1
|
Xiang G, Shi T, Nwaele CO, Xiao H, Liu Y, Wang Q, Zhang J, Zheng Y. Inhibition of the Sp1/PI3K/AKT signaling pathway exacerbates doxorubicin-induced cardiomyopathy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119960. [PMID: 40246177 DOI: 10.1016/j.bbamcr.2025.119960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/06/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
OBJECTIVE This study aimed to investigate the interaction and underlying mechanisms between specificity protein 1 (Sp1) and the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signaling pathway in the context of doxorubicin-induced cardiomyopathy (DIC). METHODS A rat model of DIC was established by intraperitoneal injection of doxorubicin (1 mg/kg) twice a week for eight weeks. Cardiac function was evaluated using echocardiography, and myocardial histopathology was assessed by hematoxylin-eosin (HE) staining. In vitro, H9c2 cardiomyocytes were treated with doxorubicin (2 μmol/L) to induce cardiotoxicity, followed by co-treatment with the Sp1 inhibitor plicamycin or the PI3K/AKT inhibitor LY294002. Cell viability was measured by the CCK-8 assay. Oxidative stress markers, including reactive oxygen species (ROS) and lactate dehydrogenase (LDH), were quantified using flow cytometry and colorimetric assays. Apoptosis was detected via TUNEL staining, and protein expression of Sp1, PI3K, AKT, and Caspase-3 was analyzed by Western blotting. RESULTS Doxorubicin treatment significantly impaired cardiac function in rats, as evidenced by an increase in both left ventricular internal diameters during diastole (LVIDd) and systole (LVIDs), along with decreased ejection fraction (EF) and fractional shortening (FS) (p < 0.01). Myocardial HE staining in doxorubicin-treated rats revealed disorganized cardiomyocyte structures, edema, and cellular necrosis. In vitro, doxorubicin exposure led to reduced H9c2 cell viability, elevated ROS and LDH levels, and increased apoptosis rates (p < 0.01). Western blotting demonstrated that doxorubicin significantly downregulated the expression of Sp1, PI3K, and AKT while upregulating Caspase-3. Inhibition of Sp1 or PI3K/AKT exacerbated these effects, resulting in further cardiac dysfunction, oxidative stress, and apoptosis. Moreover, Sp1 inhibition led to decreased PI3K/AKT pathway activation, while PI3K/AKT inhibition reciprocally suppressed Sp1 expression, indicating a bidirectional regulatory relationship. CONCLUSION Doxorubicin induces cardiotoxicity by promoting oxidative stress and apoptosis through the downregulation of the Sp1/PI3K/AKT signaling pathway. Inhibition of this pathway exacerbates cardiac injury, suggesting that targeting Sp1 and PI3K/AKT may offer novel therapeutic strategies for the prevention and treatment of DIC.
Collapse
Affiliation(s)
- Guojian Xiang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Tingting Shi
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian, China; Fuzhou Jian Jia Rehabilitation Hospital, Fuzhou 350007, Fujian, China
| | | | - Huazhen Xiao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Yucheng Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Qingfeng Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Jiancheng Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology in South Branch, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China.
| | - Yonghong Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian, China.
| |
Collapse
|
2
|
Xu Q, Lin R, Jiang T, Deng L, Wu Y, Yuan Q, Qi X, Mu P, Jiang J, Deng Y, Wen J. Enhanced SIRT1-TFEB Interaction Promotes Lysosome Biogenesis and Autophagy by Reducing TFEB Acetylation: Revealing the Enterotoxicity Disparity of Deoxynivalenol and T-2 Toxin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40373210 DOI: 10.1021/acs.jafc.5c01854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Multiple environmental factors contribute to digestive system damage caused by food contamination in both humans and animals. Mycotoxins, such as deoxynivalenol (DON) and T-2 toxin, have emerged as the most significant factors due to their extensive contamination and difficulty in removal. Transcription factor EB (TFEB) serves as a crucial transcriptional regulator governing lysosomal biogenesis and autophagy, a lysosomal-driven degradation system that safeguards cells against harmful stressors. However, little is known about whether the post-translational modification of TFEB affects autophagy activity, which could explain the toxicity disparity between DON and T-2 toxin. Here, we discovered that T-2 toxin induces excessive autophagy by significantly reducing TFEB acetylation, whereas DON surprisingly inhibits autophagy activity via maintaining high TFEB acetylation, which impairs lysosomal biogenesis, thereby boosting their respective toxicity. Mechanically, the T-2 toxin decreases TFEB acetylation via enhanced SIRT1-TFEB interaction and SIRT1 deacetylase activity, while DON maintains high TFEB acetylation by reversing the process. Together, our study revealed that the acetylation state of TFEB mediated by SIRT1 alters autophagy phenotypes in intestinal cells, shedding light on the various toxicological mechanisms and an important target of DON and T-2 toxin.
Collapse
Affiliation(s)
- Qiang Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510640, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Ruqin Lin
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Tianqing Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Luyu Deng
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yuting Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Qianqian Yuan
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xueying Qi
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Peiqiang Mu
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jun Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yiqun Deng
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510640, China
| | - Jikai Wen
- State Key Laboratory of Swine and Poultry Breeding Industry, South China Agricultural University, Guangzhou, Guangdong 510642, China
- Guangdong Provincial Key Laboratory for the Development Biology and Environmental Adaptation of Agricultural Organisms, South China Agricultural University, Guangzhou, Guangdong 510642, China
| |
Collapse
|
3
|
Yan JJ, Wang YY, Shi ZY, Ding YY, Wen HQ, Wu MP, Sun SC, Cai YF, Zhang Y. SIRT5 modulates mitochondria function via mitophagy and antioxidant mechanisms to facilitate oocyte maturation in mice. Int J Biol Macromol 2025; 306:141488. [PMID: 40015402 DOI: 10.1016/j.ijbiomac.2025.141488] [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: 01/03/2025] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
Mitochondrial homeostasis, closely associated with mitophagy and antioxidant mechanisms, is essential for proper meiotic spindle assembly and chromosome segregation during oocyte maturation. SIRT5, known to modulate mitochondrial function under various conditions, has been shown to impact oocyte quality when inhibited, however, the precise mechanisms linking SIRT5 to mitochondrial homeostasis during meiotic progression remain unclear. In this study, we demonstrate that SIRT5 localizes predominantly at the periphery of the meiotic spindle and is enriched on chromosomes during oocyte maturation. Inhibition of SIRT5 led to significant meiotic defects, including disrupted spindle organization and chromosome misalignment. These defects were associated with increased histone acetylation, which impaired kinetochore-microtubule attachments. Moreover, SIRT5 inhibition resulted in mitochondrial dysfunction, subsequently elevating ROS levels and triggering oxidative stress, which further exacerbated meiotic abnormalities. Mechanistically, SIRT5 inhibition disrupted the balance of Parkin-dependent mitophagy by inducing ULK phosphorylation. Additionally, it activated the PI3K/Akt signaling pathway, which increased NADPH consumption and reduced GSH levels. Collectively, these findings reveal that SIRT5 plays dual roles in maintaining mitochondrial homeostasis during oocyte maturation: (1) by regulating Parkin-dependent mitophagy to prevent excessive mitochondrial clearance, and (2) by preserving the NADPH/GSH antioxidant system to ensure redox balance. These insights provide potential targets for improving oocyte quality and addressing mitochondrial dysfunction-related reproductive disorders in females.
Collapse
Affiliation(s)
- Jing-Jing Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yan-Yu Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhi-Yu Shi
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuan-Yuan Ding
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Hao-Quan Wen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Meng-Ping Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Ya-Fei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
4
|
Li M, Zhang H, Wu X, Yu M, Yang Q, Sun L, Li W, Jiang Z, Xue F, Wang T, An X, Chen L. IDH1 regulates human erythropoiesis by eliciting chromatin state reprogramming. eLife 2025; 13:RP100406. [PMID: 40299922 PMCID: PMC12040319 DOI: 10.7554/elife.100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2025] Open
Abstract
Isocitrate dehydrogenase 1 (IDH1) is the key enzyme that can modulate cellular metabolism, epigenetic modification, and redox homeostasis. Gain-of-function mutations and decreased expression of IDH1 have been demonstrated to be associated with pathogenesis of various myeloid malignancies characterized by ineffective erythropoiesis, such as acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). However, the function and mechanism of IDH1 in human erythropoiesis still remains unclear. Here, utilizing the human erythropoiesis system, we present an evidence of IDH1-mediated chromatin state reprogramming besides its well-characterized metabolism effects. We found that knockdown IDH1 induced chromatin reorganization and subsequently led to abnormalities biological events in erythroid precursors, which could not be rescued by addition of reactive oxygen species (ROS) scavengers or supplementation of α-ketoglutarate (α-KG).We further revealed that knockdown IDH1 induces genome-wide changes in distribution and intensity of multiple histone marks, among which H3K79me3 was identified as a critical factor in chromatin state reprogramming. Integrated analysis of ChIP-seq, ATAC-seq, and RNA-seq recognized that SIRT1 was the key gene affected by IDH1 deficiency. Thus, our current work provided novel insights for further clarifying fundamental biological function of IDH1 which has substantial implications for an in-depth understanding of pathogenesis of diseases with IDH1 dysfunction and accordingly development of therapeutic strategies.
Collapse
Affiliation(s)
- Mengjia Li
- State Key Laboratory of Metabolic Dysregulation and Prevention and Treatment of Esophageal Cancer; School of Life Sciences, Zhengzhou UniversityZhengzhouChina
- Department of Hematology, First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hengchao Zhang
- State Key Laboratory of Metabolic Dysregulation and Prevention and Treatment of Esophageal Cancer; School of Life Sciences, Zhengzhou UniversityZhengzhouChina
| | - Xiuyun Wu
- State Key Laboratory of Metabolic Dysregulation and Prevention and Treatment of Esophageal Cancer; School of Life Sciences, Zhengzhou UniversityZhengzhouChina
| | - Mengqi Yu
- State Key Laboratory of Metabolic Dysregulation and Prevention and Treatment of Esophageal Cancer; School of Life Sciences, Zhengzhou UniversityZhengzhouChina
| | - Qianqian Yang
- State Key Laboratory of Metabolic Dysregulation and Prevention and Treatment of Esophageal Cancer; School of Life Sciences, Zhengzhou UniversityZhengzhouChina
| | - Lei Sun
- State Key Laboratory of Metabolic Dysregulation and Prevention and Treatment of Esophageal Cancer; School of Life Sciences, Zhengzhou UniversityZhengzhouChina
| | - Wei Li
- Department of Hematology, First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhongxing Jiang
- Department of Hematology, First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fumin Xue
- Department of Gastroenterology, Children's Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Ting Wang
- State Key Laboratory of Metabolic Dysregulation and Prevention and Treatment of Esophageal Cancer; School of Life Sciences, Zhengzhou UniversityZhengzhouChina
| | - Xuili An
- Laboratory of Membrane Biology, New York Blood CenterNew YorkUnited States
| | - Lixiang Chen
- State Key Laboratory of Metabolic Dysregulation and Prevention and Treatment of Esophageal Cancer; School of Life Sciences, Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
5
|
Wang L, Lin F, Miao R, Zhao T, Liu Y, Yang L, Zhang M. Cardiac protection of wogonin in mice with pulmonary fibrosis by regulating Sirt1/ γ-H2AX pathway. Front Pharmacol 2025; 16:1551141. [PMID: 40297134 PMCID: PMC12034711 DOI: 10.3389/fphar.2025.1551141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Background Clinical evidence suggests that pulmonary fibrosis (PF) and heart failure (HF) often co-exist; however, the specific impact of PF on HF remains underexplored. This gap in understanding complicates the management and treatment of HF in patients with PF. Objectives To investigate the effects of PF on cardiac function and myocardial fibrosis using a mouse PF model and evaluate the therapeutic potential of wogonin, a flavonoid compound known for its anti-PF properties. Methods A PF mouse model was established via intratracheal administration of bleomycin (BLM). Starting on day 8 post-BLM treatment, wogonin (50 mg/kg) was intraperitoneally administered every 2 days for 2 weeks. Cardiac function was assessed using echocardiography, while myocardial fibrosis was evaluated through Masson staining. In vitro, H9C2 cardiomyocytes were exposed to CoCl2 or H2O2 for 24 h with or without wogonin (20 μM) treatment. Apoptosis and DNA damage markers were analysed using immunofluorescence, immunoblotting, and the Comet assay. The interaction between wogonin and Sirt1 was examined using biotin-affinity pulldown assays and molecular docking simulations. Results Mice with PF exhibited significant cardiac dysfunction and myocardial fibrosis. Wogonin treatment markedly improved ejection fraction and attenuated myocardial fibrosis in PF mice. Mechanistic studies revealed that wogonin alleviated DNA damage and cardiomyocyte apoptosis by upregulating Sirt1 and downregulating γ-H2AX expression. Docking simulations predicted that wogonin forms a stable complex with Sirt1 through hydrogen-bonding and hydrophobic interactions, which was further validated by biotin-affinity pulldown assays. Conclusion Wogonin exerts protective effects against cardiac dysfunction and fibrosis in PF mice by modulating Sirt1/γ-H2AX-mediated pathways to reduce DNA damage and apoptosis. These findings suggest the potential of wogonin as a therapeutic agent for managing HF associated with PF.
