1
|
Xu Z, Song T, Yang X, Cong L, Yin L, Xu Y, Han X, Gao M, Xu L. TMT-based proteomics reveals methylprotodioscin alleviates oxidative stress and inflammation via COX6C in myocardial infraction. Biomed Pharmacother 2024; 180:117489. [PMID: 39321507 DOI: 10.1016/j.biopha.2024.117489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/04/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024] Open
Abstract
The effect of methylprotodioscin (MPD), a steroidal saponin obtained from medicinal plants, on myocardial infarction (MI) remains elusive. In this study, HL-1 and AC16 cells were subjected to injury induced by hypoxic environment, and a mouse model of MI was established by ligating the left anterior descending. MPD significantly increased viabilities and proliferations, improved the stability of MMP, reduced ROS and inflammatory factor levels in hypoxia cardiomyocytes. Moreover, MPD significantly improved cardiac functions, increased the ventricular ejection fraction and short axis shortening rate of mice with MI, reduced the infarction area, alleviated oxidative stress and increased ATPase activities. Then, differentially expressed proteins (DEPs) were discovered and evaluated using tandem mass tag (TMT)-based proteomics and bioinformatics approaches. Compared with sham group, there were 420 DEPs in the cardiac tissue of MI group, likewise, 163 DEPs in MPD group were identified compared to MI group. By validating, the expression of COX6C was elevated in MI group and declined in MPD groups, consistent with the TMT-based proteomics results. Correspondingly, p-NF-κB expression was downregulated, while Nrf2 and SOD expressions were upregulated by MPD. Moreover, si-COX6C transfection blocked the regulatory effects of MPD on COX6C-mediated inflammation and oxidative stress in MI. Our findings indicate that MPD, a naturally occurring active ingredient, could effectively improve cardiac function. Its ability may result from regulating COX6C to reduce oxidative stress and suppress inflammation, suggesting that MPD is very attractive for the treatment of MI.
Collapse
Affiliation(s)
- Zhihui Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Tingyu Song
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xiufang Yang
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Linhao Cong
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Meng Gao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
2
|
Singh KA, Soukar J, Zulkifli M, Kersey A, Lokhande G, Ghosh S, Murali A, Garza NM, Kaur H, Keeney JN, Banavath R, Ceylan Koydemir H, Sitcheran R, Singh I, Gohil VM, Gaharwar AK. Atomic vacancies of molybdenum disulfide nanoparticles stimulate mitochondrial biogenesis. Nat Commun 2024; 15:8136. [PMID: 39289340 PMCID: PMC11408498 DOI: 10.1038/s41467-024-52276-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
Diminished mitochondrial function underlies many rare inborn errors of energy metabolism and contributes to more common age-associated metabolic and neurodegenerative disorders. Thus, boosting mitochondrial biogenesis has been proposed as a potential therapeutic approach for these diseases; however, currently we have a limited arsenal of compounds that can stimulate mitochondrial function. In this study, we designed molybdenum disulfide (MoS2) nanoflowers with predefined atomic vacancies that are fabricated by self-assembly of individual two-dimensional MoS2 nanosheets. Treatment of mammalian cells with MoS2 nanoflowers increased mitochondrial biogenesis by induction of PGC-1α and TFAM, which resulted in increased mitochondrial DNA copy number, enhanced expression of nuclear and mitochondrial-DNA encoded genes, and increased levels of mitochondrial respiratory chain proteins. Consistent with increased mitochondrial biogenesis, treatment with MoS2 nanoflowers enhanced mitochondrial respiratory capacity and adenosine triphosphate production in multiple mammalian cell types. Taken together, this study reveals that predefined atomic vacancies in MoS2 nanoflowers stimulate mitochondrial function by upregulating the expression of genes required for mitochondrial biogenesis.
Collapse
Affiliation(s)
- Kanwar Abhay Singh
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
| | - John Soukar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA
| | - Mohammad Zulkifli
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - Anna Kersey
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, USA
| | - Giriraj Lokhande
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
| | - Sagnika Ghosh
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - Aparna Murali
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, USA
| | - Natalie M Garza
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - Harman Kaur
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA
| | - Justin N Keeney
- Department of Cell biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Ramu Banavath
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, USA
| | - Hatice Ceylan Koydemir
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, USA
| | - Raquel Sitcheran
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA
- Department of Cell biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Irtisha Singh
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
- Department of Cell biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA.
| | - Vishal M Gohil
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
- Department of Biochemistry and Biophysics, College of Agriculture & Life Sciences, Texas A&M University, College Station, TX, USA.
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
- Interdisiplinary program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
- Department of Cell biology and Genetics, College of Medicine, Texas A&M University, Bryan, TX, USA.
- Department of Material Science and Engineering, College of Engineering, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
3
|
Yu W, Peng X, Cai X, Xu H, Wang C, Liu F, Luo D, Tang S, Wang Y, Du X, Gao Y, Tian T, Liang S, Chen C, Kim NH, Yuan B, Zhang J, Jiang H. Transcriptome analysis of porcine oocytes during postovulatory aging. Theriogenology 2024; 226:387-399. [PMID: 38821784 DOI: 10.1016/j.theriogenology.2024.05.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Decreased oocyte quality is a significant contributor to the decline in female fertility that accompanies aging in mammals. Oocytes rely on mRNA stores to support their survival and integrity during the protracted period of transcriptional dormancy as they await ovulation. However, the changes in mRNA levels and interactions that occur during porcine oocyte maturation and aging remain unclear. In this study, the mRNA expression profiles of porcine oocytes during the GV, MII, and aging (24 h after the MII stage) stages were explored by transcriptome sequencing to identify the key genes and pathways that affect oocyte maturation and postovulatory aging. The results showed that 10,929 genes were coexpressed in porcine oocytes during the GV stage, MII stage, and aging stage. In addition, 3037 genes were expressed only in the GV stage, 535 genes were expressed only in the MII stage, and 120 genes were expressed only in the aging stage. The correlation index between the GV and MII stages (0.535) was markedly lower than that between the MII and aging stages (0.942). A total of 3237 genes, which included 1408 upregulated and 1829 downregulated genes, were differentially expressed during porcine oocyte postovulatory aging (aging stage vs. MII stage). Key functional genes, including ATP2A1, ATP2A3, ATP2B2, NDUFS1, NDUFA2, NDUFAF3, SREBF1, CYP11A1, CYP3A29, GPx4, CCP110, STMN1, SPC25, Sirt2, SYCP3, Fascin1/2, PFN1, Cofilin, Tmod3, FLNA, LRKK2, CHEK1/2, DDB1/2, DDIT4L, and TONSL, and key molecular pathways, such as the calcium signaling pathway, MAPK signaling pathway, TGF-β signaling pathway, PI3K/Akt signaling pathway, FoxO signaling pathway, gap junctions, and thermogenesis, were found in abundance during porcine postovulatory aging. These genes are mainly involved in the regulation of many biological processes, such as oxidative stress, calcium homeostasis, mitochondrial function, and lipid peroxidation, during porcine oocyte postovulatory aging. These results contribute to a more in-depth understanding of the biological changes, key regulatory genes and related biological pathways that are involved in oocyte aging and provide a theoretical basis for improving the efficiency of porcine embryo production in vitro and in vivo.
Collapse
Affiliation(s)
- Wenjie Yu
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Xinyue Peng
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Xiaoshi Cai
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Hong Xu
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Chen Wang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Fengjiao Liu
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Dan Luo
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Shuhan Tang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Yue Wang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Xiaoxue Du
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Yan Gao
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Tian Tian
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China; Center of Reproductive Medicine & Center of Prenatal Diagnosis, First Hospital, Jilin University, Changchun, 130062, Jilin, China
| | - Shuang Liang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Chengzhen Chen
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Nam-Hyung Kim
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Bao Yuan
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Jiabao Zhang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China
| | - Hao Jiang
- College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, China.
| |
Collapse
|
4
|
Xie Y, Chen S, Sheng L, Sun Y, Liu S. A New Landscape of Human Dental Aging: Causes, Consequences, and Intervention Avenues. Aging Dis 2023:AD.2022.1224. [PMID: 37163430 PMCID: PMC10389823 DOI: 10.14336/ad.2022.1224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/24/2022] [Indexed: 05/12/2023] Open
Abstract
Aging is accompanied by physical dysfunction and physiologic degeneration that occurs over an individual's lifetime. Human teeth, like many other organs, inevitably undergo chronological aging and age-related changes throughout the lifespan, resulting in a substantial need for preventive, restorative as well as periodontal dental care. This is particularly the case for seniors at 65 years of age and those older but economically disadvantaged. Dental aging not only interferes with normal chewing and digestion, but also affects daily appearance and interpersonal communications. Further dental aging can incur the case of multiple disorders such as oral cancer, encephalitis, and other systemic diseases. In the next decades or even hundreds of years, the proportion of the elderly in the global population will continue to rise, a tendency that attracts increasing attention across multiple scientific and medical disciplines. Dental aging will bring a variety of problems to the elderly themselves and poses serious challenges to the medical profession and social system. A reduced, but functional dentition comprising 20 teeth in occlusion has been proposed as a measurement index of successful dental aging. Healthy dental aging is critical to healthy aging, from both medical and social perspectives. To date, biomedical research on the causes, processes and regulatory mechanisms of dental aging is still in its infancy. In this article, updated insights into typical manifestations, associated pathologies, preventive strategies and molecular changes of dental aging are provided, with future research directions largely projected.
