporosis and systematically summarize the research progress on the anti-osteoporosis effects of active compounds in edible and medicinal homologous plants. The literature suggests that some naturally occurring active compounds in edible and medicinal homologous plants can inhibit bone loss, prevent the degeneration of bone cell microstructure, and reduce bone fragility through alleviating oxidative stress, regulating autophagy, anti-inflammation, improving gut flora, and regulating estrogen level with little side effects. Our review provides useful guidance for the use of edible and medicinal homologous plants and the development of safer novel anti-osteoporosis dietary supplements.
Collapse
Affiliation(s)
- Huiling Chen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Zebin Weng
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Monika Kalinowska
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Bialystok, Poland
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Jianbo Xiao
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI, Universidade de Vigo, Nutrition and Bromatology Group, Vigo, Spain
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
3
|
Nachimuthu L, Desikan R. Development of methyl 5-((cinnamoyloxy)methyl)picolinate, exploring its bio-target potential aiming at CVD mediated by multiple proteins through surface and physiochemical analysis. Sci Rep 2024; 14:13328. [PMID: 38858562 PMCID: PMC11164877 DOI: 10.1038/s41598-024-64165-7] [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: 03/03/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
The emphasis on sustainable sources of drug development seems imminent with phytochemicals emerging as promising candidates due to their minimal probability of adverse effects. This study focuses on utilizing simple cinnamic acid and nicotinic acid derivatives as starting materials, employing an efficient synthetic protocol to obtain methyl 5-((cinnamoyloxy)methyl)picolinate targeting CVD mediated by multiple enzymes such as MAPK, PCSK9, MPO, SIRT1 and TNF-α. Comprehensive characterization of synthesized molecule is achieved through 1H, 13C, FT-IR, and HRMS methods. Additionally, the crystal structure was established via SC-XRD. Comparative analysis with the DFT-optimized structure identifies key nucleophilic and electrophilic regions for determining interactions with bio-targets. Notably, Compound 5 adheres to all drug-likeness criteria, further validated through screening similar pharmacophoric drugs from databases. Targeting bio-relevant areas with a specific focus on CVD drug development. The molecular docking studies elucidate ligand-protein interactions for better binding connectivity. This investigation further underscores the importance of sustainable practices, simple chemical synthesis, and computational approaches, contributing to the pursuit of eco-friendly drug development with enhanced safety profiles (MTT assay).
Collapse
Affiliation(s)
- Lenin Nachimuthu
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, 632014, Tamilnadu, India
| | - Rajagopal Desikan
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore, 632014, Tamilnadu, India.
| |
Collapse
|
4
|
Cong X, Zhu X, Zhang X, Ning Z. Astragaloside IV inhibits angiotensin II-induced atrial fibrosis and atrial fibrillation by SIRT1/PGC-1α/FNDC5 pathway. Heliyon 2024; 10:e30984. [PMID: 38803993 PMCID: PMC11128467 DOI: 10.1016/j.heliyon.2024.e30984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
Aims and objectives Astragaloside IV (AS-IV) has been found to possess anti-oxidative, anti-inflammatory, and anti-apoptotic properties, but its effect on atrial fibrosis is yet to be determined. This research investigates the protective role of AS-IV in angiotensin II (Ang II)-induced atrial fibrosis and atrial fibrillation (AF). Methods C57BL/6 male mice aged 8-10 weeks (n = 40) were subcutaneously administered Ang II (2.0 mg/kg/day) or saline, with AS-IV (80 mg/kg) intraperitoneally administered 2 h before Ang II infusion for 4 weeks. Biochemical, histological, and morphological analyses were carried out. Using transesophageal burst pacing, AF was generated in vivo. Results Here, we report that AS-IV treatment inhibited Ang II-induced AF development in mice (58 ± 5.86 vs 15.13 ± 2.16 %, p < 0.001). Ang II + AS-IV therapy was effective in reducing the atrial fibrotic area and decreasing the increase in smooth muscle alpha-actin (α-SMA)-positive myofibroblasts brought on by Ang II treatment (fibrotic area: 26.25 ± 3.81 vs 8.62 ± 1.83 %, p < 0.001 and α-SMA: 65.62 ± 10.63 vs 17.25 ± 1.78 %, p < 0.001). The reactive oxygen species (ROS) production was reduced by pretreatment with Ang II + AS-IV (9.20 ± 0.92 vs 2.63 ± 0.22 %/sec, p < 0.001). In addition, Ang II + AS-IV treatment suppressed oxidative stress in Ang II-induced atrial fibrosis (malondialdehyde: 701.78 ± 85.01 vs 504.07 ± 25.62 pmol/mg protein, p < 0.001; superoxide dismutase: 13.82 ± 1.25 vs 29.54 ± 2.45 U/mg protein, p < 0.001 and catalase: 11.43 ± 1.19 vs 20.83 ± 3.29 U/mg protein, p < 0.001, respectively). Moreover, Ang II + AS-IV decreased the expression of α-SMA, collagen III and collagen I (3.32 ± 0.53 vs 1.41 ± 0.20 fold, p < 0.001; 3.41 ± 0.55 vs 1.48 ± 0.18 fold, p < 0.001; 2.34 ± 0.55 vs 0.99 ± 0.17 fold, p < 0.001, respectively) while increasing the protein expression of sirtuin 1 (SIRT1), peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α), and fibronectin type III domain-containing protein 5 (FNDC5) in Ang II-treated mice (0.22 ± 0.02 vs 0.57 ± 0.08 fold, p < 0.001; 0.28 ± 0.04 vs 0.72 ± 0.05 fold, p < 0.001; 0.38 ± 0.03 vs 0.68 ± 0.06 fold, p < 0.001, respectively). Conclusion Our data led us to speculate that AS-IV may protect against Ang II-induced atrial fibrosis and AF via upregulation of the SIRT1/PGC-1α/FNDC5 pathway.
Collapse
Affiliation(s)
- Xinpeng Cong
- Department of Cardiology, Shanghai Pudong New Area Zhoupu Hospital (Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital), No.1500 Zhou Yuan Road, Pudong New District, Shanghai, 201318, China
| | - Xi Zhu
- Department of Cardiology, Shanghai Pudong New Area Zhoupu Hospital (Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital), No.1500 Zhou Yuan Road, Pudong New District, Shanghai, 201318, China
| | - Xiaogang Zhang
- Department of Cardiology, Shanghai Pudong New Area Zhoupu Hospital (Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital), No.1500 Zhou Yuan Road, Pudong New District, Shanghai, 201318, China
| | - Zhongping Ning
- Department of Cardiology, Shanghai Pudong New Area Zhoupu Hospital (Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital), No.1500 Zhou Yuan Road, Pudong New District, Shanghai, 201318, China
| |
Collapse
|
5
|
Rabinovich-Nikitin I, Kirshenbaum LA. Circadian regulated control of myocardial ischemia-reperfusion injury. Trends Cardiovasc Med 2024; 34:1-7. [PMID: 36150629 DOI: 10.1016/j.tcm.2022.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 01/14/2023]
Abstract
Circadian mechanisms have been associated with the pathogenesis of a variety of cardiovascular diseases, including myocardial ischemia-reperfusion injury (I-R). Myocardial ischemia resulting from impaired oxygen delivery to cardiac muscle sets into motion a cascade of cellular events that paradoxically triggers greater cardiac dysfunction upon reinstitution of coronary blood supply, a phenomenon known as I-R. I-R injury has been attributed to a number of cellular defects including increased reactive oxygen species (ROS), increased intracellular calcium and impaired mitochondrial bioenergetics that ultimately lead to cardiac cell death, ventricular remodeling and heart failure. Emerging evidence has identified a strong correlation between cellular defects that underlie I-R and the disrupted circadian. In fact, recent studies have shown that circadian dysfunction exacerbates cardiac injury following MI from impaired cellular quality control mechanisms such as autophagy, which are vital in the clearance of damaged cellular proteins and organelles such as mitochondria from the cell. The accumulation of cellular debris is posited as the central underlying cause of excessive cardiac cell death and ventricular dysfunction following MI. The complexities that govern the interplay between circadian biology and I-R injury following MI is at its infancy and understanding how circadian misalignment, such as in shift workers impacts I-R injury is of great scientific and clinical importance toward development of new therapeutic strategies using chronotherapy and circadian regulation to mitigate cardiac injury and improve cardiac outcomes after injury. In this review, we highlight recent advances in circadian biology and adaptive cellular quality control mechanisms that influence cardiac injury in response to MI injury with a specific focus on how circadian biology can be utilized to further cardiovascular medicine and patient care.
Collapse
Affiliation(s)
- Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Canada
| | - Lorrie A Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Canada; Department of Pharmacology and Therapeutics Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba R2H 2H6, Canada.
| |
Collapse
|
6
|
Hao Y, Li B, Huber SA, Liu W. Bibliometric analysis of trends in cardiac aging research over the past 20 years. Medicine (Baltimore) 2023; 102:e34870. [PMID: 37653740 PMCID: PMC10470686 DOI: 10.1097/md.0000000000034870] [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: 07/04/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND In recent years, many studies have addressed cardiac aging and related diseases. This study aims to understand the research trend of cardiac aging and find new hot issues. METHODS We searched the web of science core collection database for articles published between 2003 and 2022 on the topic of "cardiac aging." Complete information including keywords, publication year, journal title, country, organization, and author were extracted for analysis. The VOS viewer software was used to generate network maps of keywords, countries, institutions, and author relationships for visual network analysis. RESULTS A total of 1002 papers were analyzed in the study. Overall, the number of annual publications on cardiac aging has increased since 2009, and new hot topics are emerging. The top 3 countries with the most publications were the United States (471 articles), China (209 articles) and Italy (101 articles). The University of Washington published the most papers (35 articles). The cluster analysis with author as the keyword found that the connections among different scholars are scattered and clustered in a small range. Network analysis based on keyword co-occurrence and year of publication identified relevant features and trends in cardiac aging research. According to the results of cluster analysis, all the articles are divided into 4 topics: "mechanisms of cardiac aging", "prevention and treatment of cardiac aging", "characteristics of cardiac aging", and "others." In recent years, the mechanism and treatment of cardiac aging have attracted the most attention. In both studies, animal models are used more often than in human populations. Mitochondrial dysfunction, autophagy and mitochondrial autophagy are hotspots in current research. CONCLUSION In this study, bibliometric analysis was used to analyze the research trend of cardiac aging in the past 20 years. The mechanism and treatment of cardiac aging are the most concerned contents. Mitochondrial dysfunction, autophagy and mitophagy are the focus of future research on cardiac aging.
Collapse
Affiliation(s)
- Yan Hao
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Harbin Medical University, Harbin, Heilongjiang, China
| | - Bohan Li
- Harbin Medical University, Harbin, Heilongjiang, China
| | - Sally A. Huber
- Department of Pathology and Laboratory Medicine, University of Vermont, Colchester, VT
| | - Wei Liu
- Harbin Medical University, Harbin, Heilongjiang, China
- Department of Geriatric Cardiovascular Division, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
7
|
Truong Thuy Nguyen V, Taheri N, Choi EL, Kellogg TA, Linden DR, Hayashi Y. Insulin-Like Growth Factor1 Preserves Gastric Pacemaker Cells and Motor Function in Aging via ERK1/2 Activation. Cell Mol Gastroenterol Hepatol 2023; 16:369-383. [PMID: 37301443 PMCID: PMC10372898 DOI: 10.1016/j.jcmgh.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND & AIMS Impaired gastric motor function in the elderly causes reduced food intake leading to frailty and sarcopenia. We previously found that aging-related impaired gastric compliance was mainly owing to depletion of interstitial cells of Cajal (ICC), pacemaker cells, and neuromodulator cells. These changes were associated with reduced food intake. Transformation-related protein 53-induced suppression of extracellular signal-regulated protein kinase (ERK)1/2 in ICC stem cell (ICC-SC) cell-cycle arrest is a key process for ICC depletion and gastric dysfunction during aging. Here, we investigated whether insulin-like growth factor 1 (IGF1), which can activate ERK in gastric smooth muscles and invariably is reduced with age, could mitigate ICC-SC/ICC loss and gastric dysfunction in klotho mice, a model of accelerated aging. METHODS Klotho mice were treated with the stable IGF1 analog LONG R3 recombinant human (rh) IGF1 (150 μg/kg intraperitoneally twice daily for 3 weeks). Gastric ICC/ICC-SC and signaling pathways were studied by flow cytometry, Western blot, and immunohistochemistry. Gastric compliance was assessed in ex vivo systems. Transformation-related protein 53 was induced with nutlin 3a and ERK1/2 signaling was activated by rhIGF-1 in the ICC-SC line. RESULTS LONG R3 rhIGF1 treatment prevented reduced ERK1/2 phosphorylation and gastric ICC/ICC-SC decrease. LONG R3 rhIGF1 also mitigated the reduced food intake and impaired body weight gain. Improved gastric function by LONG R3 rhIGF1 was verified by in vivo systems. In ICC-SC cultures, rhIGF1 mitigated nutlin 3a-induced reduced ERK1/2 phosphorylation and cell growth arrest. CONCLUSIONS IGF1 can mitigate age-related ICC/ICC-SC loss by activating ERK1/2 signaling, leading to improved gastric compliance and increased food intake in klotho mice.
