1
|
Tan Y, Ouyang Y, Xiao L, Huang J, Li F, Ma Z, Tan C, Feng W, Davis E, Tang Y, Chang X, Li H. Lipopolysaccharide-induced DNA damage response activates DNA-PKcs to drive actin cytoskeleton disruption and cardiac microvascular dysfunction in endotoxemia. Theranostics 2025; 15:5969-5997. [PMID: 40365284 PMCID: PMC12068286 DOI: 10.7150/thno.111266] [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: 01/29/2025] [Accepted: 04/05/2025] [Indexed: 05/15/2025] Open
Abstract
Rationale: Sepsis-induced cardiomyopathy is characterized by microvascular injury, which is linked to lipopolysaccharide (LPS)-induced DNA damage response (DDR). This study investigates the role of DNA-PKcs, a key enzyme in the DDR pathway, in driving actin disruption and microvascular dysfunction following LPS exposure. Methods: We analyzed diverse transcriptomic datasets from septic human and murine models using bioinformatics tools to assess DDR pathway activation, correlations, and prognosis. In vivo, LPS-challenged mice were treated with inhibitors of DNA-PKcs or mitochondrial fission, and we evaluated cardiac function, microvascular integrity, mitochondrial status, and actin polymerization. Results: Bioinformatic analyses consistently revealed significant activation of the DDR pathway and upregulation of key genes across diverse septic models. Notably, elevated DDR pathway activity was significantly correlated with poor 28-day survival in human sepsis patients. Single-cell analysis localized this DDR gene upregulation predominantly to cardiac endothelial cells (ECs), fibroblasts, and macrophages during sepsis. Within septic capillary ECs, DDR pathway activity scores strongly correlated spatially and functionally with heightened mitochondrial fission and cytoskeletal remodeling pathway activities. In vivo experiments confirmed that LPS induced severe systolic and diastolic dysfunction, microvascular damage, and mitochondrial fragmentation, as well as significant actin depolymerization. Inhibition of DNA-PKcs with NU7441 markedly attenuated all these LPS-induced pathologies, improving cardiac function, preserving microvascular structure, preventing mitochondrial fragmentation, and normalizing related gene expression and actin cytoskeleton stability. Additionally, inhibiting mitochondrial fission with Mdivi-1 significantly ameliorated LPS-induced cardiac dysfunction and microvascular injury. Conclusions: Our findings suggest that LPS triggers a DNA-PKcs-dependent DDR that promotes mitochondrial fragmentation and actin disruption, particularly in cardiac ECs, contributing to sepsis-induced cardiomyopathy. Targeting DNA-PKcs or mitochondrial fission may hold therapeutic potential for the treatment of sepsis-induced cardiomyopathy.
Collapse
Affiliation(s)
- Ying Tan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Critical Care Medicine, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yue Ouyang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Critical Care Medicine, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Lushan Xiao
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jianming Huang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Critical Care Medicine, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Fuye Li
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Critical Care Medicine, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Zisheng Ma
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Critical Care Medicine, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Chuhong Tan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Critical Care Medicine, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Weibin Feng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Critical Care Medicine, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Erica Davis
- School of Pharmacy, University of Phoenix, 4035 S Riverpoint Pkwy, Phoenix, AZ 85040, United States
- Faculty of International Education Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Yaoping Tang
- Faculty of International Education Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Xing Chang
- School of Pharmacy, University of Phoenix, 4035 S Riverpoint Pkwy, Phoenix, AZ 85040, United States
- Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Haixia Li
- Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
He Q, Chang X, Zhang H, Hao Q, Zhi J, Shi H, Tian Y, Zhou H, Tan Y, Zheng J, Qiu J, Tao J. Nuclear damage-induced DNA damage response coupled with IFI16-driven ECM remodeling underlies dilated cardiomyopathy. Theranostics 2025; 15:5998-6021. [PMID: 40365289 PMCID: PMC12068297 DOI: 10.7150/thno.112247] [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: 02/15/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Rationale: Dilated cardiomyopathy (DCM) is a severe cardiac condition characterized by ventricular dilation and systolic dysfunction, often leading to heart failure. While the DNA damage response (DDR) pathway is increasingly implicated in DCM pathogenesis, the precise mechanisms linking DDR activation to specific pathological features like adverse extracellular