Collapse
Affiliation(s)
- Yajia Xie
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
- Department of Endodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Shuang Chen
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Lu Sheng
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, China
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA 98195, USA
| | - Shangfeng Liu
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Li Y, Liu X, Wan L, Han B, Ma S, Pan H, Wei J, Cui X. Metformin suppresses cardiac fibroblast proliferation under high-glucose conditions via regulating the mitochondrial complex I protein Grim-19 involved in the Sirt1/Stat3 signaling pathway. Free Radic Biol Med 2023; 206:1-12. [PMID: 37353174 DOI: 10.1016/j.freeradbiomed.2023.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/23/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
Hyperglycemia associated with myocardial oxidative stress and fibrosis is the main cause of diabetic cardiomyopathy. Currently, no approved drug is available for preventing or treating diabetes-induced cardiac fibrosis. Metformin has been reported to improve glycemic control and ameliorate diabetic cardiomyopathy. This study aimed to investigate the effects and mechanism of metformin on diabetes-induced cardiac fibrosis and high glucose-induced proliferation of cardiac fibroblasts (CFs). In this study, db/db mice were treated with metformin [250 mg/kg⋅d, gavage]. CFs were cultured in high-glucose medium to mimic an in vitro diabetes model and then subjected to treatment with or without metformin. Cardiac fibrosis was analyzed using immunohistochemistry, Masson's trichrome staining, and Western blot analysis. Cell Counting Kit-8 (CCK-8) assays and cell colony formation assays were used to examine cell proliferation capacity. Transwell and scratch-wound assays were used to detect the migration ability of CFs. Retinoid-interferon-induced mortality-19 (Grim-19), sirtuin1 (Sirt1), and signal transducer and activator of transcription 3 (Stat3) were detected using Western blot analysis. The genes downstream of the Stat3 pathway were detected using quantitative reverse transcription PCR (qRT‒PCR). Metformin treatment markedly attenuated cardiac fibrosis in db/db mice and the proliferation and migration of CFs under high-glucose conditions. Mechanistically, we found an intersection between metformin and Grim-19 using bioinformatics. Metformin was found to suppress the expression of p-Stat3 and elevate the expression of mitochondrial complex I protein Grim-19 and Sirt1, thus inhibiting the proliferation and migration of CFs under high-glucose conditions. Our data suggested that metformin inhibited the proliferation and migration of CFs by regulating the expression of mitochondrial complex I Grim-19 protein involved in the Sirt1/Stat3 signaling pathway under high-glucose conditions, thus providing new ideas for treating diabetes-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Yongguang Li
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Xiangdong Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200000, People's Republic of China
| | - Lili Wan
- Division of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Beibei Han
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Shixin Ma
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China
| | - Hongyuan Pan
- Saint Paul's School, 325 Pleasant Street, Concord, NH, 03301, USA
| | - Junbo Wei
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 600 Yishan Road, Shanghai, 200233, People's Republic of China; Department of Cardiology, Renhe Hospital, 1999 Changjiang West Road, Shanghai, 200431, People's Republic of China.
| | - Xiaofang Cui
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
6
|
Tang Y, Lei S, Wang S, Lu H, Li H, Lv J, Ge RS, Ying Y. Leydig cell development in pubertal male rats is blocked by perfluorotetradecanoic acid through decreasing AMPK-mTOR-autophagy pathway. Toxicol Lett 2023:S0378-4274(23)00194-7. [PMID: 37269911 DOI: 10.1016/j.toxlet.2023.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023]
Abstract
Perfluorotetradecanoic acid (PFTeDA) is a type of perfluoroalkyl acid that has been linked to various health effects in animals and humans. The study aimed to investigate the potential impact of PFTeDA exposure on Leydig cell development in rats during puberty. Understanding the effects of PFTeDA on Leydig cells is crucial as these cells play a significant role in male reproductive function. Male Sprague-Dawley rats were gavaged with PFTeDA at doses of 0, 1, 5, and 10mg/kg/day from postnatal day 35 to 56. The serum hormone levels were measured and testicular transcriptome changes were analyzed by RNA-seq and verified by qPCR, and the levels of steroidogenesis-related proteins and energy regulators were measured. PFTeDA significantly reduced serum testosterone levels while slightly increasing LH levels. RNA-seq and qPCR analysis showed that genes responsive to oxidative phosphorylation (Naufa1 and Ndufs6) and steroidogenesis (Ldlr, Star, Cyp11a1) were markedly downregulated at ≥5mg/kg, while those related to ferroptosis (Alox15) and cell senescence (Map2k3 and RT1-CE3) were significantly upregulated. PFTeDA markedly reduced SIRT1 (silent information regulator 1) /PGC-1α (peroxisome proliferator-activated receptor gamma coactivator-1α) and AMPKA (AMP activated kinase A), LC3B and Beclin1 (biomarkers for autophagy) levels while increasing phosphorylated mTOR. In vitro treatment of PFTeDA at 5 μM significantly reduced androgen output of Leydig cells from 35-day-old male rats while ferrostatin 1 (10 μM) reversed PFTeDA-mediated inhibition. In conclusion, the inhibitory effects of PFTeDA on pubertal rat Leydig cell development are possibly regulated by inducing ferroptosis thereby downregulating SIRT1/AMPKA/ autophagy pathways, eventually resulting in reduced steroidogenesis.
Collapse
Affiliation(s)
- Yunbing Tang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shi Lei
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shaowei Wang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Han Lu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huitao Li
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University; Key Laboratory of Pediatric Anesthesiology, Ministry of Education,Wenzhou Medical University; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University
| | - Jieqiang Lv
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ren-Shan Ge
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Yingfen Ying
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
7
|
Zhao X, Xu H, Li Y, Ma R, Qi Y, Zhang M, Guo C, Sun Z, Li Y. Proteomic profiling reveals dysregulated mitochondrial complex subunits responsible for myocardial toxicity induced by SiNPs. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 857:159206. [PMID: 36198348 DOI: 10.1016/j.scitotenv.2022.159206] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
The relationship between environmental exposure to silica nanoparticles (SiNPs) and adverse cardiac outcomes has received more attention. Our recent work has revealed a size-dependent impact of the intratracheal instilled SiNPs on cardiac health of ApoE-/- mice using nanoscale SiNPs-60 and submicro-sized SiNPs-300, but the underlying mechanism of action still remains unclear. Hence, we identified proteins and protein networks perturbed by SiNPs in myocardial tissues of ApoE-/- mice by using LC-MS/MS-based quantitative proteomics. A set of 435 differentially expressed proteins (DEPs) were screened in response to SiNPs, which mainly enriched in the mitochondria and functioned in cell metabolism, biosynthesis and signal transduction. KEGG analysis showed that DEPs were significantly associated with oxidative phosphorylation and cardiomyopathy. The protein-protein interaction (PPI) network revealed 9 DEPs (e.g., Ndufs1, Ndufv1, Cox4i1) as potential biomarkers of SiNPs-induced myocardial toxicity. Of note, all the 9 candidate proteins were subunits of mitochondria respiratory chain complex, and their expressions were dependent on particle size, which were remarkably down-regulated by SiNPs-60 but not by SiNPs-300. More importantly, the correlation analysis verified the 9 dysregulated mitochondria complex protein subunits strongly correlated to the biochemical and functional indexes of cardiac injury in response to SiNPs. In conclusion, our study firstly provided significant proteomic insights into the potential molecular mechanisms underlying SiNPs-elicited cardiotoxicity, with the dysregulated mitochondrial complex subunits as core regulatory molecules. Overall, our study would provide the scientific basis for the molecular actions and mechanisms of toxicity induced by SiNPs.
Collapse
Affiliation(s)
- Xinying Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Hailin Xu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yan Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Ru Ma
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Yi Qi
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Min Zhang
- Department of Nephrology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Caixia Guo
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yanbo Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
8
|
Resveratrol and FGF1 Synergistically Ameliorates Doxorubicin-Induced Cardiotoxicity via Activation of SIRT1-NRF2 Pathway. Nutrients 2022; 14:nu14194017. [PMID: 36235670 PMCID: PMC9572068 DOI: 10.3390/nu14194017] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Doxorubicin (DOX) has received attention due to dose-dependent cardiotoxicity through abnormal redox cycling. Native fibroblast growth factor 1 (FGF1) is known for its anti-oxidative benefits in cardiovascular diseases, but possesses a potential tumorigenic risk. Coincidentally, the anti-proliferative properties of resveratrol (RES) have attracted attention as alternatives or auxiliary therapy when combined with other chemotherapeutic drugs. Therefore, the purpose of this study is to explore the therapeutic potential and underlying mechanisms of co-treatment of RES and FGF1 in a DOX-treated model. Here, various cancer cells were applied to determine whether RES could antagonize the oncogenesis effect of FGF1. In addition, C57BL/6J mice and H9c2 cells were used to testify the therapeutic potential of a co-treatment of RES and FGF1 against DOX-induced cardiotoxicity. We found RES could reduce the growth-promoting activity of FGF1. Additionally, the co-treatment of RES and FGF1 exhibits a more powerful cardio-antioxidative capacity in a DOX-treated model. The inhibition of SIRT1/NRF2 abolished RES in combination with FGF1 on cardioprotective action. Further mechanism analysis demonstrated that SIRT1 and NRF2 might form a positive feedback loop to perform the protective effect on DOX-induced cardiotoxicity. These favorable anti-oxidative activities and reduced proliferative properties of the co-treatment of RES and FGF1 provided a promising therapy for anthracycline cardiotoxicity during chemotherapy.
Collapse
|
9
|
Wang Q, Yu Q, Wu M. Antioxidant and neuroprotective actions of resveratrol in cerebrovascular diseases. Front Pharmacol 2022; 13:948889. [PMID: 36133823 PMCID: PMC9483202 DOI: 10.3389/fphar.2022.948889] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/01/2022] [Indexed: 11/15/2022] Open
Abstract
Cerebralvascular diseases are the most common high-mortality diseases worldwide. Despite its global prevalence, effective treatments and therapies need to be explored. Given that oxidative stress is an important risk factor involved with cerebral vascular diseases, natural antioxidants and its derivatives can be served as a promising therapeutic strategy. Resveratrol (3, 5, 4′-trihydroxystilbene) is a natural polyphenolic antioxidant found in grape skins, red wine, and berries. As a phytoalexin to protect against oxidative stress, resveratrol has therapeutic value in cerebrovascular diseases mainly by inhibiting excessive reactive oxygen species production, elevating antioxidant enzyme activity, and other antioxidant molecular mechanisms. This review aims to collect novel kinds of literature regarding the protective activities of resveratrol on cerebrovascular diseases, addressing the potential mechanisms underlying the antioxidative activities and mitochondrial protection of resveratrol. We also provide new insights into the chemistry, sources, and bioavailability of resveratrol.
Collapse
Affiliation(s)
- Qing Wang
- Shaanxi Prov Peoples Hospital, Shaanxi Prov Key Lab Infect and Immune Dis, Xian, China
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Qi Yu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
- Department of Histology and Embryology, Xi’an Medical University, Xi’an, China
- Department of Pharmacology, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Min Wu
- Shaanxi Prov Peoples Hospital, Shaanxi Prov Key Lab Infect and Immune Dis, Xian, China
- *Correspondence: Min Wu,
| |
Collapse
|
10
|
Metabolic Determinants in Cardiomyocyte Function and Heart Regenerative Strategies. Metabolites 2022; 12:metabo12060500. [PMID: 35736435 PMCID: PMC9227827 DOI: 10.3390/metabo12060500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Heart disease is the leading cause of mortality in developed countries. The associated pathology is characterized by a loss of cardiomyocytes that leads, eventually, to heart failure. In this context, several cardiac regenerative strategies have been developed, but they still lack clinical effectiveness. The mammalian neonatal heart is capable of substantial regeneration following injury, but this capacity is lost at postnatal stages when cardiomyocytes become terminally differentiated and transit to the fetal metabolic switch. Cardiomyocytes are metabolically versatile cells capable of using an array of fuel sources, and the metabolism of cardiomyocytes suffers extended reprogramming after injury. Apart from energetic sources, metabolites are emerging regulators of epigenetic programs driving cell pluripotency and differentiation. Thus, understanding the metabolic determinants that regulate cardiomyocyte maturation and function is key for unlocking future metabolic interventions for cardiac regeneration. In this review, we will discuss the emerging role of metabolism and nutrient signaling in cardiomyocyte function and repair, as well as whether exploiting this axis could potentiate current cellular regenerative strategies for the mammalian heart.