Collapse
Affiliation(s)
- Vy Truong Thuy Nguyen
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Negar Taheri
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Egan L Choi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Todd A Kellogg
- Department of Surgery, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - David R Linden
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Yujiro Hayashi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota.
| |
Collapse
|
8
|
Liu C, Wu X, Vulugundam G, Gokulnath P, Li G, Xiao J. Exercise Promotes Tissue Regeneration: Mechanisms Involved and Therapeutic Scope. SPORTS MEDICINE - OPEN 2023; 9:27. [PMID: 37149504 PMCID: PMC10164224 DOI: 10.1186/s40798-023-00573-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Exercise has well-recognized beneficial effects on the whole body. Previous studies suggest that exercise could promote tissue regeneration and repair in various organs. In this review, we have summarized the major effects of exercise on tissue regeneration primarily mediated by stem cells and progenitor cells in skeletal muscle, nervous system, and vascular system. The protective function of exercise-induced stem cell activation under pathological conditions and aging in different organs have also been discussed in detail. Moreover, we have described the primary molecular mechanisms involved in exercise-induced tissue regeneration, including the roles of growth factors, signaling pathways, oxidative stress, metabolic factors, and non-coding RNAs. We have also summarized therapeutic approaches that target crucial signaling pathways and molecules responsible for exercise-induced tissue regeneration, such as IGF1, PI3K, and microRNAs. Collectively, the comprehensive understanding of exercise-induced tissue regeneration will facilitate the discovery of novel drug targets and therapeutic strategies.
Collapse
Affiliation(s)
- Chang Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Xinying Wu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China
| | | | - Priyanka Gokulnath
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
9
|
Yan L, Nong X, Deng J, Yang G. Testosterone protects cardiomyocytes against hydrogen peroxide-induced aging by upregulating IGF1 and SIRT1 pathways. Physiol Int 2022. [PMID: 36001411 DOI: 10.1556/2060.2022.00191] [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: 10/26/2021] [Revised: 03/21/2022] [Accepted: 04/28/2022] [Indexed: 02/18/2024]
Abstract
Objective To investigate the role of IGF1 and SIRT1 pathways in protection of hydrogen peroxide (H2O2)-induced aging in H9c2 rat cardiomyocyte cells by testosterone. Methods The cells were treated with testosterone or up- or down-regulated for the IGF1 and SIRT1 genes and assessed for apoptosis, aging and expression of relevant genes. Results Aging was induced and the expression of SIRT1 and IGF1 was down-regulated after H2O2 treatment in H9c2 cells. The aging was attenuated in a dose-dependent manner after the cells were exposed to testosterone. Down-regulation of SIRT1 and IGF1expression was offset in the H2O2-treated cells co-treated with testosterone. Up- or down-regulation of IGF1 significantly reduced or increased senescence-associated beta-galactosidase (SA-β-gal) cells and the ROS level, respectively. In addition, SIRT1 expression was regulated by IGF1 expression. Down- or up-regulation of SIRT1 significantly decreased or increased the IGF1 levels, respectively. Furthermore, after IGF1 and SIRT1 knockdown, testosterone did not protect the cells from senescence. Testosterone, and overexpression of IGF1 and SIRT1 also up-regulated the expression of the fetal genes SERCA2 and MYH6 and down-regulated the expression of the ACTA1 and MYH7 genes. Conclusions Our data indicate that testosterone can attenuate cardiomyocyte aging induced by H2O2 and up-regulate SIRT1 and IGF1. The IGF1and SIRT1 pathway may be new targets to treat heart aging and heart failure.
Collapse
Affiliation(s)
- Li Yan
- 1 Department of Cardiology, Shaanxi Provincial People Hospital, Xian, China
| | - Xiting Nong
- 2 Department of Endocrinology, Xi'an Central Hospital, Xian, China
| | - Jizhao Deng
- 1 Department of Cardiology, Shaanxi Provincial People Hospital, Xian, China
| | - Guang Yang
- 1 Department of Cardiology, Shaanxi Provincial People Hospital, Xian, China
| |
Collapse
|
10
|
Yuan Y, Liang B, Liu XL, Liu WJ, Huang BH, Yang SB, Gao YZ, Meng JS, Li MJ, Ye T, Wang CZ, Hu XK, Xing DM. Targeting NAD+: is it a common strategy to delay heart aging? Cell Death Dis 2022; 8:230. [PMID: 35474295 PMCID: PMC9042931 DOI: 10.1038/s41420-022-01031-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/17/2022]
Abstract
Heart aging is the main susceptible factor to coronary heart disease and significantly increases the risk of heart failure, especially when the aging heart is suffering from ischemia-reperfusion injury. Numerous studies with NAD+ supplementations have suggested its use in anti-aging treatment. However, systematic reviews regarding the overall role of NAD+ in cardiac aging are scarce. The relationship between NAD+ signaling and heart aging has yet to be clarified. This review comprehensively summarizes the current studies on the role of NAD+ signaling in delaying heart aging from the following aspects: the influence of NAD+ supplementations on the aging heart; the relationship and cross-talks between NAD+ signaling and other cardiac aging-related signaling pathways; Importantly, the therapeutic potential of targeting NAD+ in delaying heart aging will be discussed. In brief, NAD+ plays a vital role in delaying heart aging. However, the abnormalities such as altered glucose and lipid metabolism, oxidative stress, and calcium overload could also interfere with NAD+ function in the heart. Therefore, the specific physiopathology of the aging heart should be considered before applying NAD+ supplementations. We believe that this article will help augment our understanding of heart aging mechanisms. In the meantime, it provides invaluable insights into possible therapeutic strategies for preventing age-related heart diseases in clinical settings.
Collapse
Affiliation(s)
- Yang Yuan
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Bing Liang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Xin-Lin Liu
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Wen-Jing Liu
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Bing-Huan Huang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Shan-Bo Yang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Yuan-Zhen Gao
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Jing-Sen Meng
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Meng-Jiao Li
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Ting Ye
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Chuan-Zhi Wang
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China
| | - Xiao-Kun Hu
- Interventional Medicine Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dong-Ming Xing
- Cancer Institute of The Affiliated Hospital of Qingdao University and Qingdao Cancer Institute, Qingdao, China. .,School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
11
|
Li J, Zeng X, Yang F, Wang L, Luo X, Liu R, Zeng F, Lu S, Huang X, Lei Y, Lan Y. Resveratrol: Potential Application in Sepsis. Front Pharmacol 2022; 13:821358. [PMID: 35222035 PMCID: PMC8864164 DOI: 10.3389/fphar.2022.821358] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/21/2022] [Indexed: 01/02/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction syndrome caused by host response disorders due to infection or infectious factors and is a common complication of patients with clinical trauma, burns, and infection. Resveratrol is a natural polyphenol compound that is a SIRT-1 activator with anti-inflammatory, antiviral, antibacterial, antifungal inhibitory abilities as well as cardiovascular and anti-tumor protective effects. In recent years, some scholars have applied resveratrol in animal models of sepsis and found that it has an organ protective effect and can improve the survival time and reduce the mortality of animals with sepsis. In this study, Medline (Pubmed), embase, and other databases were searched to retrieve literature published in 2021 using the keywords “resveratrol” and “sepsis,” and then the potential of resveratrol for the treatment of sepsis was reviewed and prospected to provide some basis for future clinical research.
Collapse
Affiliation(s)
- Jiajia Li
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoting Zeng
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fuxun Yang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Wang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiu Luo
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongan Liu
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fan Zeng
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Sen Lu
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaobo Huang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Lei
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunping Lan
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
12
|
Rahmani J, Montesanto A, Giovannucci E, Zand H, Barati M, Kopchick JJ, Mirisola MG, Lagani V, Bawadi H, Vardavas R, Laviano A, Christensen K, Passarino G, Longo VD. Association between IGF-1 levels ranges and all-cause mortality: A meta-analysis. Aging Cell 2022; 21:e13540. [PMID: 35048526 PMCID: PMC8844108 DOI: 10.1111/acel.13540] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/07/2021] [Indexed: 01/08/2023] Open
Abstract
The association between IGF‐1 levels and mortality in humans is complex with low levels being associated with both low and high mortality. The present meta‐analysis investigates this complex relationship between IGF‐1 and all‐cause mortality in prospective cohort studies. A systematic literature search was conducted in PubMed/MEDLINE, Scopus, and Cochrane Library up to September 2019. Published studies were eligible for the meta‐analysis if they had a prospective cohort design, a hazard ratio (HR) and 95% confidence interval (CI) for two or more categories of IGF‐1 and were conducted among adults. A random‐effects model with a restricted maximum likelihood heterogeneity variance estimator was used to find combined HRs for all‐cause mortality. Nineteen studies involving 30,876 participants were included. Meta‐analysis of the 19 eligible studies showed that with respect to the low IGF‐1 category, higher IGF‐1 was not associated with increased risk of all‐cause mortality (HR = 0.84, 95% CI = 0.68–1.05). Dose–response analysis revealed a U‐shaped relation between IGF‐1 and mortality HR. Pooled results comparing low vs. middle IGF‐1 showed a significant increase of all‐cause mortality (HR = 1.33, 95% CI = 1.14–1.57), as well as comparing high vs. middle IGF‐1 categories (HR = 1.23, 95% CI = 1.06–1.44). Finally, we provide data on the association between IGF‐1 levels and the intake of proteins, carbohydrates, certain vitamins/minerals, and specific foods. Both high and low levels of IGF‐1 increase mortality risk, with a specific 120–160 ng/ml range being associated with the lowest mortality. These findings can explain the apparent controversy related to the association between IGF‐1 levels and mortality.