Collapse
Affiliation(s)
- Libo Wang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Fei Lin
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Runran Miao
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Tianhao Zhao
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Yuan Liu
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Lin Yang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Min Zhang
- Department of Cardiology, Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
- King’s College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London, United Kingdom
| |
Collapse
|
6
|
Chen Y, Shan S, Xue Q, Ren Y, Wu Q, Chen J, Yang K, Cao J. Sirtuin1 mitigates hypoxia-induced cardiomyocyte apoptosis in myocardial infarction via PHD3/HIF-1α. Mol Med 2025; 31:100. [PMID: 40087582 PMCID: PMC11909899 DOI: 10.1186/s10020-025-01155-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a leading cause of mortality, characterized by myocardial ischemia that induces cardiomyocyte apoptosis and subsequent cardiac dysfunction. Sirtuin 1 (Sirt1) has emerged as a key regulator of cell survival and apoptosis, particularly under hypoxic conditions. METHODS An AMI animal model was established via ligation of the left anterior descending (LAD) coronary artery. Gene expression in the infarcted region was evaluated at various time points. Sirt1 overexpression and control lentivirus were administered to the peri-infarct region of mice heart. After LAD ligation, assessment on myocardial infarct size, cardiac function, and cardiomyocyte apoptosis were performed. In vitro, primary mouse cardiomyocytes subjected to hypoxia were analyzed for gene expression, while interactions among Sirt1, Phd3, and Hif-1α were explored using diverse treatment approaches. RESULTS A significant reduction in Sirt1 and Phd3 expression, along with an increase in Hif-1α and cleaved caspase-3, was observed in a time-dependent manner post-myocardial infarction (MI). In vitro findings revealed that hypoxia decreased nuclear Sirt1 and cytoplasmic Phd3 levels while promoting a time-dependent increase in Hif-1α and cleaved caspase-3. Furthermore, Sirt1 overexpression enhanced Phd3 expression in cardiomyocytes, suppressed Hif-1α and cleaved caspase-3 levels, and alleviated hypoxia-induced cardiomyocyte apoptosis. Notably, knockdown of Phd3 negated Sirt1's inhibitory effect on Hif-1α, whereas Hif-1α knockdown promoted Sirt1 expression. Sirt1 overexpression reduced infarct size, decreased cardiomyocyte apoptosis, and improved cardiac function. CONCLUSIONS Sirt1 effectively reduces cardiomyocyte apoptosis and myocardial infarction size while enhancing cardiac function post-MI, primarily through the Phd3/Hif-1α signaling pathway.
Collapse
Affiliation(s)
- Yafen Chen
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuyao Shan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Qiqi Xue
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Yan Ren
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Qihong Wu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China
| | - Jiawei Chen
- Department of Cardiology, Shanghai Ninth People'S Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Ke Yang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China.
| | - Jiumei Cao
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
7
|
Zhang H, Wang Y, Wang R, Yi Q, Xu H, Tan B, Zhu J. Kartogenin Improves Doxorubicin-Induced Cardiotoxicity by Alleviating Oxidative Stress and Protecting Mitochondria. Int J Mol Sci 2025; 26:2434. [PMID: 40141078 PMCID: PMC11942266 DOI: 10.3390/ijms26062434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Doxorubicin (DOX) is a common antitumor drug in clinical practice, but its clinical use is limited due to its cardiotoxic side effects. Oxidative stress and mitochondrial damage are involved in DOX-induced cardiotoxicity (DIC). Kartogenin (KGN) has been shown to have a potent ability to resist oxidative stress and maintain mitochondrial homeostasis. But the impact of KGN on DIC has not been reported. This study explores the potential protective effect of KGN on DIC. The effect of KGN on DIC was studied by establishing in vivo and in vitro DIC models. KGN reduced DOX-induced cardiac insufficiency, myocardial injury, oxidative stress damage, and mitochondrial dysfunction. Through network pharmacology and RNA sequencing (RNA-seq), the mechanism of KGN anti-DIC was highly correlated with oxidative stress and mitochondria. These findings suggest that KGN is a valuable and promising strategy for the prevention of doxorubicin cardiotoxicity.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Yunpeng Wang
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Rui Wang
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Qin Yi
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Hao Xu
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
- Department of Clinical Laboratory, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Bin Tan
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| | - Jing Zhu
- Department of Pediatric Research Institute, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Key Laboratory of Children’s Vital Organ Development and Diseases of Chongqing Health Commission, Chongqing 400014, China
| |
Collapse
|
8
|
Feng Z, Zhang N, Wang L, Guan X, Xie Y, Xia YL. CDC20 protects the heart from doxorubicin-induced cardiotoxicity by modulating CCDC69 degradation. Cell Mol Biol Lett 2025; 30:29. [PMID: 40045239 PMCID: PMC11884132 DOI: 10.1186/s11658-025-00708-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
AIMS Doxorubicin (DOX) is a potent anticancer drug; however, it is associated with significant cardiotoxicity. CDC20 is an E3 ubiquitin ligase that plays a role in cell cycle progression and apoptosis in various types of cancers. The involvement of CDC20 in DOX-induced cardiotoxicity (DIC) is poorly understood. Hence, this study aimed to explore the potential role of CDC20 in the development of DIC and assess whether CDC20 influences the antitumor effects of DOX. METHODS AND RESULTS H9C2 cells were treated with DOX, followed by transcriptomic analysis to identify differentially expressed genes. C57BL/6 mice were treated with DOX for 4 weeks after tail vein injection of CDC20 myocardial-specific knockout mice, AAV9-cTNT-(si) CDC20, or intraperitoneal injection of apcin. Cardiac function and pathological changes were evaluated by echocardiography and pathological staining, respectively. The influence of CDC20 on DOX-induced tumor inhibition was assessed in tumor-bearing mice. In vitro analysis involved treating cardiomyocytes with the Ad-CDC20 adenovirus and DOX, followed by proteomic and ubiquitination-related assays to identify potential downstream ubiquitinated CDC20 proteins. Additionally, we investigated the effect of CCDC69 on CDC20-mediated protection against DOX-induced apoptosis using CCDC69 shRNA. Transcriptome analysis revealed that DOX effectively suppressed the expression of CDC20. Cardiomyocyte-specific overexpression of CDC20 in a DOX-induced mouse model of myocardial injury effectively mitigated cardiomyocyte apoptosis, inflammation, fibrosis, and cell atrophy. Our mechanistic investigation revealed that CDC20 attenuates DOX-induced apoptosis by downregulating CCDC69 expression. Moreover, cardiomyocyte-specific overexpression of CDC20 had no effect on the therapeutic efficacy of DOX against tumors. CONCLUSION Our findings indicate that CDC20 safeguards the heart against DOX-induced cardiotoxicity by modulating CCDC69 degradation without compromising the antitumor efficacy of DOX.
Collapse
Affiliation(s)
- Zhenyu Feng
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Lianhe Road 193, Dalian, Liaoning, 116000, People's Republic of China
| | - Ningning Zhang
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Liang Wang
- Department of Pharmacy, Liaoyang City Central Hospital, Liaoyang, People's Republic of China
| | - Xumin Guan
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Lianhe Road 193, Dalian, Liaoning, 116000, People's Republic of China
| | - Yunpeng Xie
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Lianhe Road 193, Dalian, Liaoning, 116000, People's Republic of China.
| | - Yun-Long Xia
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Lianhe Road 193, Dalian, Liaoning, 116000, People's Republic of China.
| |
Collapse
|
9
|
Zhao ST, Qiu ZC, Xu ZQ, Tao ED, Qiu RB, Peng HZ, Zhou LF, Zeng RY, Lai SQ, Wan L. Curcumin attenuates myocardial ischemia‑reperfusion‑induced autophagy‑dependent ferroptosis via Sirt1/AKT/FoxO3a signaling. Int J Mol Med 2025; 55:51. [PMID: 39930816 PMCID: PMC11781526 DOI: 10.3892/ijmm.2025.5492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/04/2024] [Indexed: 02/14/2025] Open
Abstract
Curcumin (Cur) effectively attenuates myocardial ischemia/reperfusion injury (MIRI). MIRI has a complex mechanism and is associated with autophagy‑dependent ferroptosis. Therefore, the present study aimed to determine whether autophagy‑dependent ferroptosis occurs in MIRI and assess the mechanism of Cur in attenuating MIRI. The study was conducted on a Sprague‑Dawley rat MIRI model and H9c2 cell anoxia/reoxygenation (A/R) injury model. The effect of Cur pretreatment on A/R or MIRI induced autophagy‑dependent ferroptosis and its molecular mechanism were investigated. Protein expression, lysosomal, reactive oxygen species, Fe2+, oxidative systems, mitochondrial function, subcellular localization of molecules, and cardiac function assays will be employed. Cur decreased MIRI; improved myocardial histopathology; increased cardiomyocyte viability; inhibited ferroptosis, apoptosis and autophagy; reduced infarct size and maintained cardiac function. MIRI decreased silent information regulator 1 (Sirt1), decreased AKT and forkhead box O3A (FoxO3a) phosphorylation, leading to FoxO3a entry into the nucleus to activate translation of autophagy‑related genes and inducing ferroptosis, apoptosis and autophagy. However, Cur pretreatment activated AKT and FoxO3a phosphorylation via Sirt1, thereby transporting FoxO3a out of the nucleus, reducing autophagy‑related gene translation and attenuating MIRI‑induced ferroptosis, apoptosis and autophagy. Of note, the silencing of Sirt1 and administration of triciribine (an AKT inhibitor) both eliminated the protective effect of Cur. Thus, Cur maintained cardiomyocyte function by inhibiting autophagy‑dependent ferroptosis via Sirt1/AKT/FoxO3a signaling.
Collapse
Affiliation(s)
- Shi-Tao Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhi-Cong Qiu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhi-Qiang Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - En-De Tao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Rong-Bin Qiu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Han-Zhi Peng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lian-Fen Zhou
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Rui-Yuan Zeng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Song-Qing Lai
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li Wan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Institute of Cardiovascular Surgical Diseases, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
10
|
Fiorentino F, Fabbrizi E, Mai A, Rotili D. Activation and inhibition of sirtuins: From bench to bedside. Med Res Rev 2025; 45:484-560. [PMID: 39215785 PMCID: PMC11796339 DOI: 10.1002/med.22076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/27/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024]
Abstract
The sirtuin family comprises seven NAD+-dependent enzymes which catalyze protein lysine deacylation and mono ADP-ribosylation. Sirtuins act as central regulators of genomic stability and gene expression and control key processes, including energetic metabolism, cell cycle, differentiation, apoptosis, and aging. As a result, all sirtuins play critical roles in cellular homeostasis and organism wellness, and their dysregulation has been linked to metabolic, cardiovascular, and neurological diseases. Furthermore, sirtuins have shown dichotomous roles in cancer, acting as context-dependent tumor suppressors or promoters. Given their central role in different cellular processes, sirtuins have attracted increasing research interest aimed at developing both activators and inhibitors. Indeed, sirtuin modulation may have therapeutic effects in many age-related diseases, including diabetes, cardiovascular and neurodegenerative disorders, and cancer. Moreover, isoform selective modulators may increase our knowledge of sirtuin biology and aid to develop better therapies. Through this review, we provide critical insights into sirtuin pharmacology and illustrate their enzymatic activities and biological functions. Furthermore, we outline the most relevant sirtuin modulators in terms of their modes of action, structure-activity relationships, pharmacological effects, and clinical applications.