Collapse
|
11
|
The Radioprotective Activity of Resveratrol—Metabolomic Point of View. Metabolites 2022; 12:metabo12060478. [PMID: 35736411 PMCID: PMC9229206 DOI: 10.3390/metabo12060478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
Resveratrol, a plant-derived polyphenol, is an intensively studied compound with widely documented positive effects on health. Antioxidant activity is the property most often mentioned as responsible for its beneficial effects. Therefore, since the adverse effect of ionizing radiation is primarily related to the induction of oxidative stress, the question arises of whether the use of resveratrol could have a radioprotective effect. This paper summarizes the data on the cytoprotective activity of resveratrol and pieces of evidence for the potential interplay between response to radiation and resveratrol activity. The paper focuses on changes in the metabolic profile of cells and organisms induced by ionizing radiation and exposure to resveratrol. The comparison of metabolic changes induced by both factors provides a rationale for the potential mechanism of the radioprotective effects of resveratrol.
Collapse
|
12
|
Resveratrol exerts antiproliferative effects on high-glucose-cultured vascular smooth muscle cells via inhibition of STAT3 and upregulation of mitochondrial gene GRIM-19 which is responsible for STAT3 activation. Int J Diabetes Dev Ctries 2022. [DOI: 10.1007/s13410-021-00963-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
13
|
Tıraş ZŞE, Okur HH, Günay Z, Yıldırım HK. Different approaches to enhance resveratrol content in wine. CIÊNCIA E TÉCNICA VITIVINÍCOLA 2022. [DOI: 10.1051/ctv/ctv20223701013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Resveratrol is a polyphenol with antioxidant properties and possible beneficial effects on human health. Grapes, peanuts, berries, cacao beans and red wine contain resveratrol. Resveratrol attracts attention due to its bioactive properties, however, the concentration of this compound is not high in grape and wine. Therefore, different studies have been carried out to increase resveratrol level in these products. Several factors such as the grapevine variety, the climatic conditions and the viticultural practices used to create stress on the vine affect the level of resveratrol. Winemaking technologies applied during pre-fermentation, fermentation and post–fermentation stages could also have an effect on the concentration of this stilbene. In addition, recent studies have evaluated biotechnological approaches through the use of different bacteria and yeast strains to produce wine with increased resveratrol content. In this review, the most important factors contributing to increase the resveratrol concentration in grapes and wines are examined. Besides, analytical methods to determine resveratrol content in wine are addressed.
Collapse
|
14
|
Zhang X, Liu X, Wan F, You W, Tan X, Sheng Q, Li C, Hu Z, Liu G, Zhao H. Protective effect of resveratrol against hydrogen peroxide-induced oxidative stress in bovine skeletal muscle cells. Meat Sci 2021; 185:108724. [PMID: 34952489 DOI: 10.1016/j.meatsci.2021.108724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/04/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022]
Abstract
The objective of this study was to investigate the protective effects and the underlying mechanisms of resveratrol (RES) against hydrogen peroxide (H2O2)-induced oxidative stress in bovine skeletal muscle cells (BMCs). Pretreatment of BMCs with RES prior to H2O2 exposure increased cell viability, attenuated reactive oxygen species, and stabilized the redox state. H2O2 exposure activated sirtuin type 1 (SIRT1) and nuclear factor E2-related factor 2 (NRF2)-mediated signaling pathways. Pretreatment with RES did not alter SIRT1-regulated genes but inhibited the upregulation of NRF2, whereas enhanced heme oxygenase 1 (HO-1) expression. Pretreatment with RES prior to H2O2 exposure failed to suppress NRF2 expression when NRF2 was knocked down by RNA interference. However, HO-1 expression still could be induced by RES. These results suggest that RES has benifical effects against oxidative stress. NRF2-mediated pathway play an important role, and HO-1 upregulation is the key process in RES regulation. RES may be used as a therapeutic agent for meat quality improvement in beef cattle.
Collapse
Affiliation(s)
- Xianglun Zhang
- Shandong Academy of Agricultural Sciences Animal Science and Veterinary Medicine Institute, Shandong Key Lab of Animal Disease Control and Breeding, Jinan, China
| | - Xiaomu Liu
- Shandong Academy of Agricultural Sciences Animal Science and Veterinary Medicine Institute, Shandong Key Lab of Animal Disease Control and Breeding, Jinan, China
| | - Fachun Wan
- Shandong Academy of Agricultural Sciences Animal Science and Veterinary Medicine Institute, Shandong Key Lab of Animal Disease Control and Breeding, Jinan, China; College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Wei You
- Shandong Academy of Agricultural Sciences Animal Science and Veterinary Medicine Institute, Shandong Key Lab of Animal Disease Control and Breeding, Jinan, China
| | - Xiuwen Tan
- Shandong Academy of Agricultural Sciences Animal Science and Veterinary Medicine Institute, Shandong Key Lab of Animal Disease Control and Breeding, Jinan, China
| | - Qingkai Sheng
- Shandong Academy of Agricultural Sciences Animal Science and Veterinary Medicine Institute, Shandong Key Lab of Animal Disease Control and Breeding, Jinan, China
| | - Chuanhao Li
- Shandong Academy of Agricultural Sciences Animal Science and Veterinary Medicine Institute, Shandong Key Lab of Animal Disease Control and Breeding, Jinan, China
| | - Zhuran Hu
- Shandong Green and Blue Bio-technology Co. Ltd, Taian, China
| | - Guifen Liu
- Shandong Academy of Agricultural Sciences Animal Science and Veterinary Medicine Institute, Shandong Key Lab of Animal Disease Control and Breeding, Jinan, China.
| | - Hongbo Zhao
- Shandong Academy of Agricultural Sciences Animal Science and Veterinary Medicine Institute, Shandong Key Lab of Animal Disease Control and Breeding, Jinan, China.
| |
Collapse
|
15
|
Huang Y, Wang S, Meng X, Chen N, Li S. Molecular Cloning and Characterization of Sirtuin 1 and Its Potential Regulation of Lipid Metabolism and Antioxidant Response in Largemouth Bass ( Micropterus salmoides). Front Physiol 2021; 12:726877. [PMID: 34646155 PMCID: PMC8504536 DOI: 10.3389/fphys.2021.726877] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/06/2021] [Indexed: 12/24/2022] Open
Abstract
Sirtuin 1 (SIRT1) of largemouth bass (Micropterus salmoides) was cloned and characterized in the present study and the influence of SIRT1 activation induced by resveratrol inclusion on the expression of genes related to lipid metabolism and antioxidation was also investigated. The SIRT1 of largemouth bass, with full-length cDNA sequence of 3395bp encoding 695 amino acids, was mainly expressed in gonad, heart and liver. The analysis of multiple sequence alignment revealed that, in accordance with other species, SIRT1 of largemouth bass contained highly conserved substrate-binding site and NAD+ binding site. The result of subcellular localization displayed that SIRT1 of largemouth bass was mainly localized in the nucleus. The inclusion of 1.0 and 2.5‰ dietary RSV, a natural SIRT1 activator, significantly elevated the SIRT1 protein expression. Meanwhile, the phosphorylation of AKT1 and FoxO1 followed similar pattern with that of SIRT1, indicating the activation of insulin pathway, which may result in the inhibition of lipogenesis and activation of lipolysis, and reduced hepatic triglycerides content. Additionally, the activation of SIRT1 induced by dietary RSV elevated the antioxidant capacity at both transcriptional level and enzymatic level, which was probably mediated by the transcription factor Nrf2. In above, SIRT1 was suggested to be involved in improving antioxidant capacity and alleviating hepatic lipid deposition in largemouth bass.
Collapse
Affiliation(s)
- Yuting Huang
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, China
| | - Shilin Wang
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, China
| | - Xiaoxue Meng
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, China
| | - Naisong Chen
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, China.,National Demonstration Center on Experiment Teaching of Fisheries Science, Shanghai Ocean University, Shanghai, China
| | - Songlin Li
- Research Centre of the Ministry of Agriculture and Rural Affairs on Environmental Ecology and Fish Nutrition, Shanghai Ocean University, Shanghai, China.,National Demonstration Center on Experiment Teaching of Fisheries Science, Shanghai Ocean University, Shanghai, China
| |
Collapse
|
16
|
Zhang L, Chen J, Yan L, He Q, Xie H, Chen M. Resveratrol Ameliorates Cardiac Remodeling in a Murine Model of Heart Failure With Preserved Ejection Fraction. Front Pharmacol 2021; 12:646240. [PMID: 34177571 PMCID: PMC8225267 DOI: 10.3389/fphar.2021.646240] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
Objective: Accumulating evidence suggested that resveratrol (RES) could protect against adverse cardiac remodeling induced by several cardiovascular diseases. However, the role of RES in the setting of heart failure with preserved ejection fraction (HFpEF) and the underlying mechanisms of its action remain understood. This study was to determine whether RES could ameliorate HFpEF-induced cardiac remodeling and its mechanisms. Methods:In vivo, C57BL/6 mice served as either the sham or the HFpEF model. The HFpEF mice model was induced by uninephrectomy surgery and d-aldosterone infusion. RES (10 mg/kg/day, ig) or saline was administered to the mice for four weeks. In vitro, transforming growth factor β1 (TGF-β1) was used to stimulate neonatal rat cardiac fibroblasts (CFs) and Ex-527 was used to inhibit sirtuin 1 (Sirt1) in CFs. Echocardiography, hemodynamics, western blotting, quantitative real-time PCR, histological analysis, immunofluorescence, and ELISA kits were used to evaluate cardiac remodeling induced by HFpEF. Sirt1 and Smad3 expressions were measured to explore the underlying mechanisms of RES. Results: HFpEF mice developed left ventricular hypertrophy, preserved ejection fraction, diastolic dysfunction, and pulmonary congestion. Moreover, HFpEF mice showed increased infiltration of neutrophils and macrophages into the heart, including increased interleukin (IL)-1β, IL-6, and TNF-α. We also observed elevated M1 macrophages and decreased M2 macrophages, which were exhibited by increased mRNA expression of M1 markers (iNOS, CD86, and CD80) and decreased mRNA expression of M2 markers (Arg1, CD163, and CD206) in HFpEF hearts. Moreover, HFpEF hearts showed increased levels of intracellular reactive oxygen species (ROS). Importantly, HFpEF mice depicted increased collagen-I and -III and TGF-β mRNA expressions and decreased protein expression of phosphorylated endothelial nitric-oxide synthase (p-eNOS). Results of western blot revealed that the activated TGF-β/Smad3 signaling pathway mediated HFpEF-induced cardiac remodeling. As expected, this HFpEF-induced cardiac remodeling was reversed when treated with RES. RES significantly decreased Smad3 acetylation and inhibited Smad3 transcriptional activity induced by HFpEF via activating Sirt1. Inhibited Sirt1 with Ex-527 increased Smad3 acetylation, enhanced Smad3 transcriptional activity, and offset the protective effect of RES on TGF-β–induced cardiac fibroblast–myofibroblast transformation in CFs. Conclusion: Our results suggested that RES exerts a protective action against HFpEF-induced adverse cardiac remodeling by decreasing Smad3 acetylation and transcriptional activity via activating Sirt1. RES is expected to be a novel therapy option for HFpEF patients.