Collapse
Affiliation(s)
- Jamal Rahmani
- Department of Community Nutrition Faculty of Nutrition and Food Technology National Nutrition and Food Technology Research Institute Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences University of Calabria Rende Italy
| | - Edward Giovannucci
- Departments of Nutrition Harvard TH Chan School of Public Health Boston Massachusetts USA
| | - Hamid Zand
- National Nutrition and Food Technology Research Institute Faculty of Nutrition Sciences and Food Technology Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Meisam Barati
- National Nutrition and Food Technology Research Institute Faculty of Nutrition Sciences and Food Technology Shahid Beheshti University of Medical Sciences Tehran Iran
| | - John J. Kopchick
- Department of Biomedical Sciences Heritage College of Osteopathic Medicine Ohio and Edison Biotechnology Institute Ohio University Athens Ohio USA
| | - Mario G. Mirisola
- Department of Surgical, Oncological and Stomatological Disciplines Università di Palermo Palermo Italy
| | - Vincenzo Lagani
- Institute of Chemical Biology Ilia State University Tbilisi Georgia USA
- Biological and Environmental Sciences and Engineering Division (BESE) King Abdullah University of Science and Technology KAUST Thuwal Saudi Arabia
| | - Hiba Bawadi
- Human Nutrition Department College of Health Sciences QU‐Health Qatar University Doha Qatar
| | | | - Alessandro Laviano
- Department of Translational and Precision Medicine Sapienza University Rome Italy
| | - Kaare Christensen
- Danish Aging Research Center University of Southern Denmark Odense Denmark
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences University of Calabria Rende Italy
| | - Valter D. Longo
- Longevity Institute Davis School of Gerontology and Department of Biological Sciences University of Southern California Los Angeles California USA
- IFOM FIRC Institute of Molecular Oncology Milan Italy
| |
Collapse
|
13
|
Cao P, Chen Q, Shi C, Pei M, Wang L, Gong Z. Pinocembrin ameliorates acute liver failure via activating the Sirt1/PPARα pathway in vitro and in vivo. Eur J Pharmacol 2022; 915:174610. [PMID: 34951978 DOI: 10.1016/j.ejphar.2021.174610] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
Acute liver failure (ALF) is a life-threatening disease and affects multiple organ systems. Pro-inflammatory factors derived from macrophage plays a key role in septicemia. Pinocembrin is a natural favonoid compound, which can be extracted from honey, propolis and several other plants. Recent investigations demonstrate that Pinocembrin has a variety of pharmacological activities, including anti-inflammatory and antioxidant. To investigate the effects of Pinocembrin on ALF, we explored its possible molecular mechanisms through the experiments in vivo and in vitro. Pre-treatment with Pinocembrin attenuated LPS-induced hepatocyte dysfunction and reduced levels of pro-inflammatory factors and macrophages infiltration. Pinocembrin inhibited the hepatocyte apoptosis and pro-inflammatory reaction of peritoneal macrophages by reducing reactive oxygen species (ROS) via the Sirt1/PPARα signaling pathway. Our study suggests that Pinocembrin might represent a novel therapeutic drug and offers a new method for the treatment of ALF.
Collapse
Affiliation(s)
- Pan Cao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qian Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Maohua Pei
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Luwen Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
14
|
de Moraes Arnoso BJ, Magliaccio FM, de Araújo CA, de Andrade Soares R, Santos IB, de Bem GF, Fernandes-Santos C, Ognibene DT, de Moura RS, Resende AC, Daleprane JB, Costa CAD. Açaí seed extract (ASE) rich in proanthocyanidins improves cardiovascular remodeling by increasing antioxidant response in obese high-fat diet-fed mice. Chem Biol Interact 2022; 351:109721. [PMID: 34715092 DOI: 10.1016/j.cbi.2021.109721] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/24/2021] [Accepted: 10/23/2021] [Indexed: 02/06/2023]
Abstract
Obesity is recognized as an independent risk factor for cardiovascular diseases and is an important contributor to cardiac mortality. Açaí seed extract (ASE), rich in proanthocyanidins, has been shown to have potential anti-obesity effects. This study aimed to investigate the therapeutic effect of ASE in cardiovascular remodeling associated with obesity and compare it with that of rosuvastatin. Male C57BL/6 mice were fed a high-fat diet or a standard diet for 12 weeks. The ASE (300 mg/kg/day) and rosuvastatin (20 mg/kg/day) treatments started in the 8th week until the 12th week, totaling 4 weeks of treatment. Our data showed that treatment with ASE and rosuvastatin reduced body weight, ameliorated lipid profile, and improved cardiovascular remodeling. Treatment with ASE but not rosuvastatin reduced hyperglycemia and oxidative stress by reducing immunostaining of 8-isoprostane and increasing SOD-1 and GPx expression in HFD mice. ASE and rosuvastatin reduced NOX4 expression, increased SIRT-1 and Nrf2 expression and catalase and GPx activities, and improved vascular and cardiac remodeling in HFD mice. The therapeutic effect of ASE was similar to that of rosuvastatin in reducing dyslipidemia and cardiovascular remodeling but was superior in reducing oxidative damage and hyperglycemia, suggesting that ASE was a promising natural product for the treatment of cardiovascular alterations associated with obesity.
Collapse
Affiliation(s)
| | - Fabrizia Mansur Magliaccio
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Caroline Alves de Araújo
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Ricardo de Andrade Soares
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Izabelle Barcellos Santos
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Graziele Freitas de Bem
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Caroline Fernandes-Santos
- Department of Basic Sciences, Institute of Health, Fluminense Federal University, Nova Friburgo, RJ, Brazil
| | - Dayane Teixeira Ognibene
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Roberto Soares de Moura
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Angela Castro Resende
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Julio Beltrame Daleprane
- Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Cristiane Aguiar da Costa
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
15
|
Hajializadeh Z, Khaksari M. The protective effects of 17-β estradiol and SIRT1 against cardiac hypertrophy: a review. Heart Fail Rev 2021; 27:725-738. [PMID: 34537933 DOI: 10.1007/s10741-021-10171-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 12/27/2022]
Abstract
One of the major causes of morbidity and mortality worldwide is cardiac hypertrophy (CH), which leads to heart failure. Sex differences in CH can be caused by sex hormones or their receptors. The incidence of CH increases in postmenopausal women due to the decrease in female sex hormone 17-β estradiol (E2) during menopause. E2 and its receptors inhibit CH in humans and animal models. Silent information regulator 1 (SIRT1) is a NAD+-dependent HDAC (histone deacetylase) and plays a major role in biological processes, such as inflammation, apoptosis, and oxidative stress responses. Probably SIRT1 because of these effects, is one of the main suppressors of CH and has a cardioprotective effect. On the other hand, estrogen and its agonists are highly efficient in modulating SIRT1 expression. In the present study, we review the protective effects of E2 and SIRT1 against CH.
Collapse
Affiliation(s)
- Zahra Hajializadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
16
|
Valizadeh A, Asghari S, Mansouri P, Alemi F, Majidinia M, Mahmoodpoor A, Yousefi B. The roles of signaling pathways in cardiac regeneration. Curr Med Chem 2021; 29:2142-2166. [PMID: 34521319 DOI: 10.2174/0929867328666210914115411] [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: 03/03/2021] [Revised: 07/05/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
In recent years, knowledge of cardiac regeneration mechanisms has dramatically expanded. Regeneration can replace lost parts of organs, common among animal species. The heart is commonly considered an organ with terminal development, which has no reparability potential during post-natal life; however, some intrinsic regeneration capacity has been reported for cardiac muscle, which opens novel avenues in cardiovascular disease treatment. Different endogenous mechanisms were studied for cardiac repairing and regeneration in recent decades. Survival, proliferation, inflammation, angiogenesis, cell-cell communication, cardiomyogenesis, and anti-aging pathways are the most important mechanisms that have been studied in this regard. Several in vitro and animal model studies focused on proliferation induction for cardiac regeneration reported promising results. These studies have mainly focused on promoting proliferation signaling pathways and demonstrated various signaling pathways such as Wnt, PI3K/Akt, IGF-1, TGF-β, Hippo, and VEGF signaling cardiac regeneration. Therefore, in this review, we intended to discuss the connection between different critical signaling pathways in cardiac repair and regeneration.
Collapse
Affiliation(s)
- Amir Valizadeh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Samira Asghari
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Parinaz Mansouri
- Students Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Forough Alemi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia. Iran
| | - Ata Mahmoodpoor
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
17
|
Chen J, Su Y, Lin F, Iqbal M, Mehmood K, Zhang H, Shi D. Effect of paraquat on cytotoxicity involved in oxidative stress and inflammatory reaction: A review of mechanisms and ecological implications. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 224:112711. [PMID: 34455184 DOI: 10.1016/j.ecoenv.2021.112711] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/07/2021] [Accepted: 08/24/2021] [Indexed: 06/13/2023]
Abstract
Paraquat (PQ) is a cheap and an effective herbicide, which is widely being used worldwide to remove weeds in cultivated crop fields. However, it can cause soil and water pollution, and pose serious harm to the environment and organisms. Several countries have started to limit or prohibit the use of PQ because of the increasing number of human deaths. Its toxicity can damage the organisms with a multi-target mechanism, which has not been fully understood yet. That is why it is hard to treat as well. The current research on PQ focuses on its targeted organ, the lungs, in which PQ mostly trigger pulmonary fibrosis. While there is a lack of systematic research, there are few studies published discussing its toxic effects at systematic level. This review summarizes the major damages caused by PQ in different organisms and partial mechanisms by which it causes these damages. For this purpose, we consulted several research articles that studied the toxicity of PQ in various tissues. We also listed some drugs that can be used to alleviate the toxicity of PQ. However, at present, the effectiveness of these drugs is still being explored in animal experiments and the study of their mechanism will also help in understanding the poisoning mechanism of PQ, which will ultimately lead to effective treatment in future.
Collapse
Affiliation(s)
- Jiaxin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yalin Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Fei Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Mujahid Iqbal
- Department of Pathology, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur 63100, Pakistan
| | - Khalid Mehmood
- Department of Clinical Medicine and Surgery, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Dayou Shi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
18
|
Donia T, Khamis A. Management of oxidative stress and inflammation in cardiovascular diseases: mechanisms and challenges. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:34121-34153. [PMID: 33963999 DOI: 10.1007/s11356-021-14109-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/21/2021] [Indexed: 06/12/2023]
Abstract
Cardiovascular diseases (CVDs) have diverse physiopathological mechanisms with interconnected oxidative stress and inflammation as one of the common etiologies which result in the onset and development of atherosclerotic plaques. In this review, we illustrate this strong crosstalk between oxidative stress, inflammation, and CVD. Also, mitochondrial functions underlying this crosstalk, and various approaches for the prevention of redox/inflammatory biological impacts will be illustrated. In part, we focus on the laboratory biomarkers and physiological tests for the evaluation of oxidative stress status and inflammatory processes. The impact of a healthy lifestyle on CVD onset and development is displayed as well. Furthermore, the differences in oxidative stress and inflammation are related to genetic susceptibility to cardiovascular diseases and the variability in the assessment of CVDs risk between individuals; Omics technologies for measuring oxidative stress and inflammation will be explored. Finally, we display the oxidative stress-related microRNA and the functions of the redox basis of epigenetic modifications.
Collapse
Affiliation(s)
- Thoria Donia
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Abeer Khamis
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| |
Collapse
|
19
|
Wang M, Lin H. Understanding the Function of Mammalian Sirtuins and Protein Lysine Acylation. Annu Rev Biochem 2021; 90:245-285. [PMID: 33848425 DOI: 10.1146/annurev-biochem-082520-125411] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein lysine acetylation is an important posttranslational modification that regulates numerous biological processes. Targeting lysine acetylation regulatory factors, such as acetyltransferases, deacetylases, and acetyl-lysine recognition domains, has been shown to have potential for treating human diseases, including cancer and neurological diseases. Over the past decade, many other acyl-lysine modifications, such as succinylation, crotonylation, and long-chain fatty acylation, have also been investigated and shown to have interesting biological functions. Here, we provide an overview of the functions of different acyl-lysine modifications in mammals. We focus on lysine acetylation as it is well characterized, and principles learned from acetylation are useful for understanding the functions of other lysine acylations. We pay special attention to the sirtuins, given that the study of sirtuins has provided a great deal of information about the functions of lysine acylation. We emphasize the regulation of sirtuins to illustrate that their regulation enables cells to respond to various signals and stresses.
Collapse
Affiliation(s)
- Miao Wang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA;
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA; .,Howard Hughes Medical Institute, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
20
|
Differential role of SIRT1/MAPK pathway during cerebral ischemia in rats and humans. Sci Rep 2021; 11:6339. [PMID: 33737560 PMCID: PMC7973546 DOI: 10.1038/s41598-021-85577-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
Cerebral ischemia (CI) is a severe cause of neurological dysfunction and mortality. Sirtuin-1 (Silent information regulator family protein 1, SIRT1), an oxidized nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, plays an important role in protection against several neurodegenerative disorders. The present study aims to investigate the protective role of SIRT1 after CI in experimental young and aged rats and humans. Also, the study examines the possible regulatory mechanisms of neuronal death in CI settings. Immunoblotting and immunohistochemistry were used to evaluate changes in the expression of SIRT1, JNK/ERK/MAPK/AKT signaling, and pro-apoptotic caspase-3 in experimental rats and CI patients. The study findings demonstrated that, in aged experimental rats, SIRT1 activation positively influenced JNK and ERK phosphorylation and modulated neuronal survival in AKT-dependent manner. Further, the protection conferred by SIRT1 was effectively reversed by JNK inhibition and increased pro-apoptotic caspase-3 expression. In young experimental rats, SIRT1 activation decreased the phosphorylation of stress-induced JNK, ERK, caspase-3, and increased the phosphorylation of AKT after CI. Inhibition of SIRT1 reversed the protective effect of resveratrol. More importantly, in human patients, SIRT1 expression, phosphorylation of JNK/ERK/MAPK/AKT signaling and caspase-3 were up-regulated. In conclusion, SIRT1 could possibly be involved in the modulation of JNK/ERK/MAPK/AKT signaling pathway in experimental rats and humans after CI.