Collapse
Affiliation(s)
- Francesco Fiorentino
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
| | - Emanuele Fabbrizi
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
| | - Antonello Mai
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
- Pasteur Institute, Cenci‐Bolognetti FoundationSapienza University of RomeRomeItaly
| | - Dante Rotili
- Department of Drug Chemistry and TechnologiesSapienza University of RomeRomeItaly
| |
Collapse
|
11
|
Yu H, Li X, Ning B, Feng L, Ren Y, Li S, Kang Y, Ma J, Zhao M. SIRT1: a potential therapeutic target for coronary heart disease combined with anxiety or depression. J Drug Target 2025; 33:328-340. [PMID: 39470049 DOI: 10.1080/1061186x.2024.2422882] [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: 07/08/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Coronary heart disease (CHD) combined with anxiety or depression is increasingly receiving attention in the clinical field of cardiology, and exploring the comorbidity pathological mechanisms of cardiovascular disease combined with psychological disorders is a hot research topic for scholars in this field. Current research suggests that Silent Information Regulatory Factor 1 (SIRT1) may serve as a potential biomarker for the comorbidity mechanism and treatment of CHD with anxiety or depression. SIRT1 is considered a promising therapeutic target for CHD combined with anxiety or depression, with the ability to regulate inflammatory cytokine levels, alleviate oxidative stress damage, activate multiple signalling pathways, reduce platelet hyperresponsiveness, and exert neuroprotective and cardioprotective effects. In this comprehensive review, we deeply studied the structure, function, and mechanism of SIRT1, and discussed its protective effects in the cardiovascular and nervous system. The latest progress in the mechanism of SIRT1's role in CHD combined with anxiety or depression was emphasised, including its specific mechanisms in regulating inflammatory response, alleviating oxidative stress, and mediating various signalling pathways. In addition, this article also summarises the therapeutic potential of SIRT1 as a potential biomarker in patients with CHD combined with anxiety or depression.
Collapse
Affiliation(s)
- Hubin Yu
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xinping Li
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Bo Ning
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lanshuan Feng
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yaolong Ren
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Shilin Li
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yalong Kang
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jing Ma
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Air Force Military Medical University, Xi'an, China
| | - Mingjun Zhao
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
12
|
Ding YN, Wang HY, Chen XF, Tang X, Chen HZ. Roles of Sirtuins in Cardiovascular Diseases: Mechanisms and Therapeutics. Circ Res 2025; 136:524-550. [PMID: 40014680 DOI: 10.1161/circresaha.124.325440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Cardiovascular diseases (CVDs) are experiencing a rapid surge and are widely recognized as the leading cause of mortality in the current aging society. Given the multifactorial etiology of CVDs, understanding the intricate molecular and cellular mechanisms is imperative. Over the past 2 decades, many scientists have focused on Sirtuins, a family of nicotinamide adenine dinucleotide-dependent deacylases. Sirtuins are highly conserved across species, from yeasts to primates, and play a crucial role in linking aging and diseases. Sirtuins participate in nearly all key physiological and pathological processes, ranging from embryogenic development to stress response and aging. Abnormal expression and activity of Sirtuins exist in many aging-related diseases, while their activation has shown efficacy in mitigating these diseases (eg, CVDs). In terms of research, this field has maintained fast, sustained growth in recent years, from fundamental studies to clinical trials. In this review, we present a comprehensive, up-to-date discussion on the biological functions of Sirtuins and their roles in regulating cardiovascular biology and CVDs. Furthermore, we highlight the latest advancements in utilizing Sirtuin-activating compounds and nicotinamide adenine dinucleotide boosters as potential pharmacological targets for preventing and treating CVDs. The key unresolved issues in the field-from the chemicobiological regulation of Sirtuins to Sirtuin-targeted CVD investigations-are also discussed. This timely review could be critical in understanding the updated knowledge of Sirtuin biology in CVDs and facilitating the clinical accessibility of Sirtuin-targeting interventions.
Collapse
Affiliation(s)
- Yang-Nan Ding
- Department of Laboratory Medicine, Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, The Third Affiliated Hospital of Zhengzhou University, China (Y.-N.D.)
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.-N.D., H.-Y.W., H.-Z.C.)
| | - Hui-Yu Wang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.-N.D., H.-Y.W., H.-Z.C.)
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing (H.-Y.W., H.-Z.C.)
| | - Xiao-Feng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, China (X.-F.C.)
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu (X.T.)
| | - Hou-Zao Chen
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (Y.-N.D., H.-Y.W., H.-Z.C.)
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing (H.-Y.W., H.-Z.C.)
| |
Collapse
|
13
|
Liu X, Li Z. The role and mechanism of epigenetics in anticancer drug-induced cardiotoxicity. Basic Res Cardiol 2025; 120:11-24. [PMID: 38724618 DOI: 10.1007/s00395-024-01054-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/23/2024]
Abstract
Cardiovascular disease is the main factor contributing to the global burden of diseases, and the cardiotoxicity caused by anticancer drugs is an essential component that cannot be ignored. With the development of anticancer drugs, the survival period of cancer patients is prolonged; however, the cardiotoxicity caused by anticancer drugs is becoming increasingly prominent. Currently, cardiovascular disease has emerged as the second leading cause of mortality among long-term cancer survivors. Anticancer drug-induced cardiotoxicity has become a frontier and hot topic. The discovery of epigenetics has given the possibility of environmental changes in gene expression, protein synthesis, and traits. It has been found that epigenetics plays a pivotal role in promoting cardiovascular diseases, such as heart failure, coronary heart disease, and hypertension. In recent years, increasing studies have underscored the crucial roles played by epigenetics in anticancer drug-induced cardiotoxicity. Here, we provide a comprehensive overview of the role and mechanisms of epigenetics in anticancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xuening Liu
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zijian Li
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|
14
|
He Y, Li S, Jiang L, Wu K, Chen S, Su L, Liu C, Liu P, Luo W, Zhong S, Li Z. Palmitic Acid Accelerates Endothelial Cell Injury and Cardiovascular Dysfunction via Palmitoylation of PKM2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412895. [PMID: 39665133 PMCID: PMC11791964 DOI: 10.1002/advs.202412895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/17/2024] [Indexed: 12/13/2024]
Abstract
High serum level of palmitic acid(PA) is implicated in pathogenesis of cardiovascular diseases. PA serves as the substrate for protein palmitoylation. However, it is still unknown whether palmitoylation is involved in PA-induced cardiovascular dysfunction. Here, in clinical cohort studies of 1040 patients with coronary heart disease, high level of PA is associated with risk of major adverse cardiovascular events (MACE) and death. In ApoE-/-mice, 10 mg/kg-1 PA treatment induces blood pressure elevation, cardiac contractile dysfunction, endothelial dysfunction and atherosclerotic plaqueformation. In endothelial cells, inhibition of palmitoylation bypalmitoyl-transferase inhibitor 2-BP eliminates PA-induced endothelial injury, whereas promotion of palmitoylation by depalmitoylase inhibitor ML349 exacerbates the harmful effect of PA. Palmitoyl-proteomics analysis identifies pyruvate kinase isozyme type M2 (PKM2) as the palmitoylated protein responsible for PA-induced endothelial injury, and Cys31 as the predominant palmitoylated site. PKM2-C31S mutants (cysteine replaced by serine) prevents PA-induced endothelial injury. Endothelial-specific AAV-C31S PKM2endo ameliorates cardiovascular dysfunction caused by PA in ApoE-/- mice. Mechanistically, PKM2-C31 palmitoylation impairs PKM2 tetramerization to inhibit its pyruvate kinase activity and endothelial glycolysis. Finally, zDHHC13 is identified as the palmitoyl acyltransferase of PKM2. In conclusion, these findings suggest that PKM2-C31 palmitoylation contributes to PA-induced endothelial injury and cardiovascular dysfunction.
Collapse
Affiliation(s)
- Yu He
- Department of Pharmacology and ToxicologySchool of Pharmaceutical SciencesNational and Local United Engineering Lab of Druggability and New Drugs EvaluationGuangdong Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Provincial Key Laboratory of New Drug Design and EvaluationSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Senlin Li
- Department of PharmacyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080P. R. China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease PreventionGuangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080P. R. China
- School of MedicineSouth China University of TechnologyGuangzhou510006P. R. China
| | - Lujing Jiang
- Department of Pharmacology and ToxicologySchool of Pharmaceutical SciencesNational and Local United Engineering Lab of Druggability and New Drugs EvaluationGuangdong Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Provincial Key Laboratory of New Drug Design and EvaluationSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Kejue Wu
- Department of Pharmacology and ToxicologySchool of Pharmaceutical SciencesNational and Local United Engineering Lab of Druggability and New Drugs EvaluationGuangdong Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Provincial Key Laboratory of New Drug Design and EvaluationSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Shanshan Chen
- Department of Pharmacology and ToxicologySchool of Pharmaceutical SciencesNational and Local United Engineering Lab of Druggability and New Drugs EvaluationGuangdong Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Provincial Key Laboratory of New Drug Design and EvaluationSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Linjie Su
- Department of Pharmacology and ToxicologySchool of Pharmaceutical SciencesNational and Local United Engineering Lab of Druggability and New Drugs EvaluationGuangdong Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Provincial Key Laboratory of New Drug Design and EvaluationSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Cui Liu
- Department of Pharmacology and ToxicologySchool of Pharmaceutical SciencesNational and Local United Engineering Lab of Druggability and New Drugs EvaluationGuangdong Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Provincial Key Laboratory of New Drug Design and EvaluationSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Peiqing Liu
- Department of Pharmacology and ToxicologySchool of Pharmaceutical SciencesNational and Local United Engineering Lab of Druggability and New Drugs EvaluationGuangdong Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Provincial Key Laboratory of New Drug Design and EvaluationSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Wenwei Luo
- Department of PharmacyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080P. R. China
| | - Shilong Zhong
- Department of PharmacyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080P. R. China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease PreventionGuangdong Cardiovascular InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080P. R. China
- School of MedicineSouth China University of TechnologyGuangzhou510006P. R. China
| | - Zhuoming Li
- Department of Pharmacology and ToxicologySchool of Pharmaceutical SciencesNational and Local United Engineering Lab of Druggability and New Drugs EvaluationGuangdong Engineering Laboratory of Druggability and New Drug EvaluationGuangdong Provincial Key Laboratory of New Drug Design and EvaluationSun Yat‐sen UniversityGuangzhou510006P. R. China
| |
Collapse
|
15
|
Zheng H, Yang X, Zhong H, Song C, Wu Z, Yang H. HDAC6 Facilitates PRV and VSV Infection by Inhibiting Type I Interferon Production. Viruses 2025; 17:90. [PMID: 39861880 PMCID: PMC11768819 DOI: 10.3390/v17010090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
HDAC6 modulates viral infection through diverse mechanisms. Here, we investigated the role of HDAC6 in influencing viral infection in pig cells with the aim of exploiting the potential antiviral gene targets in pigs. Using gene knockout and overexpression strategies, we found that HDAC6 knockout greatly reduced PRV and VSV infectivity, whereas HDAC6 overexpression increased their infectivity in PK15 cells. Mechanistic studies identified HDAC6 as a DNA damage inhibitor in PK15 cells. HDAC6 overexpression attenuated DNA damage levels, which can further reduce type I IFN production to promote viral infection. Conversely, HDAC6 deficiency can limit viral infection by increasing DNA damage-mediated type I IFN production. This work demonstrates that HDAC6 affects the infection process of multiple viruses by modulating type I IFN production, highlighting a regulatory role of HDAC6 linking host immune response and viral infection levels in pig cells.
Collapse
Affiliation(s)
- Hu Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Xiaohui Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Haiwen Zhong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Changxu Song
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu 527400, China
| | - Huaqiang Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu 527400, China
| |
Collapse
|
16
|
Ge W, Zhang X, Lin J, Wang Y, Zhang X, Duan Y, Dai X, Zhang J, Zhang Y, Jiang M, Qiang H, Zhao Z, Zhang X, Sun D. Rnd3 protects against doxorubicin-induced cardiotoxicity through inhibition of PANoptosis in a Rock1/Drp1/mitochondrial fission-dependent manner. Cell Death Dis 2025; 16:2. [PMID: 39755713 DOI: 10.1038/s41419-024-07322-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
Doxorubicin, a representative drug of the anthracycline class, is widely used in cancer treatment. However, Doxorubicin-induced cardiotoxicity (DIC) presents a significant challenge in its clinical application. Mitochondrial dysfunction plays a central role in DIC, primarily through disrupting mitochondrial dynamics. This study aimed to investigate the impact of Rnd3 (a Rho family GTPase 3) on DIC, with a focus on mitochondrial dynamics. Cardiomyocyte-specific Rnd3 transgenic mice (Rnd3-Tg) and Rnd3LSP/LSP mice (N-Tg) were established for in vivo experiments, and adenoviruses harboring Rnd3 (Ad-Rnd3) or negative control (Ad-Control) were injected in the myocardium for in vitro experiments. The DIC model was established using wild-type, N-Tg, and Rnd3-Tg mice, with subsequent intraperitoneal injection of Dox for 4 weeks. The molecular mechanism was explored through RNA sequencing, immunofluorescence staining, co-immunoprecipitation assay, and protein-protein docking. Dox administration induced significant mitochondrial injury and cardiac dysfunction, which was ameliorated by Rnd3 overexpression. Further, the augmentation of Rnd3 expression mitigated mitochondrial fragmentation which is mediated by dynamin-related protein 1 (Drp1), thereby ameliorating the PANoptosis (pyroptosis, apoptosis, and necroptosis) response induced by Dox. Mechanically, the interaction between Rnd3 and Rho-associated kinase 1 (Rock1) may impede Rock1-induced Drp1 phosphorylation at Ser616, thus inhibiting mitochondrial fission and dysfunction. Interestingly, Rock1 knockdown nullified the effects of Rnd3 on cardiomyocytes PANoptosis, as well as Dox-induced cardiac remodeling and dysfunction elicited by Rnd3. Rnd3 enhances cardiac resilience against DIC by stabilizing mitochondrial dynamics and reducing PANoptosis. Our findings suggest that the Rnd3/Rock1/Drp1 signaling pathway represents a novel target for mitigating DIC, and modulating Rnd3 expression could be a strategic approach to safeguarding cardiac function in patients undergoing Dox treatment. The graphical abstract illustrated the cardioprotective role of Rnd3 in DIC. Rnd3 directly binds to Rock1 in cytoplasm and ameliorates mitochondrial fission by inhibiting Drp1 phosphorylation at ser616, thereby alleviating PANoptosis (apoptosis, pyroptosis, and necroptosis) in DIC.