Collapse
Affiliation(s)
- Liyun Zhang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Juan Chen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lianhua Yan
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin He
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Xie
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manhua Chen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Atale N, Yadav D, Rani V, Jin JO. Pathophysiology, Clinical Characteristics of Diabetic Cardiomyopathy: Therapeutic Potential of Natural Polyphenols. Front Nutr 2020; 7:564352. [PMID: 33344490 PMCID: PMC7744342 DOI: 10.3389/fnut.2020.564352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is an outcome of disturbances in metabolic activities through oxidative stress, local inflammation, and fibrosis, as well as a prime cause of fatality worldwide. Cardiovascular disorders in diabetic individuals have become a challenge in diagnosis and formulation of treatment prototype. It is necessary to have a better understanding of cellular pathophysiology that reveal the therapeutic targets and prevent the progression of cardiovascular diseases due to hyperglycemia. Critical changes in levels of collagen and integrin have been observed in the extracellular matrix of heart, which was responsible for cardiac remodeling in diabetic patients. This review explored the understanding of the mechanisms of how the phytochemicals provide cardioprotection under diabetes along with the caveats and provide future perspectives on these agents as prototypes for the development of drugs for managing DCM. Thus, here we summarized the effect of various plant extracts and natural polyphenols tested in preclinical and cell culture models of diabetic cardiomyopathy. Further, the potential use of selected polyphenols that improved the therapeutic efficacy against diabetic cardiomyopathy is also illustrated.
Collapse
Affiliation(s)
- Neha Atale
- Jaypee Institute of Information Technology, Noida, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Vibha Rani
- Jaypee Institute of Information Technology, Noida, India
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
18
|
Elrashidy RA, Hasan RA. Cilostazol preconditioning alleviates cyclophosphamide-induced cardiotoxicity in male rats: Mechanistic insights into SIRT1 signaling pathway. Life Sci 2020; 266:118822. [PMID: 33275987 DOI: 10.1016/j.lfs.2020.118822] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 01/08/2023]
Abstract
AIMS Cyclophosphamide (CYP) is a potent anticancer agent with well-known cardiotoxicity that limits its clinical applications. Cilostazol is a vosodilating drug, showing a cardioprotective effect in some cardiac disorders; however its effect in CYP-induced cardiotoxicity is still uncertain. We investigated the effect of cilostazol against CYP-induced cardiotoxicity and the contribution of SIRT1 signaling. MATERIALS AND METHODS 7 week-old male Wistar albino rats were treated with cilostazol (30 mg/kg/day, orally) in the absence or presence of SIRT1 inhibitor, EX-527 (5 mg/kg/day, IP) for 10 days and injected with CYP (200 mg/kg, IP) on the 7th day of the study. Age-matched rats were used as control group. On the 11th day, hearts were harvested for biochemical, immunoblotting and histological analyses. Markers of cardiac injury were assessed in plasma samples. KEY FINDINGS CYP injection contributed to cardiac injury manifested as significant increases in plasma activities of heart enzymes and cardiac troponin I levels. Cilostazol attenuated cardiac injury and minimized the histological lesions in hearts of CYP-treated rats. Cilostazol induced 3 fold up-regulation of SIRT1 and promoted the antioxidant defense response through FoxO1-related mechanism in hearts of CYP-treated rats. Cilostazol suppressed the CYP-induced up-regulation of PARP1 and p53, and blocked the NF-kB p65-mediated inflammatory response in hearts of CYP-treated rats. All the beneficial effects of cilostazol were almost abolished by EX-527. SIGNIFICANCE These data provided insights into the mechanism underlying the cardioprotective effect of cilostazol in CYP-treated rats through upregulation of SIRT1 signaling, suggesting that cilostazol might be a candidate modality for CYP-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rania A Elrashidy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| | - Rehab A Hasan
- Histology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
19
|
Effects of resveratrol on mitochondrial biogenesis and physiological diseases. ADVANCES IN TRADITIONAL MEDICINE 2020. [DOI: 10.1007/s13596-020-00492-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
20
|
Wang Z, Zhao YT, Zhao TC. Histone deacetylases in modulating cardiac disease and their clinical translational and therapeutic implications. Exp Biol Med (Maywood) 2020; 246:213-225. [PMID: 32727215 DOI: 10.1177/1535370220944128] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular diseases are the leading cause of mortality and morbidity worldwide. Histone deacetylases (HDACs) play an important role in the epigenetic regulation of genetic transcription in response to stress or pathological conditions. HDACs interact with a complex co-regulatory network of transcriptional regulators, deacetylate histones or non-histone proteins, and modulate gene expression in the heart. The selective HDAC inhibitors have been considered to be a critical target for the treatment of cardiac disease, especially for ameliorating cardiac dysfunction. In this review, we discuss our current knowledge of the cellular and molecular basis of HDACs in mediating cardiac development and hypertrophy and related pharmacologic interventions in heart disease.
Collapse
Affiliation(s)
- Zhengke Wang
- Department of Surgery, Boston University Medical School, Roger Williams Medical Center, Providence, RI 02908, USA
| | - Yu Tina Zhao
- University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Ting C Zhao
- Departments of Surgery and Plastic Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
21
|
SIRT1 Activation by Polydatin Alleviates Oxidative Damage and Elevates Mitochondrial Biogenesis in Experimental Diabetic Neuropathy. Cell Mol Neurobiol 2020; 41:1563-1577. [PMID: 32683581 DOI: 10.1007/s10571-020-00923-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 07/13/2020] [Indexed: 10/23/2022]
Abstract
Mitochondrial dysfunction has been implicated as a one of the major factors linked to the development of painful diabetic neuropathy (DN). Several studies have demonstrated that sirtuin (SIRT1) activation recuperates nerve function by activating mitochondrial biogenesis. Polydatin, a resveratrol glycoside, has been explored to improve mitochondrial function via SIRT1 activation. However, the neuroprotective effects of polydatin in DN remain elusive. In this study, polydatin (25 and 50 mg/kg, oral) was administered for last 2 weeks of 8-week study to diabetic Sprague-Dawley rats weighing 250-300 g (post 6-weeks of streptozotocin 55 mg/kg, intraperitoneal). Treatment with polydatin significantly attenuated mechanical and thermal hyperalgesia in diabetic rats. Treated diabetic rats also showed improvement in motor/sensory nerve conduction velocities and nerve blood flow. For in vitro studies, Neuro2a cells were exposed to high-glucose (30 mM) condition to simulate short-term hyperglycemia. Polydatin was evaluated for its role in SIRT1 and Nrf2 activation at a dose of 5, 10, and 20 µM concentrations. Polydatin exposure normalized the mitochondrial superoxides, membrane potentials and improved neurite outgrowth in high-glucose-exposed Neuro2a cells. Increased SIRT1 activation by polydatin resulted in peroxisome proliferator activated receptor-gamma coactivator-1α (PGC-1α) directed mitochondrial biogenesis. SIRT1 activation also facilitated Nrf2-directed antioxidant signaling. Study results inferred that decline in mitochondrial biogenesis and oxidative function in diabetic rats and high-glucose-exposed Neuro2a cells, could be counteracted by polydatin administration, postulated via enhancing SIRT1 and Nrf2 axis.
Collapse
|
22
|
The Mitochondria: A Target of Polyphenols in the Treatment of Diabetic Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21144962. [PMID: 32674299 PMCID: PMC7404043 DOI: 10.3390/ijms21144962] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a constellation of symptoms consisting of ventricular dysfunction and cardiomyocyte disarray in the presence of diabetes. The exact cause of this type of cardiomyopathy is still unknown; however, several processes involving the mitochondria, such as lipid and glucose metabolism, reactive oxygen species (ROS) production, apoptosis, autophagy and mitochondrial biogenesis have been implicated. In addition, polyphenols have been shown to improve the progression of diabetes. In this review, we discuss some of the mechanisms by which polyphenols, particularly resveratrol, play a role in slowing the progression of DCM. The most important intermediates by which polyphenols exert their protective effect include Bcl-2, UCP2, SIRT-1, AMPK and JNK1. Bcl-2 acts to attenuate apoptosis, UCP2 decreases oxidative stress, SIRT-1 increases mitochondrial biogenesis and decreases oxidative stress, AMPK increases autophagy, and JNK1 decreases apoptosis and increases autophagy. Our dissection of these molecular players aims to provide potential therapeutic targets for the treatment of DCM.
Collapse
|
23
|
Yessenkyzy A, Saliev T, Zhanaliyeva M, Masoud AR, Umbayev B, Sergazy S, Krivykh E, Gulyayev A, Nurgozhin T. Polyphenols as Caloric-Restriction Mimetics and Autophagy Inducers in Aging Research. Nutrients 2020; 12:E1344. [PMID: 32397145 PMCID: PMC7285205 DOI: 10.3390/nu12051344] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/16/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
It has been thought that caloric restriction favors longevity and healthy aging where autophagy plays a vital role. However, autophagy decreases during aging and that can lead to the development of aging-associated diseases such as cancer, diabetes, neurodegeneration, etc. It was shown that autophagy can be induced by mechanical or chemical stress. In this regard, various pharmacological compounds were proposed, including natural polyphenols. Apart from the ability to induce autophagy, polyphenols, such as resveratrol, are capable of modulating the expression of pro- and anti-apoptotic factors, neutralizing free radical species, affecting mitochondrial functions, chelating redox-active transition metal ions, and preventing protein aggregation. Moreover, polyphenols have advantages compared to chemical inducers of autophagy due to their intrinsic natural bio-compatibility and safety. In this context, polyphenols can be considered as a potential therapeutic tool for healthy aging either as a part of a diet or as separate compounds (supplements). This review discusses the epigenetic aspect and the underlying molecular mechanism of polyphenols as an anti-aging remedy. In addition, the recent advances of studies on NAD-dependent deacetylase sirtuin-1 (SIRT1) regulation of autophagy, the role of senescence-associated secretory phenotype (SASP) in cells senescence and their regulation by polyphenols have been highlighted as well. Apart from that, the review also revised the latest information on how polyphenols can help to improve mitochondrial function and modulate apoptosis (programmed cell death).