matrix (ECM) remodeling and fibrosis remain poorly understood. Interferon-inducible protein 16 (IFI16), a known DNA sensor involved in DDR and inflammatory signaling, emerges as a potential mediator in this process. This study aimed to investigate the role of the DDR-IFI16 axis in DCM, specifically exploring its connection to ECM dysregulation and cardiac dysfunction, and to evaluate its potential as a therapeutic target. Methods: W This study integrated bioinformatics analyses of human cardiac transcriptomic datasets with experimental validation in a doxorubicin-induced murine DCM model. Cardiac function was assessed by echocardiography. Key molecular pathways were investigated using qPCR, ELISA, and enrichment analyses. Mechanistic roles were tested via pharmacological DDR inhibition in vivo and targeted IFI16 siRNA knockdown in vitro, followed by analysis of fibrosis, cell viability, and cytotoxicity markers. Results: Bioinformatic analyses consistently revealed activation of DDR and cytosolic DNA sensing pathways across human iPSC-CM models and ex vivo DCM heart tissue. WGCNA identified a key gene module strongly associated with DCM, co-enriched for DDR, DNA replication, and ECM/TGF-β signaling pathways. Single-cell RNA-seq analysis confirmed significant IFI16 upregulation in human DCM samples. High IFI16 expression strongly correlated with pathways governing 'Extracellular matrix organization' and key fibrotic genes. Experimental validation in the doxorubicin mouse model confirmed DDR activation. Crucially, in vivo treatment with the DDR inhibitor NU7441 significantly attenuated IFI16 upregulation, ameliorated cardiac dysfunction, and decreased cardiac fibrosis markers. Complementarily, in vitro knockdown of IFI16 significantly reduced pro-fibrotic markers, increased cell viability, and decreased cell injury. Conclusions: Our findings delineate a novel pathogenic axis in DCM where nuclear stress-induced DDR activation drives the upregulation of the DNA sensor IFI16. IFI16 acts as a critical mediator linking DDR signaling to pathological ECM remodeling and fibrosis. Pharmacological inhibition of the upstream DDR pathway effectively mitigates IFI16 induction, attenuates cardiac fibrosis, and improves cardiac function. This study identifies the DDR-IFI16-ECM remodeling axis as a crucial contributor to DCM pathogenesis and highlights its potential as a therapeutic target for mitigating adverse cardiac remodeling and dysfunction.
Collapse
Affiliation(s)
- Qingyong He
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Hui Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Hubei University of Chinese Medicine, Hubei, Wuhan 430065, China
| | - Qianying Hao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jianguo Zhi
- The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hongshuo Shi
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yingjie Tian
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Hao Zhou
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Ying Tan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junmeng Zheng
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Junxiong Qiu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| |
Collapse
|
3
|
Wang J, Chang X, Li C, Gao J, Guo Z, Zhuang H, Wang L, Huang Y, Wang W, Li C, He Q. DNA-PKcs-Driven YAP1 Phosphorylation and Nuclear Translocation: a Key Regulator of Ferroptosis in Hyperglycemia-Induced Cardiac Dysfunction in Type 1 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412698. [PMID: 40279648 DOI: 10.1002/advs.202412698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/12/2025] [Indexed: 04/27/2025]
Abstract
The DNA-Dependent Protein Kinase catalytic subunit (DNA-PKcs) acts as a principal executor in the DNA damage response (DDR), mediating the phosphorylation of a broad spectrum of substrates integral to DNA repair and apoptosis. This investigation seeks to discern the possible association and mechanisms linking hyperglycemia-induced ferroptosis and DNA-PKcs in DCM. This data exhibits a substantial activation of DNAPKcs- dependent DDR in mice with streptozotocin-induced DCM. However, deletion of DNA-PKcs in cardiomyocytes notably mitigates DNA damage, enhances heart function and dampens the inflammatory response. Co-IP/MS analysis and subsequent validation experiments demonstrate that DNA-PKcs directly interacts with and phosphorylates YAP1 at Thr226. This phosphorylation event facilitates the nuclear retention of YAP1, where it intensifies the transcription of ferroptosis-associated genes. Knockin mice expressing a nonphosphorylatable T226A YAP1 mutant display decreased ferroptosis, reduced myocardial fibrosis and improved heart function. Taken together, this study unravels that DDR acts as an intracellular stress damage sensor, perceiving hyperglycemic conditions and subsequently transmitting the damage signal to incite ferroptosis through the interplay between DNA-PKcs and YAP1. This novel insight suggests that the DNA-PKcs-mediated YAP1 phosphorylation and the ferroptosis activation could be the promising therapeutic targets for the management of DCM.