Collapse
Affiliation(s)
- Wen Ge
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaohua Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yangyang Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yu Duan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xinchun Dai
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huanhuan Qiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhijing Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
17
|
Kuno A, Hosoda R, Saga Y, Iwahara N, Tatekoshi Y, Numazawa R, Horio Y. Resveratrol promotes autophagosome elimination via SIRT1 in cardiomyocytes. J Pharmacol Sci 2025; 157:25-34. [PMID: 39706642 DOI: 10.1016/j.jphs.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/15/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
The processes of autophagy, including autophagosome formation, fusion of autophagosomes with lysosomes, and degradation of autophagosomes by lysosomes, are regulated by various mechanisms. We recently found that treatment with resveratrol, an activator of the NAD+-dependent protein deacetylase Sirtuin-1 (SIRT1), in a mouse model prevented autophagosome accumulation in the heart with high mTORC1 activity. In this study, we investigated whether SIRT1 mediates the effects of resveratrol on autophagosome elimination using a cardiomyocyte model. In H9c2 cardiomyocytes, treatment with the mTORC1 activator MHY1485 induced autophagosome accumulation accompanied by increases in fragmented mitochondria within the autophagosomes and levels of intracellular reactive oxygen species (ROS), indicative of impaired autophagy-mediated elimination of mitochondria and resultant oxidative stress. MHY1485 suppressed the fusion of autophagosomes with lysosomes. Co-treatment with resveratrol attenuated the MHY1485-induced increases in autophagosomes, mitochondria within autophagosomes, and levels of ROS. Knockdown of Sirt1 reversed the reductions in autophagosomes and ROS levels induced by resveratrol under the condition of MHY1485 treatment. Neither resveratrol treatment nor Sirt1 knockdown modulated the phosphorylation levels of UVRAG, a target of mTORC1 for suppression of autophagosome-lysosome fusion. Our findings suggest that SIRT1 mediates the resveratrol-induced promotion of autophagosome elimination in cells with high mTORC1 activity.
Collapse
Affiliation(s)
- Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Ryusuke Hosoda
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yukika Saga
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Naotoshi Iwahara
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Neurology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuki Tatekoshi
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ryo Numazawa
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yoshiyuki Horio
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
18
|
Yang TT, Zhou LH, Gu LF, Qian LL, Bao YL, Jing P, Sun JT, Du C, Shan TK, Wang SB, Wang WJ, Chen JY, Wang ZM, Wang H, Wang QM, Wang RX, Wang LS. CHK1 attenuates cardiac dysfunction via suppressing SIRT1-ubiquitination. Metabolism 2025; 162:156048. [PMID: 39454820 DOI: 10.1016/j.metabol.2024.156048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/21/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Mitochondrial dysfunction is linked to myocardial ischemia-reperfusion (I/R) injury. Checkpoint kinase 1 (CHK1) could facilitate cardiomyocyte proliferation, however, its role on mitochondrial function in I/R injury remains unknown. METHODS To investigate the role of CHK1 on mitochondrial function following I/R injury, cardiomyocyte-specific knockout/overexpression mouse models were generated. Adult mouse cardiomyocytes (AMCMs) were isolated for in vitro study. Mass spectrometry-proteomics analysis and protein co-immunoprecipitation assays were conducted to dissect the molecular mechanism. RESULTS CHK1 was downregulated in myocardium post I/R and AMCMs post oxygen-glucose deprivation/re‑oxygenation (OGD/R). In vivo, CHK1 overexpression protected against I/R induced cardiac dysfunction, while heterogenous CHK1 knockout exacerbated cardiomyopathy. In vitro, CHK1 inhibited OGD/R-induced cardiomyocyte apoptosis and bolstered cardiomyocyte survival. Mechanistically, CHK1 attenuated oxidative stress and preserved mitochondrial metabolism in cardiomyocytes under I/R. Moreover, disrupted mitochondrial homeostasis in I/R myocardium was restored by CHK1 through the promotion of mitochondrial biogenesis and mitophagy. Through mass spectrometry analysis following co-immunoprecipitation, SIRT1 was identified as a direct target of CHK1. The 266-390 domain of CHK1 interacted with the 160-583 domain of SIRT1. Importantly, CHK1 phosphorylated SIRT1 at Thr530 residue, thereby inhibiting SMURF2-mediated degradation of SIRT1. The role of CHK1 in maintaining mitochondrial dynamics control and myocardial protection is abolished by SIRT1 inhibition, while inactivated mutation of SIRT1 Thr530 fails to reverse the impaired mitochondrial dynamics following CHK1 knockdown. CHK1 Δ390 amino acids (aa) mutant functioned similarly to full-length CHK1 in scavenging ROS and maintaining mitochondrial dynamics. Consistently, cardiac-specific SIRT1 knockdown attenuated the protective role of CHK1 in I/R injury. CONCLUSIONS Our findings revealed that CHK1 mitigates I/R injury and restores mitochondrial dynamics in cardiomyocytes through a SIRT1-dependent mechanism.
Collapse
Affiliation(s)
- Tong-Tong Yang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Liu-Hua Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ling-Feng Gu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ling-Ling Qian
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical center, Nanjing Medical University, Wuxi 214023, China
| | - Yu-Lin Bao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Peng Jing
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jia-Teng Sun
- Department of Cardiology, Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, China
| | - Chong Du
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Tian-Kai Shan
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Si-Bo Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wen-Jing Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jia-Yi Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ze-Mu Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qi-Ming Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ru-Xing Wang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical center, Nanjing Medical University, Wuxi 214023, China.
| | - Lian-Sheng Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
19
|
Qiu Y, Xu Q, Xie P, He C, Li Q, Yao X, Mao Y, Wu X, Zhang T. Epigenetic modifications and emerging therapeutic targets in cardiovascular aging and diseases. Pharmacol Res 2025; 211:107546. [PMID: 39674563 DOI: 10.1016/j.phrs.2024.107546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
The complex mechanisms underlying the development of cardiovascular diseases remain not fully elucidated. Epigenetics, which modulates gene expression without DNA sequence changes, is shedding light on these mechanisms and their heritable effects. This review focus on epigenetic regulation in cardiovascular aging and diseases, detailing specific epigenetic enzymes such as DNA methyltransferases (DNMTs), histone acetyltransferases (HATs), and histone deacetylases (HDACs), which serve as writers or erasers that modify the epigenetic landscape. We also discuss the readers of these modifications, such as the 5-methylcytosine binding domain proteins, and the erasers ten-eleven translocation (TET) proteins. The emerging role of RNA methylation, particularly N6-methyladenosine (m6A), in cardiovascular pathogenesis is also discussed. We summarize potential therapeutic targets, such as key enzymes and their inhibitors, including DNMT inhibitors like 5-azacytidine and decitabine, HDAC inhibitors like belinostat and givinotide, some of which have been approved by the FDA for various malignancies, suggesting their potential in treating cardiovascular diseases. Furthermore, we highlight the role of novel histone modifications and their associated enzymes, which are emerging as potential therapeutic targets in cardiovascular diseases. Thus, by incorporating the recent studies involving patients with cardiovascular aging and diseases, we aim to provide a more detailed and updated review that reflects the advancements in the field of epigenetic modification in cardiovascular diseases.
Collapse
Affiliation(s)
- Yurou Qiu
- GMU-GIBH Joint School of Life Sciences, Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, The Sixth School of Clinical Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Qing Xu
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Peichen Xie
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Chenshuang He
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Qiuchan Li
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Xin Yao
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Yang Mao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Xiaoqian Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| | - Tiejun Zhang
- GMU-GIBH Joint School of Life Sciences, Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, The Sixth School of Clinical Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
20
|
Cimmino A, Gioia M, Clementi ME, Faraoni I, Marini S, Ciaccio C. Polydatin-Induced Shift of Redox Balance and Its Anti-Cancer Impact on Human Osteosarcoma Cells. Curr Issues Mol Biol 2024; 47:21. [PMID: 39852136 PMCID: PMC11764470 DOI: 10.3390/cimb47010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Cancer cells demonstrate remarkable resilience by adapting to oxidative stress and undergoing metabolic reprogramming, making oxidative stress a critical target for cancer therapy. This study explores, for the first time, the redox-dependent anticancer effects of Polydatin (PD), a glucoside derivative of resveratrol, on the human Osteosarcoma (OS) cells SAOS-2 and U2OS. Using cell-based biochemical assays, we found that cytotoxic doses of PD (100-200 µM) promote ROS production, deplete glutathione (GSH), and elevate levels of both total iron and intracellular malondialdehyde (MDA), which are key markers of ferroptosis. Notably, the ROS scavenger N-acetylcysteine (NAC) and the ferroptosis inhibitor ferrostatin-1 (Fer-1) partially reverse PD's cytotoxic effects. Interestingly, PD's ability to hinder cell adhesion and migration appears independent of its pro-oxidant effect. Analysis of the oxidative stress regulators SIRT1 and Nrf2 at the gene and protein levels using real-time PCR and Western blot indicates an early oxidative response to PD treatment. PD remains effective under tumor-like conditions of hypoxia and serum starvation, and sensitizes OS cells to ROS-inducing chemotherapeutics like doxorubicin (DOX) and cisplatin (CIS). Importantly, PD exhibits minimal toxicity to non-tumorigenic cells (hFOB), suggesting a favorable therapeutic profile. Overall, our findings underscore that PD-induced redox imbalance plays a crucial role in its anti-OS effects, warranting further exploration into the molecular mechanisms behind its pro-oxidant activity.
Collapse
Affiliation(s)
- Alessio Cimmino
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy; (A.C.); (M.G.); (S.M.)
| | - Magda Gioia
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy; (A.C.); (M.G.); (S.M.)
| | - Maria Elisabetta Clementi
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta” SCITEC-CNR, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Isabella Faraoni
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy;
| | - Stefano Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy; (A.C.); (M.G.); (S.M.)
| | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Rome ‘Tor Vergata’, Via Montpellier 1, 00133 Rome, Italy; (A.C.); (M.G.); (S.M.)
| |
Collapse
|
21
|
Zhang S, Yang Y, Lv X, Zhou X, Zhao W, Meng L, Xu H, Zhu S, Wang Y. Doxorubicin-Induced Cardiotoxicity Through SIRT1 Loss Potentiates Overproduction of Exosomes in Cardiomyocytes. Int J Mol Sci 2024; 25:12376. [PMID: 39596439 PMCID: PMC11594621 DOI: 10.3390/ijms252212376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Mutual interaction between doxorubicin (DOX) and cardiomyocytes is crucial for cardiotoxicity progression. Cardiomyocyte injury is an important pathological feature of DOX-induced cardiomyopathy, and its molecular pathogenesis is multifaceted. In addition to the direct toxic effects of DOX on cardiomyocytes, DOX-induced exosomes in the extracellular microenvironment also regulate the pathophysiological states of cardiomyocytes. However, the mechanisms by which DOX regulates exosome secretion and subsequent pathogenesis remain incompletely understood. Here, we found that DOX significantly increased exosome secretion from cardiomyocytes, and inhibiting this release could alleviate cardiomyocyte injury. DOX promoted exosome secretion by reducing cardiomyocyte silencing information regulator 1 (SIRT1) expression, exacerbating cardiotoxicity. DOX impaired lysosomal acidification in cardiomyocytes, reducing the degradation of intracellular multivesicular bodies (MVBs), resulting in an increase in MVB volume before fusing with the plasma membrane to release their contents. Mechanistically, SIRT1 loss inhibited lysosomal acidification by reducing the expression of the ATP6V1A subunit of the lysosomal vacuolar-type H+ ATPase (V-ATPase) proton pump. Overexpressing SIRT1 increased ATP6V1A expression, improved lysosomal acidification, inhibited exosome secretion, and thereby alleviated DOX-induced cardiotoxicity. Interestingly, DOX also induced mitochondrial-derived vesicle formation in cardiomyocytes, which may further increase the abundance of MVBs and promote exosome release. Collectively, this study identified SIRT1-mediated impairment of lysosomal acidification as a key mechanism underlying the increased exosome secretion from cardiomyocytes induced by DOX, providing new insights into DOX-induced cardiotoxicity pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ying Wang
- Department of Forensic Medicine, School of Basic Medicine, Soochow University, Suzhou 215123, China; (S.Z.)