Collapse
Affiliation(s)
- Assylzhan Yessenkyzy
- Research Institute of Fundamental and Applied Medicine named after B. Atchabarov, S.D. Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan; (A.Y.); (T.N.)
| | - Timur Saliev
- Research Institute of Fundamental and Applied Medicine named after B. Atchabarov, S.D. Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan; (A.Y.); (T.N.)
| | - Marina Zhanaliyeva
- Department of Human Anatomy, NSC “Medical University of Astana”, Nur-Sultan 010000, Kazakhstan;
| | - Abdul-Razak Masoud
- Department of Biological Sciences, Louisiana Tech University, Ruston, LA 71270, USA;
| | - Bauyrzhan Umbayev
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (B.U.); (S.S.); (A.G.)
| | - Shynggys Sergazy
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (B.U.); (S.S.); (A.G.)
| | - Elena Krivykh
- Khanty-Mansiysk State Medical Academy, Tyumen Region, Khanty-Mansiysk Autonomous Okrug—Ugra, Khanty-Mansiysk 125438, Russia;
| | - Alexander Gulyayev
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (B.U.); (S.S.); (A.G.)
| | - Talgat Nurgozhin
- Research Institute of Fundamental and Applied Medicine named after B. Atchabarov, S.D. Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan; (A.Y.); (T.N.)
| |
Collapse
|
24
|
The roles of resveratrol on cardiac mitochondrial function in cardiac diseases. Eur J Nutr 2020; 60:29-44. [PMID: 32372266 DOI: 10.1007/s00394-020-02256-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/22/2020] [Indexed: 12/31/2022]
Abstract
Left ventricular (LV) dysfunction is commonly associated with a variety of health conditions including acute myocardial infarction and obesity/diabetes. In addition, administration of several pharmacological agents such as anticancer, antiviral, and immunosuppressive drugs has been shown to be related with LV dysfunction. The molecular mechanism responsible for LV dysfunction has been extensively studied, and it has been proposed that the overproduction of reactive oxygen species (ROS) plays a crucial role in the regulation of this function. Mitochondria require the balance between ROS production and antioxidants to maintain their appropriate function and to prevent excessive ROS production. Thus, the excessive production of ROS and the reduced scavenging process under any pathological conditions could disrupt mitochondrial function, leading to energy depletion with subsequent cell death. Therefore, maintenance of the balance between oxidative stress and antioxidants is essential. Resveratrol, a stilbene, has been investigated extensively, and potentially used to treat or prevent various cardiovascular diseases. Resveratrol directly upregulates antioxidative capacity by increasing antioxidant genes such as heme oxygenase-1, superoxide dismutase, catalase, and glutathione. In this review, accumulated data from in vitro, ex vivo, and in vivo studies regarding the effects of resveratrol on cardiac mitochondrial function in cardiac pathologies are comprehensively summarized and discussed. Since there is no conclusive available clinical study regarding the effects of resveratrol on cardiac mitochondrial function, this review also aims to encourage more clinical investigations to confirm findings from basic research. This comprehensive review will provide insight regarding the potential mechanistic roles of resveratrol in preventing and/or treating patients with cardiovascular diseases to improve LV function and their health status.
Collapse
|
25
|
Liang H, Li W, Yang H, Cao Y, Ge L, Shi R, Fan Z, Dong R, Zhang C. FAM96B inhibits the senescence of dental pulp stem cells. Cell Biol Int 2020; 44:1193-1203. [DOI: 10.1002/cbin.11319] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/02/2020] [Indexed: 01/12/2023]
Affiliation(s)
- Hanbing Liang
- Department of EndodonticsCapital Medical University School of Stomatology Beijing 100050 China
| | - Wenzhi Li
- Department of EndodonticsCapital Medical University School of Stomatology Beijing 100050 China
| | - Haoqing Yang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Yangyang Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Lihua Ge
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Ruitang Shi
- Department of EndodonticsCapital Medical University School of Stomatology Beijing 100050 China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Rui Dong
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function ReconstructionCapital Medical University School of Stomatology Beijing 100050 China
| | - Chen Zhang
- Department of EndodonticsCapital Medical University School of Stomatology Beijing 100050 China
| |
Collapse
|
26
|
Yamagata K. Polyphenols Regulate Endothelial Functions and Reduce the Risk of Cardiovascular Disease. Curr Pharm Des 2020; 25:2443-2458. [PMID: 31333108 DOI: 10.2174/1381612825666190722100504] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 07/20/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Previous studies have shown that intake of polyphenols through the consumption of vegetables and fruits reduces the risk of Cardiovascular Disease (CVD) by potentially influencing endothelial cell function. OBJECTIVE In this review, the effects and molecular mechanisms of plant polyphenols, particularly resveratrol, epigallocatechin gallate (EGCG), and quercetin, on endothelial functions, and their putative protective effects against CVD are described. METHODS Epidemiologic studies examined the effect of the CVD risk of vegetables and the fruit. Furthermore, studies within vitro models investigated the underlying molecular mechanisms of the action of the flavonoid class of polyphenols. These findings help elucidate the effect of polyphenols on endothelial function and CVD risk reduction. RESULTS Epidemiologic and in vitro studies have demonstrated that the consumption of vegetables and fruits decreases the incidence of CVDs. Furthermore, it has also been indicated that dietary polyphenols are inversely related to the risk of CVD. Resveratrol, EGCG, and quercetin prevent oxidative stress by regulating the expression of oxidase and the antioxidant enzyme genes, contributing to the prevention of stroke, hypertension, heart failure, and ischemic heart disease. CONCLUSION High intake of dietary polyphenols may help prevent CVD. Polyphenols inhibit endothelial dysfunction and induce vascular endothelium-dependent vascular relaxation viz. redox regulation and nitric oxide production. The polyphenol-induced healthy endothelial cell function may be related to CVD prevention.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Laboratory of Molecular Health Science of Food, Department of Food Science & Technology, Nihon University (NUBS), 1866 Kameino, Fujisawa, Kanagawa, 252-8510, Japan
| |
Collapse
|
27
|
Cheng CK, Luo J, Lau CW, Chen Z, Tian XY, Huang Y. Pharmacological basis and new insights of resveratrol action in the cardiovascular system. Br J Pharmacol 2020; 177:1258-1277. [PMID: 31347157 PMCID: PMC7056472 DOI: 10.1111/bph.14801] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
Resveratrol (trans-3,4',5-trihydroxystilbene) belongs to the family of natural phytoalexins. Resveratrol first came to our attention in 1992, following reports of the cardioprotective effects of red wine. Thereafter, resveratrol was shown to exert antioxidant, anti-inflammatory, anti-proliferative, and angio-regulatory effects against atherosclerosis, ischaemia, and cardiomyopathy. This article critically reviews the current findings on the molecular basis of resveratrol-mediated cardiovascular benefits, summarizing the broad effects of resveratrol on longevity regulation, energy metabolism, stress resistance, exercise mimetics, circadian clock, and microbiota composition. In addition, this article also provides an update, both preclinically and clinically, on resveratrol-induced cardiovascular protection and discusses the adverse and inconsistent effects of resveratrol reported in both preclinical and clinical studies. Although resveratrol has been claimed as a master anti-aging agent against several age-associated diseases, further detailed mechanistic investigation is still required to thoroughly unravel the therapeutic value of resveratrol against cardiovascular diseases at different stages of disease development. LINKED ARTICLES: This article is part of a themed section on The Pharmacology of Nutraceuticals. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.6/issuetoc.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Jiang‐Yun Luo
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Chi Wai Lau
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Zhen‐Yu Chen
- Food and Nutritional Sciences Programme, School of Life SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Xiao Yu Tian
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| | - Yu Huang
- Heart and Vascular Institute, Shenzhen Research Institute and Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongSARChina
| |
Collapse
|
28
|
Resveratrol protects H9c2 cells against hypoxia-induced apoptosis through miR-30d-5p/SIRT1/NF-κB axis. J Biosci 2020. [DOI: 10.1007/s12038-020-9997-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
29
|
Guo G, Gong L, Sun L, Xu H. Quercetin supports cell viability and inhibits apoptosis in cardiocytes by down-regulating miR-199a. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2909-2916. [PMID: 31307244 DOI: 10.1080/21691401.2019.1640711] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Hypoxia-caused cardiocytes insults are closely correlated with ectopic expression of genes, which might be modulated by microRNAs (miRs). Quercetin exhibits a profound protective function against hypoxic damages in cardiomyocytes. Here, we aimed to investigate a possible underpinning. H9c2 cells were pre-administrated using quercetin before hypoxia treatment. The damages were assessed using viability, apoptosis and alteration of proteins associated with apoptosis and adenosine monophosphate-activated protein (AMPK) pathway. Transfection was conducted to enforce overexpression of miR-199a or silence of sirtuin 1 (sirt1) which were confirmed by qRT-PCR. Sirt1 protein was quantified by immunoblotting. A luciferase reporter was exploited to confirm the target relationship between miR-199a and sirt1 3'-untranslated region (3'-UTR). We found quercetin mitigated hypoxia-caused viability reduction and apoptosis with restoring apoptosis-associated protein and rescuing phosphorylation of AMPK. Quercetin flattened hypoxia-evoked overexpression of miR-199a. miR-199a abrogated the protective effects of quercetin against hypoxia-elicited damages. Quercetin elevated sirt1 which was repressed by hypoxia, while this effect was slight in miR-199a-overexpressed cells. miR-199a negatively mediated sirt1 expression through directly binding its 3'-UTR. Further, quercetin facilitated the phosphorylation of AMPK by up-regulating sirt1. Collectively, quercetin participated in repressing miR-199a which negatively modulated sirt1. Mechanically, through activating AMPK, quercetin protected cardiomyocytes cells against hypoxia-caused insults. Highlights Quercetin ameliorates hypoxia-evoked apoptosis and blockage of AMPK phosphorylation; The elevated miR-199a level is eased by quercetin, which might be a protective mechanism; Quercetin restores sirt1 level by repressing miR-199a expression; By mediating miR-199a and sirt1, AMPK phosphorylation is fortified by quercetin.