Collapse
|
21
|
Asensio-Lopez MC, Sassi Y, Soler F, Fernandez Del Palacio MJ, Pascual-Figal D, Lax A. The miRNA199a/SIRT1/P300/Yy1/sST2 signaling axis regulates adverse cardiac remodeling following MI. Sci Rep 2021; 11:3915. [PMID: 33594087 PMCID: PMC7887255 DOI: 10.1038/s41598-021-82745-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/25/2021] [Indexed: 01/14/2023] Open
Abstract
Left ventricular remodeling following myocardial infarction (MI) is related to adverse outcome. It has been shown that an up-regulation of plasma soluble ST2 (sST2) levels are associated with lower pre-discharge left ventricular (LV) ejection fraction, adverse cardiovascular outcomes and mortality outcome after MI. The mechanisms involved in its modulation are unknown and there is not specific treatment capable of lowering plasma sST2 levels in acute-stage HF. We recently identified Yin-yang 1 (Yy1) as a transcription factor related to circulating soluble ST2 isoform (sST2) expression in infarcted myocardium. However, the underlying mechanisms involved in this process have not been thoroughly elucidated. This study aimed to evaluate the pathophysiological implication of miR-199a-5p in cardiac remodeling and the expression of the soluble ST2 isoform. Myocardial infarction (MI) was induced by permanent ligation of the left anterior coronary artery in C57BL6/J mice that randomly received antimiR199a therapy, antimiR-Ctrl or saline. A model of biomechanical stretching was also used to characterize the underlying mechanisms involved in the activation of Yy1/sST2 axis. Our results show that the significant upregulation of miR-199a-5p after myocardial infarction increases pathological cardiac hypertrophy by upregulating circulating soluble sST2 levels. AntimiR199a therapy up-regulates Sirt1 and inactivates the co-activator P300 protein, thus leading to Yy1 inhibition which decreases both expression and release of circulating sST2 by cardiomyocytes after myocardial infarction. Pharmacological inhibition of miR-199a rescues cardiac hypertrophy and heart failure in mice, offering a potential therapeutic approach for cardiac failure.
Collapse
Affiliation(s)
- Maria Carmen Asensio-Lopez
- Biomedical Research Institute Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Yassine Sassi
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Fernando Soler
- Biochemical and Molecular Biology Department, University of Murcia, Murcia, Spain
| | | | - Domingo Pascual-Figal
- Cardiology Department, Hospital Virgen de la Arrixaca, IMIB-Arrixaca, University of Murcia, Murcia, Spain.
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- CIBERCV, Madrid, Spain.
| | - Antonio Lax
- Biomedical Research Institute Virgen de la Arrixaca (IMIB-Arrixaca), University of Murcia, Murcia, Spain.
| |
Collapse
|
22
|
Chen L, Li S, Zhu J, You A, Huang X, Yi X, Xue M. Mangiferin prevents myocardial infarction-induced apoptosis and heart failure in mice by activating the Sirt1/FoxO3a pathway. J Cell Mol Med 2021; 25:2944-2955. [PMID: 33523605 PMCID: PMC7957271 DOI: 10.1111/jcmm.16329] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 01/04/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023] Open
Abstract
Myocardial infarction (MI) commonly leads to cardiomyocyte apoptosis and heart failure. Mangiferin is a natural glucosylxanthone extracted from mango fruits and leaves, which has anti-apoptotic and anti-inflammatory properties in experimental cardiovascular diseases. In the present study, we investigated the role and detailed mechanism of mangiferin in MI. We used ligation of the left anterior descending coronary artery to establish an MI model in vivo, and cardiomyocyte-specific Sirt1 knockout mice were used to identify the mechanism of mangiferin. For in vitro studies, oxygen and glucose deprivation (OGD) was used to mimic ischaemia in H9c2 cardiomyocytes. In mice, mangiferin treatment increased Sirt1 expression after MI, significantly reduced the infarct area, and prevented MI-induced apoptosis and heart failure. Mangiferin reduced OGD-induced cellular apoptosis in H9c2 cells. Meanwhile, Sirt1 knockout/silencing abolished the protective effects of mangiferin. Further studies revealed that mangiferin increased FoxO3a deacetylation by up-regulating Sirt1, thus preventing apoptosis, and adenovirus-mediated constitutive acetylation of FoxO3a restricted the anti-apoptotic effects of mangiferin in vivo and in vitro. Our results indicate that mangiferin prevents cardiomyocyte apoptosis and the subsequent heart failure by activating the Sirt1/FoxO3a pathway in MI, and suggest that mangiferin may have an interesting potential in following studies towards clinical evaluation.
Collapse
Affiliation(s)
- Lingli Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Santie Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Jianyu Zhu
- Department of Traumatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Anfu You
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xingzhou Huang
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinchu Yi
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China.,People's Hospital of Deyang City, Deyang, China
| | - Mei Xue
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
The relationship between coronary artery disease and SIRT1 protein. North Clin Istanb 2021; 7:631-635. [PMID: 33381707 PMCID: PMC7754863 DOI: 10.14744/nci.2020.31391] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 02/06/2020] [Indexed: 01/07/2023] Open
Abstract
Endothelial cell dysfunction proceeding with increased inflammation and monocyte increase is one of the main causes of vessel injury in CAD. SIRT1 (Sirtuin 1) protein plays an important role in the regulation of cellular physiological mechanisms. SIRT1 has roles in regulating angiogenesis and preventing endothelial dysfunction and reperfusion injury due to ischemia. Suppression of SIRT1 causes monocyte affinity due to endothelial dysfunction. Sirtuins activators are involved in pathologies of many diseases with promising treatments. The objective of this review is to summarize the current progress and future directions of sirtuin protein in the field of CAD.
Collapse
|
24
|
Szabó MR, Pipicz M, Csont T, Csonka C. Modulatory Effect of Myokines on Reactive Oxygen Species in Ischemia/Reperfusion. Int J Mol Sci 2020; 21:ijms21249382. [PMID: 33317180 PMCID: PMC7763329 DOI: 10.3390/ijms21249382] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
There is a growing body of evidence showing the importance of physical activity against acute ischemic events in various organs. Ischemia/reperfusion injury (I/R) is characterized by tissue damage as a result of restriction and subsequent restoration of blood supply to an organ. Oxidative stress due to increased reactive oxygen species formation and/or insufficient antioxidant defense is considered to play an important role in I/R. Physical activity not only decreases the general risk factors for ischemia but also confers direct anti-ischemic protection via myokine production. Myokines are skeletal muscle-derived cytokines, representing multifunctional communication channels between the contracting skeletal muscle and other organs through an endocrine manner. In this review, we discuss the most prominent members of the myokines (i.e., brain-derived neurotrophic factor (BDNF), cathepsin B, decorin, fibroblast growth factors-2 and -21, follistatin, follistatin-like, insulin-like growth factor-1; interleukin-6, interleukin-7, interleukin-15, irisin, leukemia inhibitory factor, meteorin-like, myonectin, musclin, myostatin, and osteoglycin) with a particular interest in their potential influence on reactive oxygen and nitrogen species formation or antioxidant capacity. A better understanding of the mechanism of action of myokines and particularly their participation in the regulation of oxidative stress may widen their possible therapeutic use and, thereby, may support the fight against I/R.
Collapse
Affiliation(s)
- Márton Richárd Szabó
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Márton Pipicz
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Csaba Csonka
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, University of Szeged, Dóm tér 9, 6720 Szeged, Hungary; (M.R.S.); (M.P.); (T.C.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
- Department of Sports Medicine, University of Szeged, Tisza Lajos krt 107, 6725 Szeged, Hungary
- Correspondence: ; Tel.: +36-30-5432-693
| |
Collapse
|
25
|
Yu X, Zheng J, Cai T, Wang Z, Zhu G. Testosterone antagonizes paraquat-induced cardiomyocyte senescence via the mIGF-1/SIRT1 signaling pathway. ACTA ACUST UNITED AC 2020; 53:e9849. [PMID: 32901689 PMCID: PMC7485312 DOI: 10.1590/1414-431x20209849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/29/2020] [Indexed: 12/08/2022]
Abstract
Testosterone has been demonstrated to antagonize doxorubicin-induced cardiomyocyte senescence. However, whether testosterone prevents the paraquat-induced cardiomyocyte senescence is largely unknown. The detection of SA-β-gal activity was performed using senescence β-gal staining kit and the reactive oxygen species levels were determined by reactive oxygen species assay kit. The plasmids for insulin-like growth factor 1 shRNA (sh-mIGF-1), sirtuin-1 shRNA (sh-SIRT1), scramble shRNA (sh-NC), overexpressing mIGF-1 (mIGF-1), overexpressing SIRT1 (SIRT1), and negative controls (NC) were obtained for this study. The expression of target genes was detected using quantitative real-time PCR, immunolabeling, and western blot. We found that testosterone significantly delayed the paraquat-induced HL-1 cardiomyocyte senescence as evidenced by decreasing senescence-associated β-galactosidase activity and reactive oxygen species generation, which were accompanied by the up-regulated expression of mIGF-1 and SIRT1. RNA interference to reduce mIGF-1 and SIRT1 expression showed that testosterone prevented paraquat-induced HL-1 senescence via the mIGF-1/SIRT1 signaling pathway. Furthermore, myocardial contraction was evaluated by expression of genes of the contractile proteins/enzymes, such as α-myosin heavy chain 6 (MHC6), α-myosin heavy chain 7 (MHC7), α-skeletal actin (ACTA-1), and sarco/endoplasmic reticulum calcium ATPase-2 (SERCA2). Testosterone adjusted the above four gene expressions and the adjustment was blocked by mIGF-1 or SIRT1 inhibition. Our findings suggested that the mIGF-1/SIRT1 signaling pathway mediated the protective function of testosterone against the HL-1 cardiomyocyte senescence by paraquat, which provided new clues for the mechanisms underlying the anti-aging role of testosterone in cardiomyocytes.
Collapse
Affiliation(s)
- Xing Yu
- Cardiovascular Department, First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jianyi Zheng
- Cardiovascular Department, First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Tengfei Cai
- Cardiovascular Department, First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Zhijian Wang
- Cardiovascular Department, First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Guiping Zhu
- Cardiovascular Department, First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Zhang D, Li B, Li B, Tang Y. Regulation of left atrial fibrosis induced by mitral regurgitation by SIRT1. Sci Rep 2020; 10:7278. [PMID: 32350389 PMCID: PMC7190846 DOI: 10.1038/s41598-020-64308-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/10/2020] [Indexed: 11/09/2022] Open
Abstract
SIRT1 (silent information regulator 1) is a histone deacetylase. It can sense the energy level in cells and delay cell senescence, leading to resistance to external stress and improving metabolism. Mitral regurgitation (MR) is a common disease in cardiac surgery. However, there are no previous studies on SIRT1 and left atrial fibrosis caused by MR. In this study, we aimed to explore the regulatory effect of SIRT1 on left atrial fibrosis induced by MR. We used Guizhou miniature pigs to establish an MR model and a sham operation model after anaesthesia induction and respiratory intubation, and these model animals were followed for 30 months after the surgery. The differential distribution and expression of SIRT1 and collagen I in the left atrium was determined by immunofluorescence and Western blotting. Furthermore, we treated NIH3T3 fibroblasts (CFs) with resveratrol and Angiotensin II (Ang II) to analyse the specific mechanism involved in the development of myocardial fibrosis. The results showed that the MR model was successfully constructed. There were 8 pigs in the MR group and 6 pigs in the control group. In both the animal experiments and the cell experiments, the expression of collagen I in the MR group was increased significantly compared to that in the control group, while the expression of SIRT1 was decreased.