| |
Collapse
|
22
|
Oyovwi MO, Ugwuishi EW, Udi OA, Uchechukwu GJ. Mitophagy Unveiled: Exploring the Nexus of Mitochondrial Health and Neuroendocrinopathy. J Mol Neurosci 2024; 74:107. [PMID: 39514132 DOI: 10.1007/s12031-024-02280-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Mitochondria play a pivotal role in cellular metabolism, energy production, and apoptotic signaling, making mitophagy, the selective degradation of damaged mitochondria, crucial for mitochondrial health. Dysregulation of mitophagy has been implicated in various neuroendocrinopathies, yet the mechanisms linking these processes remain poorly understood. This review aims to explore the intersection between mitophagy and neuroendocrinopathy, addressing the critical gaps in knowledge regarding how mitochondrial dysfunction may contribute to the pathophysiology of neuroendocrine disorders. We conducted a comprehensive literature review of studies published on mitophagy and neuroendocrinopathies, focusing on data that elucidate the pathways involved and the clinical implications of mitochondrial health in neuroendocrine contexts. Our findings indicate that altered mitophagy may lead to the accumulation of dysfunctional mitochondria, contributing to neuroendocrine dysregulation. We present evidence linking impaired mitochondrial clearance to disease models of conditions such as metabolic syndrome, depression, and stress-related disorders, highlighting the potential for therapeutic interventions targeting mitophagy. While significant advances have been made in understanding mitochondrial biology, the direct interplay between mitophagy and neuroendocrinopathies remains underexplored. This review underscores the necessity for further research to elucidate these connections, which may offer novel insights into disease mechanisms and therapeutic strategies for treating maladaptive neuroendocrine responses.
Collapse
Affiliation(s)
- Mega Obukohwo Oyovwi
- Department of Physiology, Faculty of Basic Medical Sciences, Adeleke University, Ede, Osun State, Nigeria.
| | | | - Onoriode Andrew Udi
- Department of Human Anatomy, Federal University Otuoke, Otuoke, Bayelsa State, Nigeria
| | - Gregory Joseph Uchechukwu
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, Adeleke University, Ede, Osun State, Nigeria
| |
Collapse
|
23
|
Yu Z, Teng Y, Yang H, Wang Y, Li X, Feng L, Xu W, Hao Y, Li Y. Inhibiting H2AX Can Ameliorate Myocardial Ischemia/Reperfusion Injury by Regulating P53/JNK Signaling Pathway. Cardiol Res Pract 2024; 2024:1905996. [PMID: 39257436 PMCID: PMC11387088 DOI: 10.1155/2024/1905996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/13/2024] [Indexed: 09/12/2024] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a significant area of focus in cardiovascular disease research. I/R injury can increase intracellular oxidative stress, leading to DNA damage. H2AX plays a crucial role in DNA repair. This study utilized mouse and cell models of myocardial I/R to investigate the impact of H2AX on cardiomyocytes during I/R. This study initially assessed the expression of H2AX in MI/R mice compared to a sham surgery group. Subsequently, cardiac function, infarct area, and mitochondrial damage were evaluated after inhibiting H2AX in MI/R mice and a negative control group. Furthermore, the study delved into the molecular mechanisms by analyzing the expression of H2AX, P53, p-JNK, SHP2, p-SHP2, p-RAS, parkin, Drp1, Cyt-C, Caspase-3, and Caspase-8 in cardiomyocytes following the addition of JNK or P53 agonists. The results from western blotting in vivo indicated significantly higher H2AX expression in the MI/R group compared to the sham group. Inhibiting H2AX improved cardiac function, reduced myocardial infarct area, and mitigated mitochondrial damage in the MI/R group. In vitro experiments demonstrated that inhibiting H2AX could attenuate mitochondrial damage and apoptosis in myocardial cells by modulating the P53 and JNK signaling pathways. These findings suggested that inhibiting H2AX may alleviate myocardial I/R injury through the regulation of the P53/JNK pathway, highlighting H2AX as a potential target for the treatment of myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ziyang Yu
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Yirong Teng
- Department of General Practice The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Hongbo Yang
- Department of Cardiology Fuwai Yunnan Hospital Chinese Academy of Medical Sciences, Kunming, Yunnan, China
| | - Yudi Wang
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Xichen Li
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Lei Feng
- Department of Laboratory Yan'an Hospital of Kunming City, Kunming, Yunnan, China
| | - Wenbo Xu
- Department of Laboratory The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Yinglu Hao
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| | - Yanping Li
- Department of Cardiology The 6th Affiliated Hospital of Kunming Medical University The People's Hospital of Yuxi City, Yuxi, Yunnan, China
| |
Collapse
|
24
|
Campagna R, Mazzanti L, Pompei V, Alia S, Vignini A, Emanuelli M. The Multifaceted Role of Endothelial Sirt1 in Vascular Aging: An Update. Cells 2024; 13:1469. [PMID: 39273039 PMCID: PMC11394039 DOI: 10.3390/cells13171469] [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: 07/17/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
NAD+-dependent deacetylase sirtuin-1 (Sirt1) belongs to the sirtuins family, known to be longevity regulators, and exerts a key role in the prevention of vascular aging. By aging, the expression levels of Sirt1 decline with a severe impact on vascular function, such as the rise of endothelial dysfunction, which in turn promotes the development of cardiovascular diseases. In this context, the impact of Sirt1 activity in preventing endothelial senescence is particularly important. Given the key role of Sirt1 in counteracting endothelial senescence, great efforts have been made to deepen the knowledge about the intricate cross-talks and interactions of Sirt1 with other molecules, in order to set up possible strategies to boost Sirt1 activity to prevent or treat vascular aging. The aim of this review is to provide a proper background on the regulation and function of Sirt1 in the vascular endothelium and to discuss the recent advances regarding the therapeutic strategies of targeting Sirt1 to counteract vascular aging.
Collapse
Affiliation(s)
- Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Laura Mazzanti
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
- Fondazione Salesi, Ospedale G. Salesi, 60100 Ancona, Italy
| | - Veronica Pompei
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Sonila Alia
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Arianna Vignini
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
- Research Center of Health Education and Health Promotion, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| |
Collapse
|
25
|
Ye C, Yan C, Bian SJ, Li XR, Li Y, Wang KX, Zhu YH, Wang L, Wang YC, Wang YY, Li TS, Qi SH, Luo L. Momordica charantia L.-derived exosome-like nanovesicles stabilize p62 expression to ameliorate doxorubicin cardiotoxicity. J Nanobiotechnology 2024; 22:464. [PMID: 39095755 PMCID: PMC11297753 DOI: 10.1186/s12951-024-02705-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Doxorubicin (DOX) is a first-line chemotherapeutic drug for various malignancies that causes cardiotoxicity. Plant-derived exosome-like nanovesicles (P-ELNs) are growing as novel therapeutic agents. Here, we investigated the protective effects in DOX cardiotoxicity of ELNs from Momordica charantia L. (MC-ELNs), a medicinal plant with antioxidant activity. RESULTS We isolated MC-ELNs using ultracentrifugation and characterized them with canonical mammalian extracellular vesicles features. In vivo studies proved that MC-ELNs ameliorated DOX cardiotoxicity with enhanced cardiac function and myocardial structure. In vitro assays revealed that MC-ELNs promoted cell survival, diminished reactive oxygen species, and protected mitochondrial integrity in DOX-treated H9c2 cells. We found that DOX treatment decreased the protein level of p62 through ubiquitin-dependent degradation pathway in H9c2 and NRVM cells. However, MC-ELNs suppressed DOX-induced p62 ubiquitination degradation, and the recovered p62 bound with Keap1 promoting Nrf2 nuclear translocation and the expressions of downstream gene HO-1. Furthermore, both the knockdown of Nrf2 and the inhibition of p62-Keap1 interaction abrogated the cardioprotective effect of MC-ELNs. CONCLUSIONS Our findings demonstrated the therapeutic beneficials of MC-ELNs via increasing p62 protein stability, shedding light on preventive approaches for DOX cardiotoxicity.
Collapse
Affiliation(s)
- Cong Ye
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou city, Jiangsu Province, 221004, PR China
| | - Chen Yan
- Department of Rheumatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang city, Jiangxi Province, PR China
| | - Si-Jia Bian
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou city, Jiangsu Province, 221004, PR China
| | - Xin-Ran Li
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou city, Jiangsu Province, 221004, PR China
| | - Yu Li
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang city, Jiangxi Province, PR China
| | - Kai-Xuan Wang
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou city, Jiangsu Province, PR China
| | - Yu-Hua Zhu
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou city, Jiangsu Province, 221004, PR China
| | - Liang Wang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou city, Jiangsu Province, 221004, PR China
| | - Ying-Chao Wang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou city, Jiangsu Province, 221004, PR China
| | - Yi-Yuan Wang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou city, Jiangsu Province, 221004, PR China
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Su-Hua Qi
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou city, Jiangsu Province, 221004, PR China.
| | - Lan Luo
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou city, Jiangsu Province, 221004, PR China.
| |
Collapse
|
26
|
Quagliariello V, Canale ML, Bisceglia I, Iovine M, Paccone A, Maurea C, Scherillo M, Merola A, Giordano V, Palma G, Luciano A, Bruzzese F, Zito Marino F, Montella M, Franco R, Berretta M, Gabrielli D, Gallucci G, Maurea N. Sodium-glucose cotransporter 2 inhibitor dapagliflozin prevents ejection fraction reduction, reduces myocardial and renal NF-κB expression and systemic pro-inflammatory biomarkers in models of short-term doxorubicin cardiotoxicity. Front Cardiovasc Med 2024; 11:1289663. [PMID: 38818214 PMCID: PMC11138344 DOI: 10.3389/fcvm.2024.1289663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 04/09/2024] [Indexed: 06/01/2024] Open
Abstract
Background Anthracycline-mediated adverse cardiovascular events are among the leading causes of morbidity and mortality in patients with cancer. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) exert multiple cardiometabolic benefits in patients with/without type 2 diabetes, chronic kidney disease, and heart failure with reduced and preserved ejection fraction. We hypothesized that the SGLT2i dapagliflozin administered before and during doxorubicin (DOXO) therapy could prevent cardiac dysfunction and reduce pro-inflammatory pathways in preclinical models. Methods Cardiomyocytes were exposed to DOXO alone or combined with dapagliflozin (DAPA) at 10 and 100 nM for 24 h; cell viability, iATP, and Ca++ were quantified; lipid peroxidation products (malondialdehyde and 4-hydroxy 2-hexenal), NLRP3, MyD88, and cytokines were also analyzed through selective colorimetric and enzyme-linked immunosorbent assay (ELISA) methods. Female C57Bl/6 mice were treated for 10 days with a saline solution or DOXO (2.17 mg/kg), DAPA (10 mg/kg), or DOXO combined with DAPA. Systemic levels of ferroptosis-related biomarkers, galectin-3, high-sensitivity C-reactive protein (hs-CRP), and pro-inflammatory chemokines (IL-1α, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-12, IL17-α, IL-18, IFN-γ, TNF-α, G-CSF, and GM-CSF) were quantified. After treatments, immunohistochemical staining of myocardial and renal p65/NF-kB was performed. Results DAPA exerts cytoprotective, antioxidant, and anti-inflammatory properties in human cardiomyocytes exposed to DOXO by reducing iATP and iCa++ levels, lipid peroxidation, NLRP-3, and MyD88 expression. Pro-inflammatory intracellular cytokines were also reduced. In preclinical models, DAPA prevented the reduction of radial and longitudinal strain and ejection fraction after 10 days of treatment with DOXO. A reduced myocardial expression of NLRP-3 and MyD-88 was seen in the DOXO-DAPA group compared to DOXO mice. Systemic levels of IL-1β, IL-6, TNF-α, G-CSF, and GM-CSF were significantly reduced after treatment with DAPA. Serum levels of galectine-3 and hs-CRP were strongly enhanced in the DOXO group; on the other hand, their expression was reduced in the DAPA-DOXO group. Troponin-T, B-type natriuretic peptide (BNP), and N-Terminal Pro-BNP (NT-pro-BNP) were strongly reduced in the DOXO-DAPA group, revealing cardioprotective properties of SGLT2i. Mice treated with DOXO and DAPA exhibited reduced myocardial and renal NF-kB expression. Conclusion The overall picture of the study encourages the use of DAPA in the primary prevention of cardiomyopathies induced by anthracyclines in patients with cancer.