Collapse
Affiliation(s)
- Gongliang Guo
- a Department of Cardiology, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Licheng Gong
- a Department of Cardiology, China-Japan Union Hospital of Jilin University , Changchun , China
| | - Liqun Sun
- b Outpatient Department of Pediatrics, The First Hospital of Jilin University , Changchun , China
| | - Haiming Xu
- a Department of Cardiology, China-Japan Union Hospital of Jilin University , Changchun , China
| |
Collapse
|
30
|
Ren Z, He H, Zuo Z, Xu Z, Wei Z, Deng J. The role of different SIRT1-mediated signaling pathways in toxic injury. Cell Mol Biol Lett 2019; 24:36. [PMID: 31164908 PMCID: PMC6543624 DOI: 10.1186/s11658-019-0158-9] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/09/2019] [Indexed: 01/07/2023] Open
Abstract
Common environmental pollutants and drugs encountered in everyday life can cause toxic damage to the body through oxidative stress, inflammatory stimulation, induction of apoptosis, and inhibition of energy metabolism. Silent information regulator 1 (SIRT1), a nicotinamide adenine dinucleotide-dependent deacetylase, is a member of the evolutionarily highly conserved Sir2 (silent information regulator 2) superprotein family, which is located in the nucleus and cytoplasm. It can deacetylate protein substrates in various signal transduction pathways to regulate gene expression, cell apoptosis and senescence, participate in the process of neuroprotection, energy metabolism, inflammation and the oxidative stress response in living organisms, and plays an important role in toxic damage caused by toxicants and in the process of SIRT1 activator/inhibitor antagonized toxic damage. This review summarizes the role that SIRT1 plays in toxic damage caused by toxicants via its interactions with protein substrates in certain signaling pathways.
Collapse
Affiliation(s)
- Zhihua Ren
- 1Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 Sichuan Province China
| | - Hongyi He
- 1Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 Sichuan Province China
| | - Zhicai Zuo
- 1Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 Sichuan Province China
| | - Zhiwen Xu
- 1Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 Sichuan Province China
| | - Zhanyong Wei
- 2The College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002 Henan Province China
| | - Junliang Deng
- 1Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 Sichuan Province China
| |
Collapse
|
31
|
Xu RY, Xu XW, Deng YZ, Ma ZX, Li XR, Zhao L, Qiu LJ, Liu HY, Chen HP. Resveratrol attenuates myocardial hypoxia/reoxygenation-induced cell apoptosis through DJ-1-mediated SIRT1-p53 pathway. Biochem Biophys Res Commun 2019; 514:401-406. [PMID: 31053297 DOI: 10.1016/j.bbrc.2019.04.165] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 04/24/2019] [Indexed: 02/04/2023]
Abstract
Resveratrol, a multi-functional phytoalexin, has been well indicated to exert cardioprotective effects by weakening ischemia/reperfusion (I/R) injury, and cell apoptosis is a vital way in I/R injury. SIRT1-p53 pathway has strong significance in regulating cell apoptosis. DJ-1 can directly bind to SIRT1 and stimulate the activity of SIRT1-p53. Therefore, the current study was determined whether Resveratrol attenuates hypoxia/reoxygenation (H/R)-induced cell apoptosis, and whether DJ-1-mediated SIRT1 activation involves in the cardioprotective effects of Resveratrol. The results showed that remarkable decrease in the number of apoptotic cells along with reduction of lactate dehydrogenase release and restoration of cell viability emerged when Resveratrol was applied in the H9c2 cells exposed to H/R. Moreover, Resveratrol increased DJ-1 expression and promoted the interaction of DJ-1 with SIRT1, which further contributed to subsequent restoration of SIRT1 activity and decrease of acetylation level of p53. However, above cardioprotective effects of Resveratrol were abrogated by DJ-1 siRNA and SIRT1 specific inhibitor Sirtinol. In conclusion, the current study demonstrated that Resveratrol suppressed H/R-induced cell apoptosis, which may be conducted by up-regulating DJ-1, and later activating SIRT1 activity and subsequently inhibiting p53 acetylation level in the H9c2 cells.
Collapse
Affiliation(s)
- Rui-Yuan Xu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - Xing-Wang Xu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - Yi-Zhang Deng
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - Zhao-Xia Ma
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - Xiao-Ran Li
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - Le Zhao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - Le-Jia Qiu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - Hao-Yue Liu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China
| | - He-Ping Chen
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, PR China.
| |
Collapse
|
32
|
Mitochondrial complex I deficiency and cardiovascular diseases: current evidence and future directions. J Mol Med (Berl) 2019; 97:579-591. [PMID: 30863992 DOI: 10.1007/s00109-019-01771-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/06/2019] [Accepted: 03/05/2019] [Indexed: 02/06/2023]
Abstract
Compelling evidence demonstrates the emerging role of mitochondrial complex I deficiency in the onset and development of cardiovascular diseases (CVDs). In particular, defects in single subunits of mitochondrial complex I have been associated with cardiac hypertrophy, ischemia/reperfusion injury, as well as diabetic complications and stroke in pre-clinical studies. Moreover, data obtained in humans revealed that genes coding for complex I proteins were associated with different CVDs. In this review, we discuss recent experimental studies that underline the contributory role of mitochondrial complex I deficiency in the etiopathogenesis of several CVDs, with a particular focus on those involving loss of function models of mitochondrial complex I. We also discuss human studies and potential therapeutic strategies able to rescue mitochondrial function in CVDs.
Collapse
|
33
|
Bakuchiol: A newly discovered warrior against organ damage. Pharmacol Res 2019; 141:208-213. [DOI: 10.1016/j.phrs.2019.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/24/2018] [Accepted: 01/01/2019] [Indexed: 12/17/2022]
|
34
|
The Effects of Resveratrol, Metformin, Cold and Strength Training on the Level of Perilipin 5 in the Heart, Skeletal Muscle and Brown Adipose Tissues in Mouse. Cell Biochem Biophys 2018; 76:471-476. [PMID: 30343407 DOI: 10.1007/s12013-018-0860-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/13/2018] [Indexed: 10/28/2022]
Abstract
The high accumulation of lipid droplets in the cell is related to metabolic disorders, such as obesity. Perilipin 5 (Plin5), plays an important role in triglyceride hydrolysis in the lipid droplets. In this study, this protein has been evaluated in different tissues and conditions in mice. Fifty male mice were divided into 5 groups and treated for 45 days with Resveratrol, Metformin, strength training, and 4 °C cold. Brown adipose tissue (BAT), gastrocnemius skeletal muscle and heart were isolated for RNA extraction. The Plin5 gene expression was evaluated, using Real-Time PCR, and the plin5 was analyzed at the protein level, using western blot. In BAT, Resveratrol significantly reduced the plin5 protein level and gene expression (p < 0.05). In heart tissue, Resveratrol and strength training, decreased (p < 0.05) the plin5 expression, but Metformin increased the gene expression (p < 0.05). In skeletal muscle, resveratrol, strength training, cold and Metformin significantly increased the plin5 expression at the gene and protein level (p < 0.05). In BAT, Resveratrol has a greater effect in decreasing lipid deposits, compared with the strength training and cold; thus, it can play a better role in preventing lipid accumulation. In heart tissue, Resveratrol probably decreases insulin resistance, due to the increased expression of plin5 in skeletal muscle.
Collapse
|
35
|
Zein-polysaccharide nanoparticles as matrices for antioxidant compounds: A strategy for prevention of chronic degenerative diseases. Food Res Int 2018; 111:451-471. [DOI: 10.1016/j.foodres.2018.05.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 05/14/2018] [Accepted: 05/18/2018] [Indexed: 02/07/2023]
|
36
|
Li N, Zhou H, Ma ZG, Zhu JX, Liu C, Song P, Kong CY, Wu HM, Deng W, Tang QZ. Geniposide Alleviates Isoproterenol-Induced Cardiac Fibrosis Partially via SIRT1 Activation in vivo and in vitro. Front Pharmacol 2018; 9:854. [PMID: 30123131 PMCID: PMC6086014 DOI: 10.3389/fphar.2018.00854] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/16/2018] [Indexed: 12/18/2022] Open
Abstract
Objective: Geniposide (GE) is a major component in the fruit of Gardenia jasminoides Ellis. Oxidative stress, endoplasmic reticulum (ER) stress, and canonical Smad3 pathway are implicated in the pathogenesis of cardiac fibrosis. We aim to investigate the protective roles of GE in isoproterenol (ISO)-induced cardiac fibrosis. Methods: ISO was used to induce cardiac fibrosis in male C57BL/6 mice. GE and the EX-527 were given for 2 weeks to detect the effects of GE on cardiac fibrosis. Levels of oxidative stress, ER stress, and Smad3 were evaluated by real time-PCR, Western blots, immunohistochemistry staining, immunofluorescence staining, and assay kits. Results: GE treatment alleviated cardiac dysfunction, fibrosis, and hypertrophy in mice response to ISO. Additionally, GE also suppressed the transformation of cardiac fibroblasts to myofibroblasts stimulated by transforming growth factor-β (TGF-β) in vitro. Mechanistically, GE inhibited the oxidative stress, ER stress, as well as Smad3 pathway activated by ISO or TGF-β. A selective antagonist of sirtuin 1 deacetylase (SIRT1), EX-527, partially counteracted the anti-fibrotic effect and weakened the inhibitory effect on the transformation of cardiac fibroblasts to myofibroblasts after the treatment of GE. Acetylated Smad3 (ac-Smad3), oxidative stress, as well as ER stress pathway were significantly enhanced after SIRT1 was blocked while phosphorylated Smad3 (P-Smad3) was not affected. Conclusion: GE could combat cardiac fibrosis in vivo and in vitro by inhibiting oxidative stress, ER stress, and ac-Smad3 in a SIRT1-dependent manner and suppressing P-Samd3 pathway independent of SIRT1 activation. GE is expected to be a promising agent against cardiac fibrosis.
Collapse
Affiliation(s)
- Ning Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jin-Xiu Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Peng Song
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chun-Yan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
37
|
Shi W, Deng H, Zhang J, Zhang Y, Zhang X, Cui G. Mitochondria-Targeting Small Molecules Effectively Prevent Cardiotoxicity Induced by Doxorubicin. Molecules 2018; 23:E1486. [PMID: 29921817 PMCID: PMC6099719 DOI: 10.3390/molecules23061486] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
Doxorubicin (Dox) is a chemotherapeutic agent widely used for the treatment of numerous cancers. However, the clinical use of Dox is limited by its unwanted cardiotoxicity. Mitochondrial dysfunction has been associated with Dox-induced cardiotoxicity. To mitigate Dox-related cardiotoxicity, considerable successful examples of a variety of small molecules that target mitochondria to modulate Dox-induced cardiotoxicity have appeared in recent years. Here, we review the related literatures and discuss the evidence showing that mitochondria-targeting small molecules are promising cardioprotective agents against Dox-induced cardiac events.