Collapse
Affiliation(s)
- Dong Zhang
- Beijing Jishuitan Hospital, Department of Thoracic Surgery, Beijing, China
| | - Bo Li
- The Seventh Affiliated Hospital, Sun Yat-sen University, Department of Cardiac Surgery, Shenzhen, China
| | - Bin Li
- Animal Experimental Centre, Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Tang
- Animal Experimental Centre, Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
27
|
Mohammed SA, Ambrosini S, Lüscher T, Paneni F, Costantino S. Epigenetic Control of Mitochondrial Function in the Vasculature. Front Cardiovasc Med 2020; 7:28. [PMID: 32195271 PMCID: PMC7064473 DOI: 10.3389/fcvm.2020.00028] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/19/2020] [Indexed: 12/24/2022] Open
Abstract
The molecular signatures of epigenetic regulation and chromatin architecture are emerging as pivotal regulators of mitochondrial function. Recent studies unveiled a complex intersection among environmental factors, epigenetic signals, and mitochondrial metabolism, ultimately leading to alterations of vascular phenotype and increased cardiovascular risk. Changing environmental conditions over the lifetime induce covalent and post-translational chemical modification of the chromatin template which sensitize the genome to establish new transcriptional programs and, hence, diverse functional states. On the other hand, metabolic alterations occurring in mitochondria affect the availability of substrates for chromatin-modifying enzymes, thus leading to maladaptive epigenetic signatures altering chromatin accessibility and gene transcription. Indeed, several components of the epigenetic machinery require intermediates of cellular metabolism (ATP, AcCoA, NADH, α-ketoglutarate) for enzymatic function. In the present review, we describe the emerging role of epigenetic modifications as fine tuners of gene transcription in mitochondrial dysfunction and vascular disease. Specifically, the following aspects are described in detail: (i) mitochondria and vascular function, (ii) mitochondrial ROS, (iii) epigenetic regulation of mitochondrial function; (iv) the role of mitochondrial metabolites as key effectors for chromatin-modifying enzymes; (v) epigenetic therapies. Understanding epigenetic routes may pave the way for new approaches to develop personalized therapies to prevent mitochondrial insufficiency and its complications.
Collapse
Affiliation(s)
- Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Thomas Lüscher
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Research, Education and Development, Royal Brompton and Harefield Hospital Trust and Imperial College, London, United Kingdom
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| |
Collapse
|
28
|
Feng J, Wu Y, Chen W, Li J, Wang X, Chen Y, Yu Y, Shen Z, Zhang Y. Sustained release of bioactive IGF-1 from a silk fibroin microsphere-based injectable alginate hydrogel for the treatment of myocardial infarction. J Mater Chem B 2019; 8:308-315. [PMID: 31808500 DOI: 10.1039/c9tb01971e] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Low circulating levels of insulin-like growth factor 1 (IGF-1) have been correlated with an increased risk for cardiovascular diseases in humans. In this work, an injectable alginate hydrogel containing silk fibroin (SF) microspheres with the capability to sustain the release of IGF-1 was prepared to induce myocardial repair after myocardial infarction (MI). First, IGF-1 was physically adsorbed onto SF microspheres prepared by the coaxial needle system, and these IGF-1-containing microspheres were subsequently encapsulated into sodium alginate solutions at different concentrations (1.0-2.5%). Finally, this solution was crosslinked with 0.68% calcium gluconate solution to prepare the composite injectable hydrogel. The composite hydrogel prepared using a sodium alginate solution at a concentration of 1.5% could promote proliferation of H9C2 cardiomyocytes and reduce the cellular apoptosis rate under hypoxic conditions. The enzyme-linked immunosorbent assay results indicated that SF microspheres as microcarriers could effectively enhance the sustained release of IGF-1 from the hydrogels, causing the composite hydrogel to possess a better sustained release ability than the system without the SF microspheres. Moreover, echocardiography, hematoxylin-eosin staining, and Masson trichrome staining results indicated that an intramyocardial injection of the composite hydrogel into the peripheral region of a MI rat model could reduce the infarct size and improve the cardiac function after 28 days. The applications of such a composite hydrogel may comprise a powerful platform in cardiac tissue engineering.
Collapse
Affiliation(s)
- Jianguo Feng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China. and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, P. R. China and The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, P. R. China
| | - Yong Wu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China. and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, P. R. China
| | - Weiqian Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China. and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, P. R. China
| | - Jingjing Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China. and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, P. R. China
| | - Xiaoyu Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China. and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, P. R. China
| | - Yueqiu Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China. and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, P. R. China
| | - Yunsheng Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China. and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, P. R. China
| | - Zhenya Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China. and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, P. R. China
| | - Yanxia Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P. R. China. and Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu 215007, P. R. China
| |
Collapse
|
29
|
Chang Z, Xia J, Wu H, Peng W, Jiang F, Li J, Liang C, Zhao H, Park K, Song G, Kim S, Huang R, Zheng L, Cai D, Qi X. Forkhead box O3 protects the heart against paraquat-induced aging-associated phenotypes by upregulating the expression of antioxidant enzymes. Aging Cell 2019; 18:e12990. [PMID: 31264342 PMCID: PMC6718552 DOI: 10.1111/acel.12990] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 05/10/2019] [Accepted: 05/23/2019] [Indexed: 11/30/2022] Open
Abstract
Paraquat (PQ) promotes cell senescence in brain tissue, which contributes to Parkinson's disease. Furthermore, PQ induces heart failure and oxidative damage, but it remains unknown whether and how PQ induces cardiac aging. Here, we demonstrate that PQ induces phenotypes associated with senescence of cardiomyocyte cell lines and results in cardiac aging‐associated phenotypes including cardiac remodeling and dysfunction in vivo. Moreover, PQ inhibits the activation of Forkhead box O3 (FoxO3), an important longevity factor, both in vitro and in vivo. We found that PQ‐induced senescence phenotypes, including proliferation inhibition, apoptosis, senescence‐associated β‐galactosidase activity, and p16INK4a expression, were significantly enhanced by FoxO3 deficiency in cardiomyocytes. Notably, PQ‐induced cardiac remolding, apoptosis, oxidative damage, and p16INK4a expression in hearts were exacerbated by FoxO3 deficiency. In addition, both in vitro deficiency and in vivo deficiency of FoxO3 greatly suppressed the activation of antioxidant enzymes including catalase (CAT) and superoxide dismutase 2 (SOD2) in the presence of PQ, which was accompanied by attenuation in cardiac function. The direct in vivo binding of FoxO3 to the promoters of the Cat and Sod2 genes in the heart was verified by chromatin immunoprecipitation (ChIP). Functionally, overexpression of Cat or Sod2 alleviated the PQ‐induced senescence phenotypes in FoxO3‐deficient cardiomyocyte cell lines. Overexpression of FoxO3 and CAT in hearts greatly suppressed the PQ‐induced heart injury and phenotypes associated with aging. Collectively, these results suggest that FoxO3 protects the heart against an aging‐associated decline in cardiac function in mice exposed to PQ, at least in part by upregulating the expression of antioxidant enzymes and suppressing oxidative stress.
Collapse
Affiliation(s)
- Zao‐Shang Chang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Jing‐Bo Xia
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Hai‐Yan Wu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Wen‐Tao Peng
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Fu‐Qing Jiang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Jing Li
- Department of Surgery, Union Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Chi‐Qian Liang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Hui Zhao
- Key Laboratory of Regenerative Medicine of Ministry of Education, School of Biomedical Sciences, Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China
| | - Kyu‐Sang Park
- Department of Physiology, Wonju College of Medicine Yonsei University Wonju Korea
| | - Guo‐Hua Song
- Institute of Atherosclerosis TaiShan Medical University Tai'an China
| | - Soo‐Ki Kim
- Department of Microbiology Wonju College of Medicine, Yonsei University Wonju Korea
| | - Ruijin Huang
- Institute of Anatomy, Department of Neuroanatomy, Medical Faculty Bonn Rheinische Friedrich-Wilhelms-University of Bonn Bonn Germany
| | - Li Zheng
- School of Environmental Science and Engineering Guangdong University of Technology Guangzhou China
| | - Dong‐Qing Cai
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Xu‐Feng Qi
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| |
Collapse
|
30
|
Fibroblast growth factor 21 protects the heart from angiotensin II-induced cardiac hypertrophy and dysfunction via SIRT1. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1241-1252. [DOI: 10.1016/j.bbadis.2019.01.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/29/2018] [Accepted: 01/16/2019] [Indexed: 12/22/2022]
|
31
|
Cx43 Inhibition Attenuates Sepsis-Induced Intestinal Injury via Downregulating ROS Transfer and the Activation of the JNK1/Sirt1/FoxO3a Signaling Pathway. Mediators Inflamm 2019; 2019:7854389. [PMID: 30948926 PMCID: PMC6425293 DOI: 10.1155/2019/7854389] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
Intestinal injury has long been considered to play a crucial role in the pathophysiology of sepsis and has even been characterized as the “motor” of it. Thus, we explored the effects of connexin43 (Cx43) on sepsis-induced intestinal injury in order to provide potential therapeutic strategies. Rat cecal ligation and puncture (CLP) models in vivo and cell models (IEC-6 cells) pretreated with LPS in vitro were used in the current study. Firstly, different methods, such as Cx43 inhibitors (18-α-GA and oleamide) or siRNA targeting Cx43 and N-acetyl cysteine (NAC) (a kind of ROS scavenger), were used to observe the effects of Cx43 channels mediating ROS transfer on intestinal injury. Secondly, the influence of ROS content on the activity of the JNK1/Sirt1/FoxO3a signaling pathway was explored through the application of NAC, sp600125 (a JNK1 inhibitor), and nicotinamide (a Sirt1 inhibitor). Finally, luciferase assays and ChIP were used to determine the direct regulation of FoxO3a on proapoptotic proteins, Bim and Puma. The results showed that sepsis-induced intestinal injury presented a dynamic change, coincident with the alternation of Cx43 expression. The inhibition of Cx43 attenuated CLP-induced intestinal injury in vivo and LPS-induced IEC-6 injury in vitro. The changes of Cx43 channel function regulated ROS transfer between the neighboring cells, which mediated the activation of the JNK1/Sirt1/FoxO3a signaling pathway. FoxO3a directly affected its downstream target genes, Bim and Puma, which are responsible for cell or tissue apoptosis. In summary, our results suggest that Cx43 inhibition suppresses ROS transfer and inactivates the JNK1/Sirt1/FoxO3a signaling pathway to protect against sepsis-induced intestinal injury.
Collapse
|
32
|
Manna D, Bhuyan R, Ghosh R. Probing the mechanism of SIRT1 activation by a 1,4-dihydropyridine. J Mol Model 2018; 24:340. [PMID: 30448921 DOI: 10.1007/s00894-018-3877-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/05/2018] [Indexed: 11/25/2022]
Abstract
The NAD+-dependent deacetylase SIRT1 plays important roles in several physiological processes such as transcription, genome stability, stress responses, and aging. Due to its diverse role in metabolisms, SIRT1 has emerged as a potential therapeutic target in many human disorders such as type II diabetes, cardiovascular and neurodegenerative diseases, and cancer. Recent studies have reported that modulation of SIRT1 activity by phenolic activators like resveratrol and some 1,4-dihydropyridines (1,4-DHPs) can inhibit tumor growth by promoting apoptosis in cancer cells. However, the mechanism of SIRT1 activation is still not clear. In this report, we have tried to elucidate the mechanism of SIRT1 activation from studies on its interaction with a synthetic 1,4-DHP derivative (DHP-8; 3,5-diethoxy carbonyl-4-(4-nitrophenyl)-2,6-dimethyl-1,4-dihydropyridine) using molecular modeling, docking, simulation, and free energy analyses. Owing to the absence of full-length human SIRT1 structure, multi-template based modeling approach was opted followed by docking of DHP-8 at its allosteric site. In presence of DHP-8, the overall conformation of SIRT1 was found to be more stable (especially at its substrate binding sites) with a large structural variation at its N-terminal domain while bound to substrate p53 or p53-W. Determination of the MM/PBSA free energy indicated that the binding of DHP-8 to SIRT1 significantly increased the binding affinity of SIRT1 to its substrate p53-W as well as to NAD+. Overall, this study depicts the atomistic detailed mechanism for the direct activation of SIRT1 by a 1,4-DHP. This would serve to develop new SIRT1 activators for future therapeutic perspectives.