Collapse
Affiliation(s)
- V. Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - M. L. Canale
- Cardiology Division, Azienda USL Toscana Nord-Ovest, Versilia Hospital, Lido di Camaiore, Italy
| | - I. Bisceglia
- Integrated Cardiology Services, Department of Cardio-Thoracic-Vascular, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| | - M. Iovine
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - A. Paccone
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - C. Maurea
- ASL NA1, UOC Neurology and Stroke Unit, Ospedale del Mare, Naples, Italy
| | - M. Scherillo
- Cardiology Department, San Pio Hospital, Benevento, Italy
| | - A. Merola
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - V. Giordano
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| | - G. Palma
- SSD Sperimentazione Animale, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italy
| | - A. Luciano
- SSD Sperimentazione Animale, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italy
| | - F. Bruzzese
- SSD Sperimentazione Animale, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italy
| | - F. Zito Marino
- Pathology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - M. Montella
- Pathology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - R. Franco
- Pathology Unit, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - M. Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - D. Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlani-ni, Roma—Fondazione per il Tuo Cuore—Heart Care Foundation, Firenze, Italy
| | - G. Gallucci
- Cardio-Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - N. Maurea
- Division of Cardiology, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, Napoli, Italia
| |
Collapse
|
27
|
Wu H, Wang L, Kang P, Zhou X, Li W, Xia Z. The SP1/SIRT1/ACLY signaling axis mediates fatty acid oxidation in renal ischemia-reperfusion-induced renal fibrosis. Int Immunopharmacol 2024; 132:112002. [PMID: 38608473 DOI: 10.1016/j.intimp.2024.112002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Renal ischemia-reperfusion is the primary cause of acute kidney injury (AKI). Clinically, most patients who experience ischemia-reperfusion injury eventually progress gradually to renal fibrosis and chronic kidney disease (CKD). However, the underlying mechanism for AKI to CKD transition remain absent. Our study demonstrated that the downregulation of sirtuin 1 (Sirt1)-mediated fatty acid oxidation (FAO) facilitates IRI-induced renal fibrosis. METHODS The IRI animal model was established, and ribonucleic acid (RNA) sequencing was used to explore potential differentially expressed genes (DEGs) and pathways. The SIRT1 knockout mice were generated, and a recombinant adeno-associated virus that overexpresses SIRT1 was injected into mice to explore the function of SIRT1 in renal fibrosis induced by renal IRI. In vitro, hypoxia/reoxygenation (H/R) was used to establish the classical model of renal IRI and overexpression or knockdown of SIRT1 to investigate the SIRT1 function through lentiviral plasmids. The underlying molecular mechanism was explored through RNA sequencing, bioinformatics analysis, and chromatin immunoprecipitation assay. RESULTS RNA sequencing analysis and western blot demonstrated that the expression of SIRT1 was significantly decreased in IRI mice. Overexpression of SIRT1 improved renal function and reduced lipid deposition and renal fibrosis. On the contrary, knockout of SIRT1 aggravated kidney injury and renal fibrosis. RNA sequencing, bioinformatics analysis, and chromatin immunoprecipitation assay mechanistically revealed that SIRT1 impairs the acetylation of histone H3K27 on the promoter region of ACLY, thereby impeding FAO activity and promoting renal fibrosis. Additionally, SP1 regulated FAO by directly modulating SIRT1 expression. CONCLUSION Our findings highlight that downregulation of SIRT1-modulated FAO facilitated by the SP1/SIRT1/ACLY axis in the kidney increases IRI, suggesting SIRT1 to be a potential therapeutic target for renal fibrosis induced by renal IRI.
Collapse
Affiliation(s)
- Huailiang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Liyan Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Peng Kang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
28
|
Fanelli G, Alloisio G, Lelli V, Marini S, Rinalducci S, Gioia M. Mechano-induced cell metabolism disrupts the oxidative stress homeostasis of SAOS-2 osteosarcoma cells. Front Mol Biosci 2024; 10:1297826. [PMID: 38726050 PMCID: PMC11079223 DOI: 10.3389/fmolb.2023.1297826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/18/2023] [Indexed: 05/12/2024] Open
Abstract
There has been an increasing focus on cancer mechanobiology, determining the underlying-induced changes to unlock new avenues in the modulation of cell malignancy. Our study used LC-MS untargeted metabolomic approaches and real-time polymerase chain reaction (PCR) to characterize the molecular changes induced by a specific moderate uniaxial stretch regimen (i.e., 24 h-1 Hz, cyclic stretch 0,5% elongation) on SAOS-2 osteosarcoma cells. Differential metabolic pathway analysis revealed that the mechanical stimulation induces a downregulation of both glycolysis and the tricarboxylic acid (TCA) cycle. At the same time, the amino acid metabolism was found to be dysregulated, with the mechanical stimulation enhancing glutaminolysis and reducing the methionine cycle. Our findings showed that cell metabolism and oxidative defense are tightly intertwined in mechanically stimulated cells. On the one hand, the mechano-induced disruption of the energy cell metabolism was found correlated with an antioxidant glutathione (GSH) depletion and an accumulation of reactive oxygen species (ROS). On the other hand, we showed that a moderate stretch regimen could disrupt the cytoprotective gene transcription by altering the expression levels of manganese superoxide dismutase (SOD1), Sirtuin 1 (SIRT1), and NF-E2-related factor 2 (Nrf2) genes. Interestingly, the cyclic applied strain could induce a cytotoxic sensitization (to the doxorubicin-induced cell death), suggesting that mechanical signals are integral regulators of cell cytoprotection. Hence, focusing on the mechanosensitive system as a therapeutic approach could potentially result in more effective treatments for osteosarcoma in the future.
Collapse
Affiliation(s)
- Giuseppina Fanelli
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Viterbo, Italy
| | - Giulia Alloisio
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Veronica Lelli
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Viterbo, Italy
| | - Stefano Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Sara Rinalducci
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Viterbo, Italy
| | - Magda Gioia
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|
29
|
Manawy SM, Faruk EM, Hindawy RF, Hassan MM, Farrag DMG, Bashar MAE, Fouad H, Bagabir RA, Hassan DAA, Zaazaa AM, Hablas MGA, Kamal KM. Modulation of the Sirtuin-1 signaling pathway in doxorubicin-induced nephrotoxicity (synergistic amelioration by resveratrol and pirfenidone). Tissue Cell 2024; 87:102330. [PMID: 38412579 DOI: 10.1016/j.tice.2024.102330] [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: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/29/2024]
Abstract
The current study was conducted to determine the precise mechanisms of Sirtuin-1 (Sirt-1), TGF- β (Transforming Growth Factor-β), and long non-coding RNA Metastasis Associated Lung Adenocarcinoma Transcript 1 (LncRNA MALAT-1) in signaling pathways in doxorubicin (DOX)-induced nephrotoxicity. The potential therapeutic effect of Resveratrol and Pirfenidone in DOX toxicity was also assessed. Thirty-six male adult rats were evenly distributed into four groups: Group 1: control rats. Group 2: DOX exposed rats' group, each animal received 7.5 mg/kg DOX as a single intravenous dose, Group 3: DOX exposed group subjected to oral resveratrol (20 mg/kg/daily for two weeks), Group 4: DOX exposed group subjected to oral Pirfenidone (200 mg/kg once daily for 10 days). At the planned time, animals were sacrificed. Renal tissue was collected to assess matrix metalloproteinase-9 (MMP9), inflammatory and apoptotic markers: tumor necrosis factor-alpha (TNF- β, caspase-3, cyclo-oxygenase-2 (COX-2), and oxidative stress markers: nitric oxide (NO), Glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD). Sirtuin-1 (Sirt-1), TGF-β, and LncRNA MALAT-1 were quantitatively assessed by real-time RT-PCR in the whole blood. Results showed that the DOX group exhibited a significant increase in oxidative stress markers, and inflammatory, and apoptotic markers in the renal tissue. Histologically, the renal tubule lining cells exhibited vacuolar alterations in the cytoplasm, glomerular atrophy, and vascular congestion. Furthermore, renal degeneration was evident, as confirmed by the heightened immuno-expression of MMP9. Exposure to DOX resulted in a significant decrease in Sirtuin-1 (Sirt-1) with a significant increase in the TGFβ, and LncRNA MALAT-1 gene expression. However, pre-treatment with either resveratrol/or Pirefenidone ameliorated the histological renal alterations, regulated the pathways of Sirt-1, TGFβ, and LncRNA MALAT-1, and decreased all oxidative stress, inflammatory and apoptotic markers. In conclusion, DOX exposure leads to renal toxicity by inducing renal degeneration, oxidative stress, and apoptosis. Administration of either resveratrol or Pirfenidone counteracted these changes and protected the kidney against DOX-induced renal damage.
Collapse
Affiliation(s)
- Samia Mahmoud Manawy
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Benha, Egypt.
| | - Eman Mohamed Faruk
- Anatomy Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia; Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, Egypt.
| | - Rabab Fawzy Hindawy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha, Egypt.
| | - Mahmoud M Hassan
- Department of Physiology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Diaa M G Farrag
- Marine Biology Branch, Zoology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt.
| | - Mansour A E Bashar
- Marine Biology Branch, Zoology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo 11884, Egypt.
| | - Hanan Fouad
- Basic Medical Sciences, Faculty of Medicine, Galala University, Galala City, POB 43711, ATTAKA, Suez Governorate, Egypt; Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo POB 12613, Egypt.
| | - Rania Abubaker Bagabir
- College of Medicine, Hematology and Immunology Department, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Ahmed Mohammed Zaazaa
- Students at Faculty of Medicine, Benha National University, Benha Colleges in Cairo, Egypt
| | | | - K Mostafa Kamal
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
30
|
Zhang W, Shi C, Yao Z, Qian S. Bardoxolone methyl attenuates doxorubicin-induced cardiotoxicity by inhibiting the TXNIP-NLRP3 pathway through Nrf2 activation. ENVIRONMENTAL TOXICOLOGY 2024; 39:1936-1950. [PMID: 38064254 DOI: 10.1002/tox.24075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 03/09/2024]
Abstract
Bardoxolone methyl, which triggers nuclear factor erythroid 2-related factor (Nrf2), has therapeutic effects against myocardial infarction, heart failure, and other diseases. Nrf2 can inhibit the activation of the thioredoxin-interacting protein (TXNIP)/NLR family pyrin domain-containing protein 3 (NLRP3) pathway. Doxorubicin is an anthracycline chemotherapeutic drug associated with cardiotoxicity, limiting its clinical use. In this study, we explored the specific mechanism of the Nrf2-TXNIP-NLRP3 pathway in doxorubicin-induced cardiotoxicity using bardoxolone methyl in animal and cell models. Using in vivo and in vitro experiments, we show that doxorubicin can induce oxidative stress and pyroptosis in the heart. Western blot and co-immunoprecipitation experimental results found that doxorubicin can reduce the interaction between TXNIP and TRX, increase the interaction between TXNIP and NLRP3, and activate the pyroptosis process. Bardoxolone methyl reduces the accumulation of reactive oxygen species in cardiomyocytes through the Nrf2 pathway, inhibits the interaction between TXNIP and NLRP3, and alleviates the progression of myocardial damage and cardiac fibrosis. Bardoxolone methyl lost its therapeutic effect when the expression of Nrf2 was decreased. Additionally, repressing the expression of TXNIP can inhibit the activation of NLRP3 and alleviate myocardial damage caused by doxorubicin. Collectively, our findings confirm that bardoxolone methyl alleviates doxorubicin-induced cardiotoxicity by activating Nrf2 and inhibiting the TXNIP-NLRP3 pathway.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Chao Shi
- Department of Cardiac Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Zhuoya Yao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Shaohuan Qian
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
31
|
Shen J, Lan Y, Ji Z, Liu H. Sirtuins in intervertebral disc degeneration: current understanding. Mol Med 2024; 30:44. [PMID: 38553713 PMCID: PMC10981339 DOI: 10.1186/s10020-024-00811-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/20/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is one of the etiologic factors of degenerative spinal diseases, which can lead to a variety of pathological spinal conditions such as disc herniation, spinal stenosis, and scoliosis. IVDD is a leading cause of lower back pain, the prevalence of which increases with age. Recently, Sirtuins/SIRTs and their related activators have received attention for their activity in the treatment of IVDD. In this paper, a comprehensive systematic review of the literature on the role of SIRTs and their activators on IVDD in recent years is presented. The molecular pathways involved in the regulation of IVDD by SIRTs are summarized, and the effects of SIRTs on senescence, inflammatory responses, oxidative stress, and mitochondrial dysfunction in myeloid cells are discussed with a view to suggesting possible solutions for the current treatment of IVDD. PURPOSE This paper focuses on the molecular mechanisms by which SIRTs and their activators act on IVDD. METHODS A literature search was conducted in Pubmed and Web of Science databases over a 13-year period from 2011 to 2024 for the terms "SIRT", "Sirtuin", "IVDD", "IDD", "IVD", "NP", "Intervertebral disc degeneration", "Intervertebral disc" and "Nucleus pulposus". RESULTS According to the results, SIRTs and a large number of activators showed positive effects against IVDD.SIRTs modulate autophagy, myeloid apoptosis, oxidative stress and extracellular matrix degradation. In addition, they attenuate inflammatory factor-induced disc damage and maintain homeostasis during disc degeneration. Several clinical studies have reported the protective effects of some SIRTs activators (e.g., resveratrol, melatonin, honokiol, and 1,4-dihydropyridine) against IVDD. CONCLUSION The fact that SIRTs and their activators play a hundred different roles in IVDD helps to better understand their potential to develop further treatments for IVDD. NOVELTY This review summarizes current information on the mechanisms of action of SIRTs in IVDD and the challenges and limitations of translating their basic research into therapy.