Collapse
Affiliation(s)
- Wei Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| | - Hongkuan Deng
- School of Life Sciences, Shandong University of Technology, Zibo 255000, China.
| | - Jianyong Zhang
- Pharmacy School, Zunyi Medical University, Zunyi 563003, China.
| | - Ying Zhang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| | - Xiufang Zhang
- School of Life Sciences, Shandong University of Technology, Zibo 255000, China.
| | - Guozhen Cui
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| |
Collapse
|
38
|
Siasos G, Tsigkou V, Kosmopoulos M, Theodosiadis D, Simantiris S, Tagkou NM, Tsimpiktsioglou A, Stampouloglou PK, Oikonomou E, Mourouzis K, Philippou A, Vavuranakis M, Stefanadis C, Tousoulis D, Papavassiliou AG. Mitochondria and cardiovascular diseases-from pathophysiology to treatment. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:256. [PMID: 30069458 DOI: 10.21037/atm.2018.06.21] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mitochondria are the source of cellular energy production and are present in different types of cells. However, their function is especially important for the heart due to the high demands in energy which is achieved through oxidative phosphorylation. Mitochondria form large networks which regulate metabolism and the optimal function is achieved through the balance between mitochondrial fusion and mitochondrial fission. Moreover, mitochondrial function is upon quality control via the process of mitophagy which removes the damaged organelles. Mitochondrial dysfunction is associated with the development of numerous cardiac diseases such as atherosclerosis, ischemia-reperfusion (I/R) injury, hypertension, diabetes, cardiac hypertrophy and heart failure (HF), due to the uncontrolled production of reactive oxygen species (ROS). Therefore, early control of mitochondrial dysfunction is a crucial step in the therapy of cardiac diseases. A number of anti-oxidant molecules and medications have been used but the results are inconsistent among the studies. Eventually, the aim of future research is to design molecules which selectively target mitochondrial dysfunction and restore the capacity of cellular anti-oxidant enzymes.
Collapse
Affiliation(s)
- Gerasimos Siasos
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.,Division of Cardiovascular, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vasiliki Tsigkou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Marinos Kosmopoulos
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimosthenis Theodosiadis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Spyridon Simantiris
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Nikoletta Maria Tagkou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athina Tsimpiktsioglou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiota K Stampouloglou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Konstantinos Mourouzis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Anastasios Philippou
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Dimitris Tousoulis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
39
|
Aras-López R, Almeida L, Andreu-Fernández V, Tovar J, Martínez L. Anti-oxidants correct disturbance of redox enzymes in the hearts of rat fetuses with congenital diaphragmatic hernia. Pediatr Surg Int 2018; 34:307-313. [PMID: 29079903 DOI: 10.1007/s00383-017-4201-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/12/2017] [Indexed: 01/20/2023]
Abstract
AIM To evaluate if the redox system is unbalanced in the hearts of nitrofen-induced congenital diaphragmatic hernia (CDH) animals and to study the possible preventive effects of two anti-oxidant treatments, apocynin and epigallocatechin-3-gallate (EGCG). METHODS Adult rats were divided into four groups. Group 1: rats received only vehicle on day E9.5. Group 2: rats received 100 mg nitrofen on day E9.5. Group 3: 1 month before mating rats received apocynin 1.5 mM and, when pregnant, 100 mg nitrofen on day E9.5. Group 4: same than group 3 but with EGCG 30 mg/kg. All fetuses were recovered at term and the hearts were processed. Nox activity and mRNA levels of Nox1, Nox2, Nox4, SOD1, SOD2, SOD3, catalase, and GPX1 were analyzed. Nox, SOD, and Catalase activity and H2O2 production were also evaluated. RESULTS Nox activity, H2O2 production and Nox1, Nox2, and Nox4 mRNA levels were increased in the hearts of fetuses with CDH. There were no changes in SOD1 levels, whereas those of SOD2, SOD3, catalase, and GPX1 mRNA were decreased. Apocynin and EGCG treatments attenuated the increment of Nox and SOD activities and H2O2 production was only decreased by apocynin. CONCLUSION These findings suggest a possible preventive effect on the abnormal redox metabolism of anti-oxidant treatments in the hearts from rat fetuses with CDH. If the same occurs in humans, it could represent a potential tool in future prenatal treatment.
Collapse
Affiliation(s)
- Rosa Aras-López
- Congenital Malformations Lab, Institute of Medicine and Molecular Genetic (INGEMM), Institute for Health Research of La Paz Universitary Hospital (IdiPAZ), Madrid, Spain.
| | - L Almeida
- BCNatal, Barcelona Center for Maternal-Fetal Medicine and Neonatology, Hospital Clinic and Hospital San Joan de Deu, IdiBaps, University of Barcelona, Barcelona, Spain
| | - V Andreu-Fernández
- Fundació Clínic per la Recerca Biomèdica, BCNatal, GRIE, ICGON, Barcelona Center for Maternal-Fetal Medicine and Neonatology, Servicio de Neonatologia, Hospital Clinic-Maternitat, Barcelona, Spain
| | - J Tovar
- Department of Pediatric Surgery, Hospital Universitario La Paz, Madrid, Spain
| | - L Martínez
- Congenital Malformations Lab, Institute of Medicine and Molecular Genetic (INGEMM), Institute for Health Research of La Paz Universitary Hospital (IdiPAZ), Madrid, Spain.,Department of Pediatric Surgery, Hospital Universitario La Paz, Madrid, Spain
| |
Collapse
|
40
|
Zhang Y, Li XR, Zhao L, Duan GL, Xiao L, Chen HP. DJ-1 preserving mitochondrial complex I activity plays a critical role in resveratrol–mediated cardioprotection against hypoxia/reoxygenation–induced oxidative stress. Biomed Pharmacother 2018; 98:545-552. [DOI: 10.1016/j.biopha.2017.12.094] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 01/03/2023] Open
|
41
|
Suppression of Oxidative Stress and Apoptosis in Electrically Stimulated Neonatal Rat Cardiomyocytes by Resveratrol and Underlying Mechanisms. J Cardiovasc Pharmacol 2017; 70:396-404. [DOI: 10.1097/fjc.0000000000000534] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
42
|
Fernandes GFS, Silva GDB, Pavan AR, Chiba DE, Chin CM, Dos Santos JL. Epigenetic Regulatory Mechanisms Induced by Resveratrol. Nutrients 2017; 9:nu9111201. [PMID: 29104258 PMCID: PMC5707673 DOI: 10.3390/nu9111201] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022] Open
Abstract
Resveratrol (RVT) is one of the main natural compounds studied worldwide due to its potential therapeutic use in the treatment of many diseases, including cancer, diabetes, cardiovascular diseases, neurodegenerative diseases and metabolic disorders. Nevertheless, the mechanism of action of RVT in all of these conditions is not completely understood, as it can modify not only biochemical pathways but also epigenetic mechanisms. In this paper, we analyze the biological activities exhibited by RVT with a focus on the epigenetic mechanisms, especially those related to DNA methyltransferase (DNMT), histone deacetylase (HDAC) and lysine-specific demethylase-1 (LSD1).
Collapse
Affiliation(s)
- Guilherme Felipe Santos Fernandes
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
- Institute of Chemistry, São Paulo State University (UNESP), 14800060 Araraquara, Brazil.
| | | | - Aline Renata Pavan
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
| | - Diego Eidy Chiba
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
| | - Chung Man Chin
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
| | - Jean Leandro Dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
| |
Collapse
|
43
|
Yu M, Wang C, Zeng G, Zeng G, Zhou L, Chen T, Tan X, Wang Y. Toll‑like receptor 4 is expressed and functional in late endothelial progenitor cells. Mol Med Rep 2017; 16:5549-5554. [PMID: 28849058 DOI: 10.3892/mmr.2017.7291] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 07/07/2017] [Indexed: 02/05/2023] Open
Abstract
It has been previously demonstrated that lipopolysaccharides (LPS) inhibit the viability, migration, adhesion and in vitro angiogenesis of late endothelial progenitor cells (EPCs). However, the mechanisms underlying this LPS‑induced impairment of late EPC functional activity are unknown. The aim of the present study was to investigate whether Toll‑like receptor 4 (TLR4) is expressed and functional on late EPCs, using late EPCs of 3‑5 passages. Cells were deprived of serum for 24 h prior to experiments and incubated with 10 µg/ml LPS for 24 h with or without pretreatment with 2 µg/ml TLR4 signaling inhibitor CLI‑095 for 30 min. The viability, migration, adhesion and in vitro angiogenesis, as well as the expression of silent information regulator 1 (SIRT1), in late EPCs were evaluated. Treatment with 10 µg/ml LPS decreased the viability, migration and adhesion abilities, and in vitro angiogenesis of late EPCs. Pretreatment with the TLR4 signaling inhibitor reversed this LPS‑induced dysfunction of late EPCs. LPS downregulated the expression of SIRT1 protein, however, blocking TLR4 attenuated the effect of LPS on SIRT1 expression. Therefore, the results of the present study indicate that LPS impaired the functional activity of late EPCs via TLR4, which may be associated with decreased SIRT1 expression.
Collapse
Affiliation(s)
- Min Yu
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Cantian Wang
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Guoning Zeng
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Guoning Zeng
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Lihong Zhou
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Tingting Chen
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xuerui Tan
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yanping Wang
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
44
|
Mattera R, Benvenuto M, Giganti MG, Tresoldi I, Pluchinotta FR, Bergante S, Tettamanti G, Masuelli L, Manzari V, Modesti A, Bei R. Effects of Polyphenols on Oxidative Stress-Mediated Injury in Cardiomyocytes. Nutrients 2017; 9:nu9050523. [PMID: 28531112 PMCID: PMC5452253 DOI: 10.3390/nu9050523] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/09/2017] [Accepted: 05/16/2017] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases are the main cause of mortality and morbidity in the world. Hypertension, ischemia/reperfusion, diabetes and anti-cancer drugs contribute to heart failure through oxidative and nitrosative stresses which cause cardiomyocytes nuclear and mitochondrial DNA damage, denaturation of intracellular proteins, lipid peroxidation and inflammation. Oxidative or nitrosative stress-mediated injury lead to cardiomyocytes apoptosis or necrosis. The reactive oxygen (ROS) and nitrogen species (RNS) concentration is dependent on their production and on the expression and activity of anti-oxidant enzymes. Polyphenols are a large group of natural compounds ubiquitously expressed in plants, and epidemiological studies have shown associations between a diet rich in polyphenols and the prevention of various ROS-mediated human diseases. Polyphenols reduce cardiomyocytes damage, necrosis, apoptosis, infarct size and improve cardiac function by decreasing oxidative stress-induced production of ROS or RNS. These effects are achieved by the ability of polyphenols to modulate the expression and activity of anti-oxidant enzymes and several signaling pathways involved in cells survival. This report reviews current knowledge on the potential anti-oxidative effects of polyphenols to control the cardiotoxicity induced by ROS and RNS stress.