Collapse
Affiliation(s)
- Junyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Chun Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jing Gao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Zhijiang Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Haowen Zhuang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Lingjun Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yusheng Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Wei Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Chao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Qingyong He
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| |
Collapse
|
4
|
Wang Z, Yang Y, Wang N, Lu L, Xu C, Ren J, Yang L. RIP3 orchestrates oxidative stress and pyroptosis in doxorubicin-induced cardiotoxicity through regulation of AKT/Nrf2 signaling cascade. Mol Cell Biochem 2025; 480:2331-2343. [PMID: 38955910 DOI: 10.1007/s11010-024-05029-6] [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: 02/21/2024] [Accepted: 05/04/2024] [Indexed: 07/04/2024]
Abstract
This study was designed to explore the role of RIP3 in DOX-induced cardiotoxicity and its underlying molecular mechanisms. Our results demonstrate that RIP3 exacerbates DOX-induced cardiotoxicity through promoting oxidative stress and pyroptosis by regulating the AKT/Nuclear factor erythroid 2-related factor 2 (Nrf2) signal pathway. Inhibition of RIP3 using GSK-872 attenuated DOX-induced cardiac remodeling and contractile dysfunction. Moreover, using GSK-872 in vivo, the results revealed that inhibition of RIP3 alleviated DOX-induced cardiotoxicity by the resulting inhibition of oxidative stress and pyroptosis. In addition, inhibition of RIP3 increased the protein levels of AKT and Nrf2 in DOX-treated mouse hearts. Furthermore, the AKT inhibitor LY294002 lessened RIP3 reduction-offered protection against DOX-induced H9c2 cell injury by moderating oxidative stress and pyroptosis. Taken together, these data demonstrate that RIP3 activation orchestrates DOX-induced cardiotoxicity through elevated oxidative stress and pyroptosis in an AKT/Nrf2-dependent manner. Those findings highlight the clinical relevance and therapeutic potential of targeting RIP3 for the treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhenyi Wang
- Department of Anesthesiology, Children's Hospital Affiliated to Xi'an Jiao Tong University, Xi'an, 710003, Shaanxi, China
| | - Yitong Yang
- Department of Children's Respiratory Asthma, Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xian Yang, 712046, Shaanxi, China
| | - Nisha Wang
- Department of Anesthesiology, Children's Hospital Affiliated to Xi'an Jiao Tong University, Xi'an, 710003, Shaanxi, China
| | - Linhe Lu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Chennian Xu
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhong Shan Hospital Fudan University, Shanghai, China
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - Lifang Yang
- Department of Anesthesiology, Children's Hospital Affiliated to Xi'an Jiao Tong University, Xi'an, 710003, Shaanxi, China.
| |
Collapse
|
5
|
Wang Z, Zhu Y, Yao Y, Zhang W, Wang B, Wang J, Yang Y, Liu L. Natural products targeting regulated cell deaths for adriamycin-induced cardiotoxicity. Cell Death Discov 2025; 11:112. [PMID: 40118839 PMCID: PMC11928682 DOI: 10.1038/s41420-025-02389-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/01/2025] [Accepted: 03/06/2025] [Indexed: 03/24/2025] Open
Abstract
Adriamycin (ADR), as an anti-cancer drug in routine clinical application, is utilized to treat various cancers such as ovarian cancer, hematological malignant tumor, and endometrial carcinoma. However, its serious dose-dependent cardiotoxicity extremely limits its clinical application. Currently, there remains a dearth of therapeutic agents to mitigate ADR-induced cardiotoxicity. Extensive research has demonstrated that ADR can simultaneously trigger various regulated cell death (RCD) pathways, such as apoptosis, autophagy, ferroptosis, necroptosis, and pyroptosis. Therefore, drugs targeting these RCD pathways may represent effective strategies for treating ADR-induced cardiotoxicity. Natural products, with their wide availability, low cost, and diverse pharmacological activities, have increasingly gained attention. Various natural products, including polyphenols, flavonoids, terpenoids, and alkaloids, can target the RCD pathways involved in ADR-induced cardiotoxicity. Furthermore, these natural products have exhibited excellent properties in preclinical studies or in vitro experiments. This review summarizes the mechanisms of RCD in ADR-induced cardiotoxicity and systematically reviews the natural products targeting these RCD pathways. Finally, we propose future research directions of natural products in this field.