Collapse
Affiliation(s)
- Jianlin Shen
- Department of Orthopaedics, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China
- Central Laboratory, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China
| | - Yujian Lan
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ziyu Ji
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Huan Liu
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- The Third People's Hospital of Longmatan District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
32
|
Jin Q, Ma F, Liu T, Yang L, Mao H, Wang Y, Peng L, Li P, Zhan Y. Sirtuins in kidney diseases: potential mechanism and therapeutic targets. Cell Commun Signal 2024; 22:114. [PMID: 38347622 PMCID: PMC10860260 DOI: 10.1186/s12964-023-01442-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/12/2023] [Indexed: 02/15/2024] Open
Abstract
Sirtuins, which are NAD+-dependent class III histone deacetylases, are involved in various biological processes, including DNA damage repair, immune inflammation, oxidative stress, mitochondrial homeostasis, autophagy, and apoptosis. Sirtuins are essential regulators of cellular function and organismal health. Increasing evidence suggests that the development of age-related diseases, including kidney diseases, is associated with aberrant expression of sirtuins, and that regulation of sirtuins expression and activity can effectively improve kidney function and delay the progression of kidney disease. In this review, we summarise current studies highlighting the role of sirtuins in renal diseases. First, we discuss sirtuin family members and their main mechanisms of action. We then outline the possible roles of sirtuins in various cell types in kidney diseases. Finally, we summarise the compounds that activate or inhibit sirtuin activity and that consequently ameliorate renal diseases. In conclusion, targeted modulation of sirtuins is a potential therapeutic strategy for kidney diseases. Video Abstract.
Collapse
Affiliation(s)
- Qi Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
33
|
Shen S, Shen M, Kuang L, Yang K, Wu S, Liu X, Wang Y, Wang Y. SIRT1/SREBPs-mediated regulation of lipid metabolism. Pharmacol Res 2024; 199:107037. [PMID: 38070792 DOI: 10.1016/j.phrs.2023.107037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024]
Abstract
Sirtuins, also called silent information regulator 2, are enzymes that rely on nicotinamide adenine dinucleotide (NAD+) to function as histone deacetylases. Further investigation is warranted to explore the advantageous impacts of Sirtuin 1 (SIRT1), a constituent of the sirtuin group, on lipid metabolism, in addition to its well-researched involvement in extending lifespan. The regulation of gene expression has been extensively linked to SIRT1. Sterol regulatory element-binding protein (SREBP) is a substrate of SIRT1 that has attracted significant interest due to its role in multiple cellular processes including cell cycle regulation, DNA damage repair, and metabolic functions. Hence, the objective of this analysis was to investigate and elucidate the correlation between SIRT1 and SREBPs, as well as assess the contribution of SIRT1/SREBPs in mitigating lipid metabolism dysfunction. The objective of this research was to investigate whether SIRT1 and SREBPs could be utilized as viable targets for therapeutic intervention in managing complications associated with diabetes.
Collapse
Affiliation(s)
- Shan Shen
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Mingyang Shen
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Lirun Kuang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Keyu Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Shiran Wu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xinde Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yuting Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yong Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
34
|
Liao Y, Meng Q. Protection against cancer therapy-induced cardiovascular injury by planed-derived polyphenols and nanomaterials. ENVIRONMENTAL RESEARCH 2023; 238:116896. [PMID: 37586453 DOI: 10.1016/j.envres.2023.116896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/18/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Cancer therapy-induced heart injury is a significant concern for cancer patients undergoing chemotherapy, radiotherapy, immunotherapy, and also targeted molecular therapy. The use of these treatments can lead to oxidative stress and cardiomyocyte damage in the heart, which can result in heart failure and other cardiac complications. Experimental studies have revealed that chemotherapy drugs such as doxorubicin and cyclophosphamide can cause severe side effects such as cardiac fibrosis, electrophysiological remodeling, chronic oxidative stress and inflammation, etc., which may increase risk of cardiac disorders and attacks for patients that underwent chemotherapy. Similar consequences may also be observed for patients that undergo radiotherapy for left breast or lung malignancies. Polyphenols, a group of natural compounds with antioxidant and anti-inflammatory properties, have shown the potential in protecting against cancer therapy-induced heart injury. These compounds have been found to reduce oxidative stress, necrosis and apoptosis in the heart, thereby preserving cardiac function. In recent years, nanoparticles loaded with polyphenols have also provided for the delivery of these compounds and increasing their efficacy in different organs. These nanoparticles can improve the bioavailability and efficacy of polyphenols while minimizing their toxicity. This review article summarizes the current understanding of the protective effects of polyphenols and nanoparticles loaded with polyphenols against cancer therapy-induced heart injury. The article discusses the mechanisms by which polyphenols protect the heart, including antioxidant and anti-inflammation abilities. The article also highlights the potential benefits of using nanoparticles for the delivery of polyphenols.
Collapse
Affiliation(s)
- Yunshu Liao
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China
| | - Qinghua Meng
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
35
|
Lin X, Wu G, Wang S, Huang J. Bibliometric and visual analysis of doxorubicin-induced cardiotoxicity. Front Pharmacol 2023; 14:1255158. [PMID: 38026961 PMCID: PMC10665513 DOI: 10.3389/fphar.2023.1255158] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Doxorubicin-induced cardiotoxicity represents a prevalent adverse effect encountered in patients undergoing treatment with doxorubicin. To date, there has been no bibliometric study to summarize the field of doxorubicin-induced cardiotoxicity. In our study, we aim to determine the current status and frontiers of doxorubicin-induced cardiotoxicity by bibliometric analysis. Methods: The documents concerning doxorubicin-induced cardiotoxicity are obtained from the Web of Science Core Collection database (WOSCC), and VOSviewer 1.6.16, CiteSpace 5.1.3 and the WOSCC's literature analysis wire were used to conduct the bibliometric analysis. Results: In total, 7,021 publications were encompassed, which are produced by 37,152 authors and 6,659 organizations, 1,323 journals, and 101 countries/regions. The most productive author, institution, country and journal were Bonnie Ky with 35 publications, University of Texas with 190 documents, the United States with 1,912 publications, and PLOS ONE with 120 documents. The first high-cited article was published in the NEJM with 8,134 citations authored by DJ Slamon et al., in 2001. For keyword analysis, there are four clusters depicted in distinct directions. The keywords in the red cluster are oxidative stress, apoptosis, and cardiomyopathy. The keywords in the green cluster are cardiotoxicity, heart failure, and anthracycline. The keywords in the blue cluster are chemotherapy, trastuzumab, and paclitaxel. The keywords in the purple cluster are doxorubicin, adriamycin, and cancer. Most of the documents were derived from the United States, China and Italy (4,080/7,021, 58.1%). The number of studies from other countries should be increased. Conclusion: In conclusion, the main research hotspots and frontiers in the field of doxorubicin-induced cardiotoxicity include the role of doxorubicin in cardiotoxicity, the mechanisms underlying doxorubicin-induced cardiotoxicity, and the development of treatment strategies for doxorubicin-induced cardiotoxicity. More studies are needed to explore the mechanisms and treatment of doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | - Shuai Wang
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinyu Huang
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
36
|
Han J, Wang S, Wang H, Zhang T, Yang Y, Zhao T, Chen Z, Xia G, Wang C. SIRT1 reduction contributes to doxorubicin-induced oxidative stress and meiotic failure in mouse oocytes. Toxicol Appl Pharmacol 2023; 476:116671. [PMID: 37633598 DOI: 10.1016/j.taap.2023.116671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Impaired fertility is the major side effect of chemotherapy for female cancer patients, accumulated evidence indicates this is associated with damage on oocyte quality, but the underlying mechanisms remain unclear. Previously we reported that doxorubicin (DXR) exposure, one of the most widely used chemotherapy drugs, disrupted mouse oocyte meiotic maturation in vitro. In the current study, we identified that SIRT1 expression was remarkably reduced in DXR exposure oocytes. Next, we found that increasing SIRT1 expression by resveratrol partially alleviated the effects of DXR exposure on oocyte maturation, which was counteracted by SIRT1 inhibition. Furthermore, we revealed that increasing SIRT1 expression mitigated DXR induced oocyte damage through reducing ROS levels, increasing antioxidant enzyme MnSOD expression, and preventing spindle and chromosome disorganization, lowering the incidence of aneuploidy. Importantly, by performing in vitro fertilization and embryo transfer assays, we demonstrated that increasing SIRT1 expression significantly improved the fertilization ability, developmental competence of oocytes and early embryos. In summary, our data uncover that SIRT1 reduction represents one mechanism that mediates the effects of DXR exposure on oocyte quality.
Collapse
Affiliation(s)
- Jun Han
- Jiangsu Academy of Agricultural Sciences, Nanjing 21000, China; State Key Laboratory of Livestock and Poultry Biotechnology Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shuo Wang
- State Key Laboratory of Livestock and Poultry Biotechnology Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Huarong Wang
- State Key Laboratory of Livestock and Poultry Biotechnology Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Tuo Zhang
- State Key Laboratory of Livestock and Poultry Biotechnology Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Ye Yang
- State Key Laboratory of Livestock and Poultry Biotechnology Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Ting Zhao
- State Key Laboratory of Livestock and Poultry Biotechnology Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Ziqi Chen
- State Key Laboratory of Livestock and Poultry Biotechnology Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Guoliang Xia
- State Key Laboratory of Livestock and Poultry Biotechnology Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China; Ningxia University, Ningxia 750021, China
| | - Chao Wang
- State Key Laboratory of Livestock and Poultry Biotechnology Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
37
|
Li X. Doxorubicin-mediated cardiac dysfunction: Revisiting molecular interactions, pharmacological compounds and (nano)theranostic platforms. ENVIRONMENTAL RESEARCH 2023; 234:116504. [PMID: 37356521 DOI: 10.1016/j.envres.2023.116504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023]
Abstract
Although chemotherapy drugs are extensively utilized in cancer therapy, their administration for treatment of patients has faced problems that regardless of chemoresistance, increasing evidence has shown concentration-related toxicity of drugs. Doxorubicin (DOX) is a drug used in treatment of solid and hematological tumors, and its function is based on topoisomerase suppression to impair cancer progression. However, DOX can also affect the other organs of body and after chemotherapy, life quality of cancer patients decreases due to the side effects. Heart is one of the vital organs of body that is significantly affected by DOX during cancer chemotherapy, and this can lead to cardiac dysfunction and predispose to development of cardiovascular diseases and atherosclerosis, among others. The exposure to DOX can stimulate apoptosis and sometimes, pro-survival autophagy stimulation can ameliorate this condition. Moreover, DOX-mediated ferroptosis impairs proper function of heart and by increasing oxidative stress and inflammation, DOX causes cardiac dysfunction. The function of DOX in mediating cardiac toxicity is mediated by several pathways that some of them demonstrate protective function including Nrf2. Therefore, if expression level of such protective mechanisms increases, they can alleviate DOX-mediated cardiac toxicity. For this purpose, pharmacological compounds and therapeutic drugs in preventing DOX-mediated cardiotoxicity have been utilized and they can reduce side effects of DOX to prevent development of cardiovascular diseases in patients underwent chemotherapy. Furthermore, (nano)platforms are used comprehensively in treatment of cardiovascular diseases and using them for DOX delivery can reduce side effects by decreasing concentration of drug. Moreover, when DOX is loaded on nanoparticles, it is delivered into cells in a targeted way and its accumulation in healthy organs is prevented to diminish its adverse impacts. Hence, current paper provides a comprehensive discussion of DOX-mediated toxicity and subsequent alleviation by drugs and nanotherapeutics in treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai, 200072, China.