Collapse
Affiliation(s)
- Rosanna Mattera
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Maria Gabriella Giganti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Ilaria Tresoldi
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | | | - Sonia Bergante
- IRCCS "S. Donato" Hospital, San Donato Milanese, Piazza Edmondo Malan, 20097 Milan, Italy.
| | - Guido Tettamanti
- IRCCS "S. Donato" Hospital, San Donato Milanese, Piazza Edmondo Malan, 20097 Milan, Italy.
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", 00164 Rome, Italy.
| | - Vittorio Manzari
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
- Center for Regenerative Medicine (CIMER), University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
- Center for Regenerative Medicine (CIMER), University of Rome "Tor Vergata", 00133 Rome, Italy.
| |
Collapse
|
45
|
Feng J, Yang Y, Zhou Y, Wang B, Xiong H, Fan C, Jiang S, Liu J, Ma Z, Hu W, Li T, Feng X, Xu J, Jin Z. Bakuchiol attenuates myocardial ischemia reperfusion injury by maintaining mitochondrial function: the role of silent information regulator 1. Apoptosis 2016; 21:532-45. [PMID: 27000151 DOI: 10.1007/s10495-016-1225-6] [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] [Indexed: 01/10/2023]
Abstract
Ischemia reperfusion (IR) injury (IRI) is associated with poor prognoses in the settings of both cardiac surgery and ischemic heart disease and causes mitochondrial oxidative stress and cell death. Silent information regulator 1 (SIRT1), a member of the histone deacetylase family, exerts anti-IRI effects. Bakuchiol (BAK), an analog of resveratrol and a monoterpene phenol isolated from the seeds of Psoralea corylifolia (Leguminosae), protects tissues from injury. This study was designed to investigate the protective effects of BAK treatment in the setting of myocardial IRI and to elucidate the potential mechanism of those effects. Prior to induction of IR, isolated rat hearts or cardiomyocytes were exposed to BAK in either the absence or presence of the SIRT1 inhibitors Sirtinol and SIRT1 siRNA. BAK exerted cardioprotective effects, as evidenced by the improvements noted in cardiac function following ischemia, attenuated myocardial apoptosis, and changes in several biochemical parameters (including increases in the level of the anti-apoptotic protein Bcl2, decreases in the level of the pro-apoptotic protein Bax, and decreases in the cleaved Caspase 3 level). However, Sirtinol and SIRT1 siRNA each blocked BAK-induced cardioprotection by inhibiting SIRT1 signaling. Additionally, BAK significantly increased the activities of mitochondrial succinate dehydrogenase, cytochrome c oxidase, and mitochondrial superoxide dismutase and decreased the production of malondialdehyde. These findings suggested that BAK significantly attenuated IR-induced mitochondrial oxidative damage. However, Sirtinol and SIRT1 siRNA abolished BAK-dependent mitochondrial function. In summary, our results demonstrate that BAK treatment attenuates IRI by attenuating IR-induced mitochondrial oxidative damage via the activation of SIRT1/PGC-1α signaling.
Collapse
Affiliation(s)
- Jianyu Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Yang Yang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Yajun Zhou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China
- Department of Cardiothoracic Surgery, The 94th Hospital of Chinese PLA, 1028 Jingangshan Road, Nanchang, 330000, China
| | - Bodong Wang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Hongyan Xiong
- Department of Cardiothoracic Surgery, Central Hospital of Xi'an, 185 Houzaimen Road, Xi'an, 710033, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Shuai Jiang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Jun Liu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Xiao Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Jianjun Xu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China.
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
46
|
An R, Zhao L, Xu J, Xi C, Li H, Shen G, Zhang W, Zhang S, Sun L. Resveratrol alleviates sepsis-induced myocardial injury in rats by suppressing neutrophil accumulation, the induction of TNF-α and myocardial apoptosis via activation of Sirt1. Mol Med Rep 2016; 14:5297-5303. [DOI: 10.3892/mmr.2016.5861] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 08/24/2016] [Indexed: 11/06/2022] Open
|
47
|
Activation of sirtuin 1/3 improves vascular hyporeactivity in severe hemorrhagic shock by alleviation of mitochondrial damage. Oncotarget 2016; 6:36998-7011. [PMID: 26473372 PMCID: PMC4741911 DOI: 10.18632/oncotarget.6076] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 09/24/2015] [Indexed: 12/11/2022] Open
Abstract
Vascular hyporeactivity is one of the major causes responsible for refractory hypotension and associated mortality in severe hemorrhagic shock. Mitochondrial permeability transition (mPT) pore opening in arteriolar smooth muscle cells (ASMCs) is involved in the pathogenesis of vascular hyporeactivity. However, the molecular mechanism underlying mitochondrial injury in ASMCs during hemorrhagic shock is not well understood. Here we produced an in vivo model of severe hemorrhagic shock in adult Wistar rats. We found that sirtuin (SIRT)1/3 protein levels and deacetylase activities were decreased in ASMCs following severe shock. Immunofluorescence staining confirmed reduced levels of SIRT1 in the nucleus and SIRT3 in the mitochondria, respectively. Acetylation of cyclophilin D (CyPD), a component of mPT pore, was increased. SIRT1 activators suppressed mPT pore opening and ameliorated mitochondrial injury in ASMCs after severe shock. Furthermore, administration of SIRT1 activators improved vasoreactivity in rats under severe shock. Our data suggest that epigenetic mechanisms, namely histone post-translational modifications, are involved in regulation of mPT by SIRT1/SIRT3- mediated deacetylation of CyPD. SIRT1/3 is a promising therapeutic target for the treatment of severe hemorrhagic shock.
Collapse
|
48
|
Iwata H. Age-associated events in bovine oocytes and possible countermeasures. Reprod Med Biol 2016; 15:155-164. [PMID: 29259432 PMCID: PMC5715852 DOI: 10.1007/s12522-015-0233-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/19/2015] [Indexed: 01/31/2023] Open
Abstract
Maternal aging profoundly affects oocyte quality. This has become common knowledge in industrialized countries and extensive studies addressing the causes and possible countermeasures against age-associated deterioration of oocytes suggest that mitochondrial dysfunction is a causal factor in infertility. However, almost all studies addressing age-associated events in oocytes have used mice as an animal model, and the reproductive life of mice is very short, making it difficult to study the gradual decline in fertility observed in humans. In the present review, age-associated changes in the quality and quantity of bovine oocytes and possible countermeasures related to mitochondrial quality control are introduced.
Collapse
Affiliation(s)
- Hisataka Iwata
- Tokyo University of AgricultureFunako 1737243‐034AtsugiKanagawaJapan
| |
Collapse
|
49
|
Hu MZ, Zhou B, Mao HY, Sheng Q, Du B, Chen JL, Pang QF, Ji Y. Exogenous Hydrogen Sulfide Postconditioning Protects Isolated Rat Hearts From Ischemia/Reperfusion Injury Through Sirt1/PGC-1α Signaling Pathway. Int Heart J 2016; 57:477-82. [PMID: 27357440 DOI: 10.1536/ihj.15-506] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sirt1 is a highly conserved nicotinamide adenine dinucleotide (NAD(+)) dependent histone deacetylase which plays an important role in heart diseases. Studies performed with Sirt1 activators indicated that it protects cells from ischemia/ reperfusion (I/R) injury. The protective effects of H2S against I/R injury also have been recognized. Hence, the present study was designed to explore whether Sirt1/PGC-1α participates in the protection of exogenous H2S postconditioning against I/R injury in isolated rat hearts. Isolated rat hearts were subjected to 30 minutes of global ischemia followed by 60 minutes of reperfusion after 20 minutes of equilibrium. During this procedure, the hearts were exposed to NaHS (10 μmol/L) treatment in the absence or presence of the selective Sirt1 inhibitor EX-527 (10 μmol/L). NaHS exerted a protective effect on isolated rat hearts subjected to I/R, as shown by the improved expression of Sirt1/PGC-1α associated with restoration of Sirt1 nuclear localization, cardiac function, decreased myocardial infarct size, decreased myocardial enzyme release, and several biochemical parameters, including up-regulation of the ATP and SOD levels, and down-regulation of the MDA level. However, treatment with EX-527 could partially prevent the above effects of NaHS postconditioning. These results indicate that H2S confers protective effects against I/R injury through the activation of Sirt1/PGC1α.
Collapse
|
50
|
Liu MH, Shan J, Li J, Zhang Y, Lin XL. Resveratrol inhibits doxorubicin-induced cardiotoxicity via sirtuin 1 activation in H9c2 cardiomyocytes. Exp Ther Med 2016; 12:1113-1118. [PMID: 27446329 DOI: 10.3892/etm.2016.3437] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/25/2016] [Indexed: 12/22/2022] Open
Abstract
Doxorubicin (DOX) is an efficient drug used in cancer therapy; however, it can induce severe cytotoxicity, which limits its clinical application. In the present study, the effects of resveratrol (RES) on sirtuin 1 (SIRT1) activation in mediating DOX-induced cytotoxicity in H9c2 cardiac cells was investigated. H9c2 cells were exposed to 5 µM DOX for 24 h to establish a model of DOX cardiotoxicity. Apoptosis of H9c2 cardiomyocytes was assessed using the MTT assay and Hoechst nuclear staining. The results demonstrated that pretreating H9c2 cells with RES prior to the exposure of DOX resulted in increased cell viability and a decreased quantity of apoptotic cells. Western blot analysis demonstrated that DOX decreased the expression level of SIRT1. These effects were significantly alleviated by co-treatment with RES. In addition, the results demonstrated that DOX administration amplified forkhead box O1 (FoxO1) and P53 expression levels in H9c2 cells. RES was also found to protect against DOX-induced increases of FoxO1 and P53 expression levels in H9c2 cells. Furthermore, the protective effects of RES were arrested by the SIRT1 inhibitor nicotinamide. In conclusion, the results demonstrated that RES protected H9c2 cells against DOX-induced injuries via SIRT1 activation.
Collapse
Affiliation(s)
- Mi-Hua Liu
- Department of Clinical Laboratory, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jian Shan
- Department of Pathology, Zhongshan Torch Development Zone Hospital, Zhongshan, Guangdong 528437, P.R. China
| | - Jian Li
- Department of Ultrasonic Diagnosis, Bo'Ai Hospital of Zhongshan, Zhongshan, Guangdong 528403, P.R. China
| | - Yuan Zhang
- Department of Pathology, Mawangdui Hospital, Changsha, Hunan 410016, P.R. China
| | - Xiao-Long Lin
- Department of Pathology, The Third People's Hospital of Huizhou, Affiliated Huizhou Hospital of Guangzhou Medical University, Huizhou, Guangdong 516002, P.R. China
| |
Collapse
|