Collapse
Affiliation(s)
- Zheng Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, 627 Wuluo Road, Wuhan, 430070, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yu Yao
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Wenyu Zhang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Bo Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Jing Wang
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Liwen Liu
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
6
|
Jiang R, Guo J, Yang S, Zeng H, Wei J, Jin X, Zheng X, Sun W, Zhang Y, Hu Z. Trichodenoids A and B, Two Skeletally Unprecedented Polyketides from Trichoderma reesei with Cardioprotective Effects against H 2O 2-Induced Injury. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6736-6744. [PMID: 40047798 DOI: 10.1021/acs.jafc.5c00957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Trichoderma reesei, recognized by the FDA as a food-safe strain, plays a vital role in food fermentation. Although the enzymatic applications of T. reesei are well-established, the health benefits of its fermentation-derived metabolites are yet to be fully explored. Trichodenoids A (1) and B (2), two skeletally unprecedented polyketides, were isolated from the endophytic fungus T. reesei originating from the plant Gastrodia elata Blume. Their structures were elucidated via spectroscopic data, single-crystal X-ray crystallographic analysis, quantum chemical DP4+ analysis, and ECD calculation. Compounds 1 and 2 were uniquely defined by the unusual 6/6/5 and 6/6/5/6 ring systems, respectively, which were proposed to be formed through key Diels-Alder and Baeyer-Villiger reactions during biosynthesis. Compound 2 had the potential to mitigate H2O2-induced oxidative stress in H9C2 cells by reducing intracellular ROS levels, restoring mitochondrial function, and regulating the mRNA expression related to oxidative stress, inflammation, and autophagy. These findings highlight compound 2 as a potential candidate for natural antioxidants and even as dietary supplements for cardiovascular health.
Collapse
Affiliation(s)
- Rui Jiang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Jiayi Guo
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Shenglan Yang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Hanxiao Zeng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Jiangchun Wei
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Xiaoqi Jin
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Xinyu Zheng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Weiguang Sun
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Zhengxi Hu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311403, People's Republic of China
- Hubei Shizhen Laboratory, Wuhan 430061, People's Republic of China
| |
Collapse
|
7
|
Meng C, Wang Y, Zheng T, Rong Z, Lv Z, Wu C, Zhou X, Mao W. A novel approach to the prevention and management of chemotherapy-induced cardiotoxicity: PANoptosis. Chem Biol Interact 2025; 407:111379. [PMID: 39788474 DOI: 10.1016/j.cbi.2025.111379] [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: 11/11/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
As a fundamental component of antitumor therapy, chemotherapy-induced cardiotoxicity (CIC) has emerged as a leading cause of long-term mortality in patients with malignant tumors. Unfortunately, there are currently no effective therapeutic preventive or treatment strategies, and the underlying pathophysiological mechanisms of CIC remain inadequately understood. A growing number of studies have shown that different mechanisms of cell death, such as apoptosis, pyroptosis, and necroptosis, are essential for facilitating the cardiotoxic effects of chemotherapy. The PANoptosis mode represents a highly synchronized and dynamically balanced programmed cell death (PCD) process that integrates the principal molecular characteristics of necroptosis, apoptosis, and pyroptosis. Recent research has revealed a significant correlation between PANoptosis and the apoptosis of tumor cells. Chemotherapy drugs can activate PANoptosis, which is involved in the development of cardiovascular diseases. These findings suggest that PANoptosis marks the point where the effectiveness of chemotherapy against tumors overlaps with the onset and development of cardiovascular diseases. Furthermore, previous studies have demonstrated that CIC can simultaneously induce pyrodeath, apoptosis, and necrotic apoptosis. Therefore, PANoptosis may represent a potential mechanism and target for the prevention of CIC. This study explored the interactions among the three main mechanisms of PCD, pyroptosis, apoptosis, and necroptosis in CICs and analyzed the relevant literature on PANoptosis and CICs. The purpose of this work is to serve as a reference for future investigations on the role of PANoptosis in the development and mitigation of cardiotoxicity associated with chemotherapy.