| |
Collapse
|
38
|
Basak M, Das K, Mahata T, Kumar D, Nagar N, Poluri KM, Kumar P, Das P, Stewart A, Maity B. RGS7 balances acetylation/de-acetylation of p65 to control chemotherapy-dependent cardiac inflammation. Cell Mol Life Sci 2023; 80:255. [PMID: 37589751 PMCID: PMC11071981 DOI: 10.1007/s00018-023-04895-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/28/2023] [Accepted: 07/22/2023] [Indexed: 08/18/2023]
Abstract
Cardiotoxicity remains a major limitation in the clinical utility of anthracycline chemotherapeutics. Regulator of G-protein Signaling 7 (RGS7) and inflammatory markers are up-regulated in the hearts of patients with a history of chemotherapy particularly those with reduced left-ventricular function. RGS7 knockdown in either the murine myocardium or isolated murine ventricular cardiac myocytes (VCM) or cultured human VCM provided marked protection against doxorubicin-dependent oxidative stress, NF-κB activation, inflammatory cytokine production, and cell death. In exploring possible mechanisms causally linking RGS7 to pro-inflammatory signaling cascades, we found that RGS7 forms a complex with acetylase Tip60 and deacetylase sirtuin 1 (SIRT1) and controls the acetylation status of the p65 subunit of NF-κB. In VCM, the detrimental impact of RGS7 could be mitigated by inhibiting Tip60 or activating SIRT1, indicating that the ability of RGS7 to modulate cellular acetylation capacity is critical for its pro-inflammatory actions. Further, RGS7-driven, Tip60/SIRT1-dependent cytokines released from ventricular cardiac myocytes and transplanted onto cardiac fibroblasts increased oxidative stress, markers of transdifferentiation, and activity of extracellular matrix remodelers emphasizing the importance of the RGS7-Tip60-SIRT1 complex in paracrine signaling in the myocardium. Importantly, while RGS7 overexpression in heart resulted in sterile inflammation, fibrotic remodeling, and compromised left-ventricular function, activation of SIRT1 counteracted the detrimental impact of RGS7 in heart confirming that RGS7 increases acetylation of SIRT1 substrates and thereby drives cardiac dysfunction. Together, our data identify RGS7 as an amplifier of inflammatory signaling in heart and possible therapeutic target in chemotherapeutic drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Madhuri Basak
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Kiran Das
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Tarun Mahata
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Dinesh Kumar
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Nupur Nagar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Pranesh Kumar
- Institute of Pharmaceutical Sciences, University of Lucknow, Lucknow, Uttar Pradesh, 226025, India
| | - Priyadip Das
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamilnadu, 603203, India
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA.
| | - Biswanath Maity
- Centre of Biomedical Research (CBMR), SGPGI, SGPGI Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India.
| |
Collapse
|
39
|
Peng B, Rao L, Yang J, Ku X, Kong B, Shuai W, Huang H. Columbianadin attenuates doxorubicin-induced cardiac injury, oxidative stress, and apoptosis via Sirt1/FOXO1 signaling pathway. Acta Cir Bras 2023; 38:e382223. [PMID: 37377248 DOI: 10.1590/acb382223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/11/2023] [Indexed: 06/29/2023] Open
Abstract
PURPOSE Oxidative stress and apoptosis contribute to the pathological basis of doxorubicin (DOX)-induced cardiotoxicity. Columbianadin (CBN) is one of the main bioactive constituents isolated from the root of Angelica pubescens. Herein, we intended to explore the potential role and molecular basis of CBN in DOX-induced cardiotoxicity. METHODS C57BL/6 mice were subjected to DOX (15 mg/kg/day, i.p.) to generate DOX-induced cardiotoxicity. CBN (10 mg/kg/day, i.p.) was administered for four week following DOX injection. RESULTS DOX administered markedly dampened cardiac function, increased cardiac injury, excessive reactive oxygen species (ROS) production, and cardiomyocyte loss. These alterations induced by DOX significantly alleviated by CBN treatment. Mechanistically, our results demonstrated that the CBN exerts cardioprotection role against DOX by up-regulating silent information regulator 1 (Sirt1) and decreasing acetylation of forkhead box O1 (FOXO1). Moreover, Sirt1 inhibition with Ex-527 significantly blunt the beneficial effect of CBN on DOX-induced cardiotoxicity, including cardiac dysfunction, ROS, and apoptosis. CONCLUSION Collectively, CBN attenuated oxidative stress and cardiomyocyte apoptosis in DOX-induced cardiotoxicity through maintaining Sirt1/FOXO1 signaling pathway. Our results demonstrated that CBN might be used to treat DOX-related cardiotoxicity.
Collapse
Affiliation(s)
- Bo Peng
- Wuhan University - Renmin Hospital - Department of Cardiology - Hubei, China
- Wuhan University - Cardiovascular Research Institute - Hubei, China
- Hubei Key Laboratory of Cardiology - Hubei, China
| | - Li Rao
- Wuhan University - Renmin Hospital - Department of Geriatrics - Hubei, China
| | - Jiaolong Yang
- Wuhan University - Renmin Hospital - Department of Neurology - Hubei, China
| | - Xiaowei Ku
- Wuhan University - Renmin Hospital - Department of Endocrinology - Hubei, China
| | - Bin Kong
- Wuhan University - Renmin Hospital - Department of Cardiology - Hubei, China
- Wuhan University - Cardiovascular Research Institute - Hubei, China
- Hubei Key Laboratory of Cardiology - Hubei, China
| | - Wei Shuai
- Wuhan University - Renmin Hospital - Department of Cardiology - Hubei, China
| | - He Huang
- Wuhan University - Renmin Hospital - Department of Cardiology - Hubei, China
- Wuhan University - Cardiovascular Research Institute - Hubei, China
- Hubei Key Laboratory of Cardiology - Hubei, China
| |
Collapse
|
40
|
Hosoda R, Nakashima R, Yano M, Iwahara N, Asakura S, Nojima I, Saga Y, Kunimoto R, Horio Y, Kuno A. Resveratrol, a SIRT1 activator, attenuates aging-associated alterations in skeletal muscle and heart in mice. J Pharmacol Sci 2023; 152:112-122. [PMID: 37169475 DOI: 10.1016/j.jphs.2023.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 05/13/2023] Open
Abstract
Aging is associated with impairment of multiple organs, including skeletal muscle and heart. In this study, we investigated whether resveratrol, an activator of an NAD+-dependent protein deacetylase Sirtuin-1 (SIRT1), attenuates age-related sarcopenia and cardiomyocyte hypertrophy in mice. Treatment of mice with resveratrol (0.4 g/kg diet) from 28 weeks of age for 32 weeks prevented aging-associated shortening of rotarod riding time. In the tibialis anterior (TA) muscle, histogram analysis showed that the atrophic muscle was increased in 60-week-old (wo) mice compared with 20-wo mice, which was attenuated by resveratrol. In the heart, resveratrol attenuated an aging-associated increase in the cardiomyocyte diameter. Acetylated proteins were increased and autophagic activity was reduced in the TA muscle of 60-wo mice compared with those of 20-wo mice. Resveratrol treatment reduced levels of acetylated proteins and restored autophagic activity in the TA muscle. Aging-related reduction in myocardial autophagy was also suppressed by resveratrol. Skeletal muscle-specific SIRT1 knockout mice showed increases in acetylated proteins and atrophic muscle fibers and reduced autophagic activity in the TA muscle. These results suggest that activation of SIRT1 by treatment with resveratrol suppresses sarcopenia and cardiomyocyte hypertrophy by restoration of autophagy in mice.
Collapse
Affiliation(s)
- Ryusuke Hosoda
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Ryuta Nakashima
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Masaki Yano
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Naotoshi Iwahara
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Seidai Asakura
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Iyori Nojima
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Yukika Saga
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Risa Kunimoto
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Yoshiyuki Horio
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan
| | - Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University School of Medicine, Japan.
| |
Collapse
|
41
|
Korste S, Settelmeier S, Michel L, Odersky A, Stock P, Reyes F, Haj-Yehia E, Anker MS, Grüneboom A, Hendgen-Cotta UB, Rassaf T, Totzeck M. Anthracycline Therapy Modifies Immune Checkpoint Signaling in the Heart. Int J Mol Sci 2023; 24:ijms24076052. [PMID: 37047026 PMCID: PMC10094326 DOI: 10.3390/ijms24076052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
Cancer survival rates have increased significantly because of improvements in therapy regimes and novel immunomodulatory drugs. Recently, combination therapies of anthracyclines and immune checkpoint inhibitors (ICIs) have been proposed to maximize neoplastic cell removal. However, it has been speculated that a priori anthracycline exposure may prone the heart vulnerable to increased toxicity from subsequent ICI therapy, such as an anti-programmed cell death protein 1 (PD1) inhibitor. Here, we used a high-dose anthracycline mouse model to characterize the role of the PD1 immune checkpoint signaling pathway in cardiac tissue using flow cytometry and immunostaining. Anthracycline treatment led to decreased heart function, increased concentration of markers of cell death after six days and a change in heart cell population composition with fewer cardiomyocytes. At the same time point, the number of PD1 ligand (PDL1)-positive immune cells and endothelial cells in the heart decreased significantly. The results suggest that PD1/PDL1 signaling is affected after anthracycline treatment, which may contribute to an increased susceptibility to immune-related adverse events of subsequent anti-PD1/PDL1 cancer therapy.
Collapse
Affiliation(s)
- Sebastian Korste
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Stephan Settelmeier
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Andrea Odersky
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Pia Stock
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Fabrice Reyes
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Elias Haj-Yehia
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Markus S Anker
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, 12203 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
| | - Anika Grüneboom
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany
| | - Ulrike B Hendgen-Cotta
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, 45147 Essen, Germany
| |
Collapse
|
42
|
Chen Y, Shi S, Dai Y. Research progress of therapeutic drugs for doxorubicin-induced cardiomyopathy. Biomed Pharmacother 2022; 156:113903. [PMID: 36279722 DOI: 10.1016/j.biopha.2022.113903] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 12/06/2022] Open
Abstract
Doxorubicin (DOX), as a kind of chemotherapy agent with remarkable therapeutic effect, can be used to treat diverse malignant tumors clinically. Dose-dependent cardiotoxicity is the most serious adverse reaction after DOX treatment, which eventually leads to cardiomyopathy and greatly limits the clinical application of DOX. DOX-induced cardiomyopathy is not a result of a single mechanistic action, and multiple mechanisms have been discovered and demonstrated experimentally, such as oxidative stress, inflammation, mitochondrial damage, calcium homeostasis disorder, ferroptosis, autophagy and apoptosis. Dexrazoxane (DEX) is the only protective agent approved by FDA for the treatment of DOX cardiomyopathy, but its clinical treatment still has some limitations. Therefore, we need to find other effective therapeutic drugs as soon as possible. In this paper, the drugs that effectively improve cardiomyopathy in recent years are mainly described from the aspects of natural drugs, endogenous substances, new dosage forms, herbal medicines, chemical modification and marketed drugs. The aim of the present study is to evaluate the effects of these drugs on DOX-induced anticancer and cardiomyopathy curative effects, so as to provide some reference value for clinical treatment of DOX-induced cardiomyopathy in the future.
Collapse
Affiliation(s)
- Ye Chen
- Department of pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China; School of pharmacy, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Saixian Shi
- Department of pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China; School of pharmacy, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Yan Dai
- Department of pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province 646000, China.
| |
Collapse
|
43
|
Wu L, Sowers JR, Zhang Y, Ren J. OUP accepted manuscript. Cardiovasc Res 2022; 119:691-709. [PMID: 35576480 DOI: 10.1093/cvr/cvac080] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular diseases (CVDs) arise from a complex interplay among genomic, proteomic, and metabolomic abnormalities. Emerging evidence has recently consolidated the presence of robust DNA damage in a variety of cardiovascular disorders. DNA damage triggers a series of cellular responses termed DNA damage response (DDR) including detection of DNA lesions, cell cycle arrest, DNA repair, cellular senescence, and apoptosis, in all organ systems including hearts and vasculature. Although transient DDR in response to temporary DNA damage can be beneficial for cardiovascular function, persistent activation of DDR promotes the onset and development of CVDs. Moreover, therapeutic interventions that target DNA damage and DDR have the potential to attenuate cardiovascular dysfunction and improve disease outcome. In this review, we will discuss molecular mechanisms of DNA damage and repair in the onset and development of CVDs, and explore how DDR in specific cardiac cell types contributes to CVDs. Moreover, we will highlight the latest advances regarding the potential therapeutic strategies targeting DNA damage signalling in CVDs.
Collapse
Affiliation(s)
- Lin Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri Columbia, Columbia, MO 65212, USA
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|