Collapse
Affiliation(s)
- Debashri Manna
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Rajabrata Bhuyan
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Rita Ghosh
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, West Bengal, 741235, India.
| |
Collapse
|
33
|
Bernardo BC, Ooi JYY, Weeks KL, Patterson NL, McMullen JR. Understanding Key Mechanisms of Exercise-Induced Cardiac Protection to Mitigate Disease: Current Knowledge and Emerging Concepts. Physiol Rev 2018; 98:419-475. [PMID: 29351515 DOI: 10.1152/physrev.00043.2016] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The benefits of exercise on the heart are well recognized, and clinical studies have demonstrated that exercise is an intervention that can improve cardiac function in heart failure patients. This has led to significant research into understanding the key mechanisms responsible for exercise-induced cardiac protection. Here, we summarize molecular mechanisms that regulate exercise-induced cardiac myocyte growth and proliferation. We discuss in detail the effects of exercise on other cardiac cells, organelles, and systems that have received less or little attention and require further investigation. This includes cardiac excitation and contraction, mitochondrial adaptations, cellular stress responses to promote survival (heat shock response, ubiquitin-proteasome system, autophagy-lysosomal system, endoplasmic reticulum unfolded protein response, DNA damage response), extracellular matrix, inflammatory response, and organ-to-organ crosstalk. We summarize therapeutic strategies targeting known regulators of exercise-induced protection and the challenges translating findings from bench to bedside. We conclude that technological advancements that allow for in-depth profiling of the genome, transcriptome, proteome and metabolome, combined with animal and human studies, provide new opportunities for comprehensively defining the signaling and regulatory aspects of cell/organelle functions that underpin the protective properties of exercise. This is likely to lead to the identification of novel biomarkers and therapeutic targets for heart disease.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Jenny Y Y Ooi
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Natalie L Patterson
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| |
Collapse
|
34
|
Yamac AH, Huyut MA, Yilmaz E, Celikkale I, Bacaksiz A, Demir Y, Demir AR, Erturk M, Bakhshaliyev N, Ozdemir R, Kilic U. MicroRNA 199a Is Downregulated in Patients After Coronary Artery Bypass Graft Surgery and Is Associated with Increased Levels of Sirtuin 1 (SIRT 1) Protein and Major Adverse Cardiovascular Events at 3-Year Follow-Up. Med Sci Monit 2018; 24:6245-6254. [PMID: 30192743 PMCID: PMC6139112 DOI: 10.12659/msm.912065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background The cardioprotective protein SIRT1 is elevated in patients with coronary artery disease (CAD) to compensate for the disease-related adverse effects, but less is known about the prognostic role of SIRT 1 regulating microRNAs in patients after coronary artery bypass graft (CABG) surgery. Material/Methods The expression of the SIRT 1-specific microRNAs miR-199a and miR-195 was analyzed using real-time PCR in 68 patients referred for CABG surgery and 34 control patients undergoing heart valve surgery. In CABG patients, major adverse cardiac and cerebrovascular events (MACCEs), including all-cause death, myocardial infarction (MI), re-vascularization, heart failure symptoms ≥NYHA II, re-hospitalization for any cardiovascular reason, and stroke, were analyzed at a median follow-up (FU) of 3.2 years (range: 3.0–3.6). Results The level of miR-199a in patients with CAD was significantly reduced compared to the control group (relative expression: 0.89±0.49 vs. 1.90±0.90, p=0.001), while SIRT 1 protein was markedly enhanced (p<0.001). In patients undergoing CABG who had MACCEs, miR-199a was significantly lower compared to patients with an uneventful FU (0.71±0.25 vs. 0.98±0.53, p=0.007). Heart failure status, death, and total MACCEs rate were inversely correlated with the amount of miR-199a (p=0.039) at 3-year FU. Conclusions Altered expression of miR-199a in myocardial tissue was found to be associated with SIRT 1 upregulation in patients with CAD undergoing CABG and was associated with an increased MACCEs rate at mid-term follow-up.
Collapse
Affiliation(s)
- Aylin Hatice Yamac
- Department of Cardiology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Mustafa Ahmet Huyut
- Department of Cardiology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Emre Yilmaz
- Department of Cardiology, Mehmet Akif Ersoy Heart Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ilke Celikkale
- Department of Cardiology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ahmet Bacaksiz
- Department of Cardiology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Yusuf Demir
- Department of Cardiology, Mehmet Akif Ersoy Heart Hospital, University of Health Sciences, Istanbul, Turkey
| | - Ali Riza Demir
- Department of Cardiology, Mehmet Akif Ersoy Heart Hospital, University of Health Sciences, Istanbul, Turkey
| | - Mehmet Erturk
- Department of Cardiology, Mehmet Akif Ersoy Heart Hospital, University of Health Sciences, Istanbul, Turkey
| | - Nijad Bakhshaliyev
- Department of Cardiology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ramazan Ozdemir
- Department of Cardiology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ulkan Kilic
- Department of Medical Biology, Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
35
|
Li Y, Peng Z, Wang C, Li L, Leng Y, Chen R, Yuan H, Zhou S, Zhang Z, Chen AF. Novel role of PKR in palmitate-induced Sirt1 inactivation and endothelial cell senescence. Am J Physiol Heart Circ Physiol 2018; 315:H571-H580. [PMID: 29906232 DOI: 10.1152/ajpheart.00038.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Endothelial cell senescence is regarded as a vital characteristic of cardiovascular diseases. Elevated palmitate (PA) is an independent risk factor of cardiovascular diseases, but its role in endothelial cell senescence is currently unknown. During the course of studying the prosenescent role of PA, we discovered a key role of dsRNA-dependent protein kinase [protein kinase R (PKR)] in endothelial senescence. Exposure of human umbilical vein endothelial cells (HUVECs) to PA-induced cell senescence is characterized by increased levels of senescence-associated β-galactose glucosidase activity, excessive production of reactive oxygen species production, impaired cellular proliferation, and G1 phase arrest. This phenomenon is associated with an increase of PKR autophosphorylation and decreased activity of sirtuin 1 (Sirt1), a pivotal antisenescent factor. PKR inactivation by PKR siRNA or its phosphorylation inhibitor 2-aminopurine significantly attenuated PA-induced HUVEC senescence by reversing Sirt1 activity and its downstream signaling. Moreover, to study the regulatory mechanism between PKR and Sirt1, we found that PKR promotes JNK activation to inhibit Sirt1 activity and that this effect could be reversed by the JNK inhibitor SP600125. These findings provide evidence that PKR mediates PA-induced HUVEC senescence by inhibiting Sirt1 signaling. Our study provides novel insights into the actions and mechanisms of PKR in endothelial senescence. NEW & NOTEWORTHY This study first provides a novel observation that dsRNA-dependent protein kinase (PKR) mediates palmitate-induced sirtuin 1 inactivation and subsequent human umbilical vein endothelial cell senescence. Most importantly, these new findings will provide a potential therapeutic strategy to improve free fatty acid-induced endothelial senescence by targeting PKR in cardiovascular diseases.
Collapse
Affiliation(s)
- Yapei Li
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhouyangfan Peng
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Chunle Wang
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Le Li
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Yiping Leng
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
| | - Ruifang Chen
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hong Yuan
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shenghua Zhou
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhen Zhang
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
- The Center of Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Alex F. Chen
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital, Central South University, Changsha, China
- Department of Surgery, University of Pittsburgh School of Medicine, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Murofushi T, Tsuda H, Mikami Y, Yamaguchi Y, Suzuki N. CAY10591, a SIRT1 activator, suppresses cell growth, invasion, and migration in gingival epithelial carcinoma cells. J Oral Sci 2018; 59:415-423. [PMID: 28904318 DOI: 10.2334/josnusd.16-0696] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
SIRT1 is a NAD-dependent histone deacetylase that is important in a wide variety of physiological and pathophysiological processes. Although many studies have examined the relationship between SIRT1 and cancer, the role of SIRT1 in tumor malignancy is controversial. Here, we examined the effects of the SIRT1 activator CAY10591 in gingival epithelial carcinoma Ca9-22 cells. CAY10591 treatment dose- and time-dependently increased SIRT1 level and activity. The treatment decreased cell growth and induced cell-cycle repressor p21 levels. In addition, dimethyl sulfoxide significantly reduced cellular invasion and migration, and CAY10591 enhanced this decrease. Quantitative PCR analysis showed that CAY10591 decreased expression of several invasion/migration promoter genes and induced repressor genes. Our findings suggest that CAY10591 suppresses cell growth and invasion/migration activity in gingival squamous cell carcinoma Ca9-22 cells.
Collapse
Affiliation(s)
| | - Hiromasa Tsuda
- Department of Biochemistry, Nihon University School of Dentistry.,Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry
| | - Yoshikazu Mikami
- Division of Microscopic Anatomy, Niigata University Graduate School of Medical and Dental Sciences
| | - Yoko Yamaguchi
- Department of Biochemistry, Nihon University School of Dentistry.,Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry
| | - Naoto Suzuki
- Department of Biochemistry, Nihon University School of Dentistry.,Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry
| |
Collapse
|
37
|
Matsumoto R, Koga M, Kasayama S, Fukuoka H, Iguchi G, Odake Y, Yoshida K, Bando H, Suda K, Nishizawa H, Takahashi M, Ogawa W, Takahashi Y. Factors correlated with serum insulin-like growth factor-I levels in health check-up subjects. Growth Horm IGF Res 2018; 40:55-60. [PMID: 29395967 DOI: 10.1016/j.ghir.2018.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/04/2018] [Accepted: 01/18/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Most of circulating IGF-I is derived from the liver and circulating IGF-I levels are decreased in several pathological conditions, such as liver cirrhosis, uncontrolled diabetes, renal failure, and malnutrition. However, it has not fully been elucidated which factors modify IGF-I level in a physiological condition. OBJECTIVE To identify the factors which are associated with circulating IGF-I levels. DESIGN Cross-sectional study. METHODS This study included 428 subjects who undertook health check-up. Subjects diagnosed with non-alcoholic fatty liver disease (NAFLD) by ultrasonography were analyzed separately. Univariate and multivariate regression analyses were conducted to identify the factors associated with circulating IGF-I levels. RESULTS Regression analyses revealed that serum albumin levels, total-bilirubin levels, calcium levels, and HOMA-IR were positively correlated with IGF-I levels. Serum transaminase levels and habitual drinking (ethanol intake >20 g/day) were negatively correlated with serum IGF-I levels. Although serum IGF-I standard deviation scores (SDS) in subjects with and without NAFLD were comparable, after adjusting confounding factors clarified by multivariate regression analysis, IGF-I SDS negatively correlated with the presence of NAFLD. CONCLUSION In this study, we demonstrated that serum bilirubin and calcium levels are correlated with serum IGF-I levels. Although further study is necessary, these data suggest a presence of interaction between GH-IGF-I axis and bilirubin and calcium metabolism.