Collapse
Affiliation(s)
- Chenchen Meng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Yali Wang
- Department of Cardiology, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China
| | - Tiantian Zheng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Zheng Rong
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Zhengtian Lv
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Chenxia Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053, Hangzhou, China; Department of Cardiology, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China
| | - Xinbin Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 310006, Hangzhou, Zhejiang, China.
| | - Wei Mao
- Department of Cardiology, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China; Zhejiang Key Laboratory of Integrative Chinese and Western Medicine for Diagnosis and Treatment of Circulatory Diseases, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China; Zhejiang Engineering Research Center for Precise Diagnosis and Innovative Traditional Chinese Medicine for Cardiovascular Diseases, Zhejiang Hospital (Affiliated Zhejiang Hospital, Zhejiang University School of Medicine), Hangzhou, Zhejiang, 310007, China.
| |
Collapse
|
8
|
Qaed E, Almoiliqy M, Liu W, Al-Mashriqi HS, Alyafeai E, Aldahmash W, Mahyoub MA, Tang Z. Protective effects of phosphocreatine against Doxorubicin-Induced cardiotoxicity through mitochondrial function enhancement and apoptosis suppression via AMPK/PGC-1α signaling pathway. Int Immunopharmacol 2025; 144:113677. [PMID: 39580863 DOI: 10.1016/j.intimp.2024.113677] [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: 09/05/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Doxorubicin (DOX), a potent chemotherapy drug, is limited by its cardiotoxic effects, which can lead to heart damage. This study explores the cardioprotective potential of Phosphocreatine (PCr) in vitro and in vivo models, focusing on its impact on the AMPK and PGC-1α pathways, apoptosis reduction, and mitochondrial function preservation. Advanced methodologies, including high-resolution respirometry (HRR), were employed to assess mitochondrial bioenergetics, AMPK activity, and apoptotic rates in cardiomyocytes. Electrocardiography (ECG) and echocardiography (echo) were used to monitor cardiac function in vivo. Results showed that PCr significantly activated the AMPK and PGC-1α pathways, reduced apoptosis, and stabilized mitochondrial function in cardiomyocytes exposed to DOX. There was an upregulation of AMPK and PGC-1α target genes, stabilization of mitochondrial membranes, and improvements in cellular energy production and antioxidant defenses. PCr also markedly reduced apoptotic markers, enhancing cardiomyocyte viability. ECG and echocardiography revealed that PCr preserved cardiac function, indicated by improved heart rate variability, reduced QT interval prolongation, and enhanced ejection fraction. These findings highlight PCr's potential in mitigating DOX-induced cardiotoxicity by enhancing mitochondrial function and reducing apoptosis. The study underscores the promise of PCr as an agent to reduce chemotherapy-related cardiac injuries, paving the way for further research to improve patient outcomes in cancer treatment.
Collapse
Affiliation(s)
- Eskandar Qaed
- Collage of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China; State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Marwan Almoiliqy
- Collage of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China
| | - Wu Liu
- Collage of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China
| | | | - Eman Alyafeai
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Waleed Aldahmash
- Zoology Department, College of Science, King Saud University, P. O. Box 2455, 11451 Riyadh, Saudi Arabia
| | - Mueataz A Mahyoub
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zeyao Tang
- Collage of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China.
| |
Collapse
|
9
|
Szponar J, Ciechanski E, Ciechanska M, Dudka J, Mandziuk S. Evolution of Theories on Doxorubicin-Induced Late Cardiotoxicity-Role of Topoisomerase. Int J Mol Sci 2024; 25:13567. [PMID: 39769331 PMCID: PMC11678604 DOI: 10.3390/ijms252413567] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Doxorubicin (DOX) has been widely used as a cytotoxic chemotherapeutic. However, DOX has a number of side effects, such as myelotoxicity or gonadotoxicity, the most dangerous of which is cardiotoxicity. Cardiotoxicity can manifest as cardiac arrhythmias, myocarditis, and pericarditis; life-threatening late cardiotoxicity can result in heart failure months or years after the completion of chemotherapy. The development of late cardiomyopathy is not yet fully understood. The most important question is how DOX reprograms the cardiomyocyte, after which DOX is excreted from the body, initially without symptoms. However, clinically overt cardiomyopathy develops over the following months and years. Since the 1980s, DOX-induced disorders in cardiomyocytes have been thought to be related to oxidative stress and dependent on the Fe/reactive oxygen species (ROS) mechanism. That line of evidence was supported by dexrazoxane (DEX) protection, the only Food and Drug Administration (FDA)-approved drug for preventing DOX-induced cardiomyopathy, which complexes iron. Thus, the hypothesis related to Fe/ROS provides a plausible explanation for the induction of the development of late cardiomyopathy via DOX. However, in subsequent studies, DEX was used to identify another important mechanism in DOX-induced cardiomyopathy that is related to topoisomerase 2β (Top2β). Does the Top2β hypothesis explain the mechanisms of the development of DOX-dependent late heart failure? Several of these mechanisms have been identified to date, proving the involvement of Top2β in the regulation of the redox balance, including oxidative stress. Thus, the development of late cardiomyopathy can be explained based on mechanisms related to Top2β. In this review, we highlight free radical theory, iron imbalance, calcium overload, and finally, a theory based on Top2β.