Collapse
Affiliation(s)
- Ryusaku Matsumoto
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Masafumi Koga
- Department of Internal Medicine, Hakuhokai Central Hospital, Hyogo, Japan
| | - Soji Kasayama
- Department of Medicine, Nissay Hospital, Osaka, Japan
| | - Hidenori Fukuoka
- Department of Diabetes and Endocrinology, Kobe University Hospital, Hyogo, Japan
| | - Genzo Iguchi
- Department of Diabetes and Endocrinology, Kobe University Hospital, Hyogo, Japan
| | - Yukiko Odake
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Kenichi Yoshida
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Hironori Bando
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Kentaro Suda
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Hitoshi Nishizawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Michiko Takahashi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Yutaka Takahashi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Hyogo, Japan.
| |
Collapse
|
38
|
|
39
|
D'Onofrio N, Servillo L, Balestrieri ML. SIRT1 and SIRT6 Signaling Pathways in Cardiovascular Disease Protection. Antioxid Redox Signal 2018; 28:711-732. [PMID: 28661724 PMCID: PMC5824538 DOI: 10.1089/ars.2017.7178] [Citation(s) in RCA: 280] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Oxidative stress represents the common hallmark of pathological conditions associated with cardiovascular disease (CVD), including atherosclerosis, heart failure, hypertension, aging, diabetes, and other vascular system-related diseases. The sirtuin (SIRT) family, comprising seven proteins (SIRT1-SIRT7) sharing a highly conserved nicotinamide adenine dinucleotide (NAD+)-binding catalytic domain, attracted a great attention for the past few years as stress adaptor and epigenetic enzymes involved in the cellular events controlling aging-related disorder, cancer, and CVD. Recent Advances: Among sirtuins, SIRT1 and SIRT6 are the best characterized for their protective roles against inflammation, vascular aging, heart disease, and atherosclerotic plaque development. This latest role has been only recently unveiled for SIRT6. Of interest, in recent years, complex signaling networks controlled by SIRT1 and SIRT6 common to stress resistance, vascular aging, and CVD have emerged. CRITICAL ISSUES We provide a comprehensive overview of recent developments on the molecular signaling pathways controlled by SIRT1 and SIRT6, two post-translational modifiers proven to be valuable tools to dampen inflammation and oxidative stress at the cardiovascular level. FUTURE DIRECTIONS A deeper understanding of the epigenetic mechanisms through which SIRT1 and SIRT6 act in the signalings responsible for onset and development CVD is a prime scientific endeavor of the upcoming years. Multiple "omic" technologies will have widespread implications in understanding such mechanisms, speeding up the achievement of selective and efficient pharmacological modulation of sirtuins for future applications in the prevention and treatment of CVD. Antioxid. Redox Signal. 28, 711-732.
Collapse
Affiliation(s)
- Nunzia D'Onofrio
- Department of Biochemistry, Biophysics and General Pathology, School of Medicine and Surgery, Università degli Studi della Campania , Naples, Italy
| | - Luigi Servillo
- Department of Biochemistry, Biophysics and General Pathology, School of Medicine and Surgery, Università degli Studi della Campania , Naples, Italy
| | - Maria Luisa Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, School of Medicine and Surgery, Università degli Studi della Campania , Naples, Italy
| |
Collapse
|
40
|
Abstract
Heart diseases are major causes of mortality. Cardiac hypertrophy, myocardial infarction (MI), viral cardiomyopathy, ischemic and reperfusion (I/R) heart injury finally lead to heart failure and death. Insulin and IGF1 signal pathways play key roles in normal cardiomyocyte growth and physiological cardiac hypertrophy while inflammatory signal pathway is associated with pathological cardiac hypertrophy, MI, viral cardiomyopathy, I/R heart injury, and heart failure. Adapter proteins are the major family proteins, which transduce signals from insulin, IGF1, or cytokine receptors to the downstream pathways and have been shown to regulate variety of heart diseases. Here, we summarized the recent advances in understanding the physiological and pathological roles of adapter proteins in heart failure.
Collapse
Affiliation(s)
- Li Tao
- Cardiovascular Center, 305 Hospital of People's Liberation Army, Beijing, 100017, China
| | - Linna Jia
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, Jilin, China
| | - Yuntian Li
- Cardiovascular Center, 305 Hospital of People's Liberation Army, Beijing, 100017, China
| | - Chengyun Song
- Cardiovascular Center, 305 Hospital of People's Liberation Army, Beijing, 100017, China.
| | - Zheng Chen
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, Jilin, China.
| |
Collapse
|
41
|
Xing Z, Pan W, Zhang J, Xu X, Zhang X, He X, Fan M. Hydrogen Rich Water Attenuates Renal Injury and Fibrosis by Regulation Transforming Growth Factor-β Induced Sirt1. Biol Pharm Bull 2018; 40:610-615. [PMID: 28458345 DOI: 10.1248/bpb.b16-00832] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The current research was designed to study the role of hydrogen in renal fibrosis and the renal epithelial to mesenchymal transition (EMT) induced by transforming growth factor-β1 (TGF-β1). Hydrogen rich water (HW) was used to treat animal and cell models. Unilateral ureteral obstruction (UUO) was performed on Balb/c mice to create a model of renal fibrosis. Human kidney proximal tubular epithelial cells (HK-2 cells) were treated with TGF-β1 for 36 h to induce EMT. Serum creatinine (Scr) and blood urea nitrogen (BUN) were measured to test renal function, in addition, kidney histology and immunohistochemical staining of alpha-smooth muscle actin (α-SMA) positive cells was performed to examine the morphological changes. The treatment with UUO induced a robust fibrosis of renal interstitium, shrink of glomerulus and partial fracture of basement membrane. Renal function was also impaired in the experimental group with UUO, with an increase of Scr and BUN in serum. After that, Western-blot was performed to examine the expression of α-SMA, fibronectin, E-cadherin, Smad2 and Sirtuin-1 (Sirt1). The treatment with HW attenuated the development of fibrosis and deterioration of renal function in UUO model. In HK-2 cells, the pretreatment of HW abolished EMT induced by TGF-β1. The down-regulation the expression of Sirt1 induced by TGF-β1 which was dampened by the treatment with HW. Sirtinol, a Sirt1 inhibitor, reversed the effect of HW on EMT induced by TGF-β1. HW can inhibit the development of fibrosis in kidney and prevents HK-2 cells from undergoing EMT which is mediated through Sirt1, a downstream molecule of TGF-β1.
Collapse
Affiliation(s)
- Zhaoyu Xing
- Department of Urology, The Third Affiliated Hospital of Soochow University
| | - Wanma Pan
- Department of Pharmacology, School of Pharmacy, Fudan University
| | - Jing Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University
| | - Xianlin Xu
- Department of Urology, The Third Affiliated Hospital of Soochow University
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University
| | - Xiaozhou He
- Department of Urology, The Third Affiliated Hospital of Soochow University
| | - Min Fan
- Department of Urology, The Third Affiliated Hospital of Soochow University
| |
Collapse
|
42
|
Choupani J, Mansoori Derakhshan S, Bayat S, Alivand MR, Shekari Khaniani M. Narrower insight to SIRT1 role in cancer: A potential therapeutic target to control epithelial-mesenchymal transition in cancer cells. J Cell Physiol 2018; 233:4443-4457. [PMID: 29194618 DOI: 10.1002/jcp.26302] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is a highly networked cellular process which involves cell transition from the immotile epithelial to the motile mesenchymal phenotype, whereby cells lose their cell-cell adhesion and cell polarity. This important process is one of the underlying mechanisms for enabling invasion and metastasis of cancer cells which is considered as malignant phase of tumor progression. However, the molecular mechanisms of this process are not fully clarified. It is reported that Sirtuin1 (SIRT1), a NAD+ dependent class III histone deacetylase is associated with tumor metastasis through positive regulation of EMT in several types of cancers. Recent studies confirmed that up and down regulation of SIRT1 expression remarkably change the migration ability of different cancer cells in vitro and tumor metastasis in vivo. Also, according to this fact that carcinomas as the main human solid tumors, originate from different epithelial cell types, SIRT1 role in EMT has received a great attention due to its potential role in tumor development and metastasis. Therefore, SIRT1 has been proposed as a key regulator of cancer metastasis by promoting EMT, although little is known about the cleared effect of SIRT1 in this transition. Our aim in this review is to explain in more detail the role of SIRT1 in various signaling pathways related to carcinogenesis, with the focus on the promoting role of SIRT1 in EMT as a potential therapeutic target to control EMT and to prevent cancer progression.
Collapse
Affiliation(s)
- Jalal Choupani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sima Mansoori Derakhshan
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Bayat
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahmoud Shekari Khaniani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
43
|
Lewis FC, Kumar SD, Ellison-Hughes GM. Non-invasive strategies for stimulating endogenous repair and regenerative mechanisms in the damaged heart. Pharmacol Res 2018; 127:33-40. [DOI: 10.1016/j.phrs.2017.08.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 08/14/2017] [Accepted: 08/30/2017] [Indexed: 01/04/2023]
|
44
|
Lin N, Cai Y, Zhang L, Chen Y. Identification of key genes associated with congenital heart defects in embryos of diabetic mice. Mol Med Rep 2017; 17:3697-3707. [PMID: 29286097 PMCID: PMC5802176 DOI: 10.3892/mmr.2017.8330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022] Open
Abstract
Maternal diabetes has been reported to be a critical factor for congenital heart defects (CHD) in offspring. The present study aimed to screen the key genes that may be involved in CHD in offspring of diabetic mothers. The present study obtained the gene expression profile of GSE32078, including three embryonic heart tissue samples at embryonic day 13.5 (E13.5), three embryonic heart tissue samples at embryonic day 15.5 (E15.5) from diabetic mice and their respective controls from normal mice. The cut-off criterion of P<0.08 was set to screen differentially expressed genes (DEGs). Their enrichment functions were predicted by Gene Ontology. The enriched pathways were forecasted by Kyoto Encyclopedia of Genes and Genomes and Reactome analysis. Protein-protein interaction (PPI) networks for DEGs were constructed using Cytoscape. The present study identified 869 and 802 DEGs in E13.5 group and E15.5 group, respectively and 182 DEGs were shared by the two developmental stages. The pathway enrichment analysis results revealed that DEGs including intercellular adhesion molecule 1 (Icam1) and H2-M9 were enriched in cell adhesion molecules; DEGs including bone morphogenetic protein receptor type 1A, transforming growth factor β receptor 1 and SMAD specific E3 ubiquitin protein ligase 1 were enriched in the tumor growth factor-β signaling pathway. In addition, DEGs including Icam1, C1s and Fc fragment of IgG receptor IIb were enriched in Staphylococcus aureus infection. Furthermore, the shared DEGs including Icam1, nuclear receptor corepressor 1 (Ncor1) and AKT serine/threonine kinase 3 (Akt3) had high connectivity degrees in the PPI network. The shared DEGs including Icam1, Ncor1 and Akt3 may be important in the cardiogenesis of embryos. These genes may be involved in the development of CHD in the offspring of diabetic mothers.
Collapse
Affiliation(s)
- Nan Lin
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yan Cai
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Linlin Zhang
- Gastroenterology Department, Harbin The First Hospital, Harbin, Heilongjiang 150001, P.R. China
| | - Yahang Chen
- Department of Obstetrics and Gynecology, The Hospital of Heilongjiang, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
45
|
Nuclear sirtuins and inflammatory signaling pathways. Cytokine Growth Factor Rev 2017; 38:98-105. [DOI: 10.1016/j.cytogfr.2017.11.001] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 02/06/2023]
|
46
|
Oxidative stress promotes SIRT1 recruitment to the GADD34/PP1α complex to activate its deacetylase function. Cell Death Differ 2017; 25:255-267. [PMID: 28984870 DOI: 10.1038/cdd.2017.152] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/01/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022] Open
Abstract
Phosphorylation of the eukaryotic translation initiation factor, eIF2α, by stress-activated protein kinases and dephosphorylation by the growth arrest and DNA damage-inducible protein (GADD34)-containing phosphatase is a central node in the integrated stress response. Mass spectrometry demonstrated GADD34 acetylation at multiple lysines. Substituting K315 and K322 with alanines or glutamines did not impair GADD34's ability to recruit protein phosphatase 1α (PP1α) or eIF2α, suggesting that GADD34 acetylation did not modulate eIF2α phosphatase activity. Arsenite (Ars)-induced oxidative stress increased cellular GADD34 levels and enhanced Sirtuin 1 (SIRT1) recruitment to assemble a cytoplasmic complex containing GADD34, PP1α, eIF2α and SIRT1. Induction of GADD34 in WT MEFs paralleled the dephosphorylation of eIF2α (phosphoserine-51) and SIRT1 (phosphoserine-47). By comparison, eIF2α and SIRT1 were persistently phosphorylated in Ars-treated GADD34-/- MEFs. Expressing WT GADD34, but not a mutant unable to bind PP1α in GADD34-/- MEFs restored both eIF2α and SIRT1 dephosphorylation. SIRT1 dephosphorylation increased its deacetylase activity, measured in vitro and in cells. Loss of function of GADD34 or SIRT1 enhanced cellular p-eIF2α levels and attenuated cell death following Ars exposure. These results highlighted a novel role for the GADD34/PP1α complex in coordinating the dephosphorylation and reactivation of eIF2α and SIRT1 to determine cell fate following oxidative stress.
Collapse
|
47
|
Insulin-like growth factor-1 signaling in cardiac aging. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1931-1938. [PMID: 28847512 DOI: 10.1016/j.bbadis.2017.08.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in most developed countries. Aging is associated with enhanced risk of CVD. Insulin-like growth factor-1 (IGF-1) binds to its cognate receptor, IGF-1 receptor (IGF-1R), and exerts pleiotropic effects on cell growth, differentiation, development, and tissue repair. Importantly, IGF-1/IGF-1R signaling is implicated in cardiac aging and longevity. Cardiac aging is an intrinsic process that results in cardiac dysfunction, accompanied by molecular and cellular changes. In this review, we summarize the current state of knowledge regarding the link between the IGF-1/IGF-1R system and cardiac aging. The biological effects of IGF-1R and insulin receptor will be discussed and compared. Furthermore, we describe data regarding how deletion of IGF-1R in cardiomyocytes of aged knockout mice may delay the development of senescence-associated myocardial pathologies. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
|
48
|
Charles S, Raj V, Arokiaraj J, Mala K. Caveolin1/protein arginine methyltransferase1/sirtuin1 axis as a potential target against endothelial dysfunction. Pharmacol Res 2017; 119:1-11. [PMID: 28126510 DOI: 10.1016/j.phrs.2017.01.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/20/2016] [Accepted: 01/22/2017] [Indexed: 12/23/2022]
Abstract
Endothelial dysfunction (ED), an established response to cardiovascular risk factors, is characterized by increased levels of soluble molecules secreted by endothelial cells (EC). Evidence suggest that ED is an independent predictor of cardiac events and that it is associated with a deficiency in production or bioavailability of nitric oxide (NO) and/or an imbalance in the relative contribution of endothelium-derived relaxing and contracting factors. ED can be reversed by treating cardiovascular risk factors, hence, beyond ambiguity, ED contributes to initiation and progression of atherosclerotic disease. Majority of cardiovascular risk factors act by a common pathway, oxidative stress (OS), characterized by an imbalance in bioavailability of NO and reactive oxygen species (ROS). Enhanced ROS, through several mechanisms, alters competence of EC that leads to ED, reducing its potential to maintain homeostasis and resulting in development of cardiovascular disease (CVD). Influential mechanisms that have been implicated in the development of ED include (i) presence of elevated levels of NOS inhibitor, asymmetric dimethylarginine (ADMA) due to augmented enzyme activity of protein arginine methyl transferase-1 (PRMT1); (ii) decrease in NO generation by endothelial nitric oxide synthase (eNOS) uncoupling, or by reaction of NO with free radicals and (iii) impaired post translational modification of protein (PTM) such as eNOS, caveolin-1 (cav1) and sirtuin-1 (SIRT1). However, the inter-related mechanisms that concur to developing ED is yet to be understood. The events that possibly overlay include OS-induced sequestration of SIRT1 to caveolae facilitating cav1-SIRT1 association; potential increase in lysine acetylation of enzymes such as eNOS and PRMT1 leading to enhanced ADMA formation; imbalance in acetylation-methylation ratio (AMR); diminished NO generation and ED. Here we review current literature from research showing interdependent association between cav1-PRMT1-SIRT1 to the outcomes of experimental and clinical research aiming to preserve endothelial function with gene- or pharmaco-therapy.
Collapse
Affiliation(s)
- Soniya Charles
- Department of Biotechnology, School of Bioengineering, SRM University, Potheri 603203, Tamil Nadu, India
| | - Vijay Raj
- Medical College Hospital and Research Center, SRM University, Potheri 603203, Tamil Nadu, India
| | - Jesu Arokiaraj
- Department of Biotechnology, Faculty of Science and Humanities, SRM University, Potheri 603203, Tamil Nadu, India
| | - Kanchana Mala
- Medical College Hospital and Research Center, SRM University, Potheri 603203, Tamil Nadu, India.
| |
Collapse
|
49
|
Tzanis G, Philippou A, Karatzanos E, Dimopoulos S, Kaldara E, Nana E, Pitsolis T, Rontogianni D, Koutsilieris M, Nanas S. Effects of High-Intensity Interval Exercise Training on Skeletal Myopathy of Chronic Heart Failure. J Card Fail 2017; 23:36-46. [PMID: 27327970 DOI: 10.1016/j.cardfail.2016.06.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 06/11/2016] [Accepted: 06/16/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND It remains controversial which type of exercise elicits optimum adaptations on skeletal myopathy of heart failure (HF). Our aim was to evaluate the effect of high-intensity interval training (HIIT), with or without the addition of strength training, on skeletal muscle of HF patients. METHODS AND RESULTS Thirteen male HF patients (age 51 ± 13 years, body mass index 27 ± 4 kg/m2) participated in either an HIIT (AER) or an HIIT combined with strength training (COM) 3-month program. Biopsy samples were obtained from the vastus lateralis. Analyses were performed on muscle fiber type, cross-section area (CSA), capillary density, and mRNA expression of insulin-like growth factor (IGF) 1 isoforms (ie, IGF-1Ea, IGF-1Eb, IGF-1Ec), type-1 receptor (IGF-1R), and binding protein 3 (IGFBP-3). Increased expression of IGF-1Ea, IGF-1Eb, IGF-1Ec, and IGFBP-3 transcripts was found (1.7 ± 0.8, 1.5 ± 0.8, 2.0 ± 1.32.4 ± 1.4 fold changes, respectively; P < .05). Type I fibers increased by 21% (42 ± 10% to 51 ± 7%; P < .001) and capillary/fiber ratio increased by 24% (1.27 ± 0.22 to 1.57 ± 0.41; P = .005) in both groups as a whole. Fibers' mean CSA increased by 10% in total, but the increase in type I fibers' CSA was greater after AER than COM (15% vs 6%; P < .05). The increased CSA correlated with the increased expression of IGF-1Ea and IGF-1Εb. CONCLUSIONS HIIT reverses skeletal myopathy of HF patients, with the adaptive responses of the IGF-1 bioregulation system possibly contributing to these effects. AER program seemed to be superior to COM to induce muscle hypertrophy.
Collapse
Affiliation(s)
- Georgios Tzanis
- 1st Critical Care Medicine Department, Cardiopulmonary Exercise Testing and Rehabilitation Laboratory, "Evgenidio Hospital," National & Kapodistrian University of Athens, Greece
| | - Anastassios Philippou
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Eleftherios Karatzanos
- 1st Critical Care Medicine Department, Cardiopulmonary Exercise Testing and Rehabilitation Laboratory, "Evgenidio Hospital," National & Kapodistrian University of Athens, Greece
| | - Stavros Dimopoulos
- 1st Critical Care Medicine Department, Cardiopulmonary Exercise Testing and Rehabilitation Laboratory, "Evgenidio Hospital," National & Kapodistrian University of Athens, Greece
| | - Elisavet Kaldara
- 3rd Cardiology Department, "Laiko" Hospital, National and Kapodistrian University of Athens, Greece
| | - Emmeleia Nana
- 3rd Cardiology Department, "Laiko" Hospital, National and Kapodistrian University of Athens, Greece
| | - Theodoros Pitsolis
- 1st Critical Care Medicine Department, Cardiopulmonary Exercise Testing and Rehabilitation Laboratory, "Evgenidio Hospital," National & Kapodistrian University of Athens, Greece
| | | | - Michael Koutsilieris
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Serafim Nanas
- 1st Critical Care Medicine Department, Cardiopulmonary Exercise Testing and Rehabilitation Laboratory, "Evgenidio Hospital," National & Kapodistrian University of Athens, Greece.
| |
Collapse
|
50
|
Kong D, Zhan Y, Liu Z, Ding T, Li M, Yu H, Zhang L, Li H, Luo A, Zhang D, Wang Y, Wang S, Zhang Z, Zhang H, Huang X, Yao P, Ding Y, Liu Z. SIRT1-mediated ERβ suppression in the endothelium contributes to vascular aging. Aging Cell 2016; 15:1092-1102. [PMID: 27470296 PMCID: PMC6398526 DOI: 10.1111/acel.12515] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2016] [Indexed: 01/23/2023] Open
Abstract
SIRT1 has many important molecular functions in aging, and the estrogen receptors (ERs) have a vasculoprotective effect, although the detailed mechanism for the roles of SIRT1 and ERs in vascular aging remains unclear. We found that ERβ expression in the endothelium was reduced in aging mice, and the expression of ERα and SIRT1 did not change, while SIRT1 activity declined. Further investigation showed that the ERβ expression was regulated by SIRT1 through complexes of SIRT1‐PPARγ/RXR‐p300 that bind to a PPRE (PPAR response element) site on the ERβ promoter, and the declined SIRT1 function in aging mice was due to compromised phosphorylation at S154. A single‐mutant SIRT1‐C152(D) restored the reduced ERβ expression in the endothelium with minimized reactive oxygen species generation and DNA damage and increased mitochondrial function and fatty acid metabolism. In high‐fat diet aging mice, the endothelium‐specific delivery of ERβ or SIRT1‐C152(D) on the vascular wall reduced the circulating lipids with ameliorated vascular damage, including the restored vessel tension and blood pressure. We conclude that SIRT1‐mediated ERβ suppression in the endothelium contributes to vascular aging, and the modulation of SIRT1 phosphorylation through a single‐mutant SIRT1‐C152(D) restores this effect.
Collapse
Affiliation(s)
- Danli Kong
- School of Public Health Guangdong Medical College Dongguan 523808 China
| | - Ying Zhan
- Tongji Wenchang Hospital Huazhong University of Science and Technology Wenchang 571321 China
- Tongji Hospital Huazhong University of Science and Technology Wuhan 430030 China
| | - Zhaoyu Liu
- Tongji Hospital Huazhong University of Science and Technology Wuhan 430030 China
| | - Ting Ding
- Tongji Hospital Huazhong University of Science and Technology Wuhan 430030 China
| | - Min Li
- Tongji Wenchang Hospital Huazhong University of Science and Technology Wenchang 571321 China
- Inner Mongolia University for the Nationalities #1742 Huolinhe Str. Tongliao Inner Mongolia 028000 China
| | - Haibing Yu
- School of Public Health Guangdong Medical College Dongguan 523808 China
| | - Laxi Zhang
- Tongji Hospital Huazhong University of Science and Technology Wuhan 430030 China
| | - Huawen Li
- School of Public Health Guangdong Medical College Dongguan 523808 China
| | - Aiyue Luo
- Tongji Hospital Huazhong University of Science and Technology Wuhan 430030 China
| | - Dongwei Zhang
- Inner Mongolia University for the Nationalities #1742 Huolinhe Str. Tongliao Inner Mongolia 028000 China
| | - Yifei Wang
- Guangzhou Biomedical Research and Development Center Jinan University Guangzhou 510632 China
| | - Shixuan Wang
- Tongji Hospital Huazhong University of Science and Technology Wuhan 430030 China
| | - Zhefan Zhang
- Personalized Treatment Research Center The Third Hospital of Wuhan Wuhan 430060 China
| | - Hongyu Zhang
- Department of Hematology Peking University ShenZhen Hospital ShenZhen 518036 China
| | - Xiaodong Huang
- Personalized Treatment Research Center The Third Hospital of Wuhan Wuhan 430060 China
| | - Paul Yao
- School of Public Health Guangdong Medical College Dongguan 523808 China
- Tongji Wenchang Hospital Huazhong University of Science and Technology Wenchang 571321 China
| | - Yuanling Ding
- School of Public Health Guangdong Medical College Dongguan 523808 China
| | - Zhengxiang Liu
- Tongji Wenchang Hospital Huazhong University of Science and Technology Wenchang 571321 China
| |
Collapse
|