Collapse
Affiliation(s)
- Jaroslaw Szponar
- Toxicology Clinic, Faculty of Medicine, Medical University of Lublin, Krasnicka 100, 20-718 Lublin, Poland;
- Clinical Department of Toxicology and Cardiology, Regional Specialist Hospital, Krasnicka 100, 20-718 Lublin, Poland
| | - Erwin Ciechanski
- Department of Cardiology, Regional Specialist Hospital, Krasnicka 100, 20-718 Lublin, Poland
| | - Magda Ciechanska
- Department of Pulmonary Diseases and Children Rheumatology, Medical University of Lublin, Antoniego Gebali 6, 20-093 Lublin, Poland
| | - Jaroslaw Dudka
- Department of Toxicology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Sławomir Mandziuk
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland;
| |
Collapse
|
10
|
Zhao M, Zheng Z, Liu J, Xu Y, Zhang J, Peng S, Qin JJ, Wan J, Wang M. LGR6 protects against myocardial ischemia-reperfusion injury via suppressing necroptosis. Redox Biol 2024; 78:103400. [PMID: 39471639 PMCID: PMC11550357 DOI: 10.1016/j.redox.2024.103400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024] Open
Abstract
Regulated necrosis (necroptosis) and apoptosis are important biological features of ischemia-reperfusion (I/R) injury. However, the molecular mechanisms underlying myocardial necroptosis remain elusive. Leucine rich repeat containing G protein-coupled receptor 6 (LGR6) has been reported to play important roles in various cardiovascular disease. In this study, we aimed to determine whether LGR6 suppresses I/R-induced myocardial necroptosis and the underlying molecular mechanisms. We generated LGR6 knockout mice and used ligation of left anterior descending coronary artery to produce an in vivo I/R model. The effects of LGR6 and its downstream molecules were subsequently identified using RNA sequencing and CHIP assays. We observed significantly downregulated LGR6 expression in hearts post myocardial I/R and cardiomyocytes post hypoxia and reoxygenation (HR). LGR6 deficiency promoted and LGR6 overexpression inhibited necroptosis and acute myocardial injury after I/R. Mechanistically, in vivo and in vitro experiments suggest that LGR6 regulates the expression of STAT2 and ZBP1 by activating the Wnt signaling pathway, thereby inhibiting cardiomyocyte necroptosis after HR. Inhibiting STAT2 and ZBP1 effectively alleviated the aggravating effect of LGR6 deficiency on myocardial necroptosis after I/R. Furthermore, activating LGR6 with RSPO3 also effectively protected mice from acute myocardial I/R injury. Our findings reveal that RSPO3-LGR6 axis downregulates the expression of STAT2 and ZBP1 through the Wnt signaling pathway, thereby inhibiting I/R-induced myocardial injury and necroptosis. Targeting the RSPO3-LGR6 axis may be a potential therapeutic strategy to treat myocardial I/R injury.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
11
|
Wang J, Ma H, Guo H, Chen Y, Liu Y. Clinical applications of phosphocreatine and related mechanisms. Life Sci 2024; 355:123012. [PMID: 39181314 DOI: 10.1016/j.lfs.2024.123012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Phosphocreatine (PCr), a naturally occurring creatine phosphorylated molecule, is a high-energy phosphate compound that is one of the most important substances involved in cell energy metabolism, and also has anti-apoptosis and anti-oxidative stress effects. It is precisely because of its role in maintaining energy homeostasis that PCr is widely used in diseases related to energy damage. In the regulation of cell signal, PCr mainly plays a role through MAPK, NF-κB, PI3K/AKT, ERK/Nrf2/HO-1 and JAK2/STAT3. In clinical applications, PCr is commonly used as a cardioprotective drug, such as ischemic heart disease, myocardial fibrosis, myocardial infarction, arrhythmia, and myocarditis. In recent years, further research on PCr has found that PCr also has a positive role in the treatment of other diseases, including diabetes-induced liver injury, kidney injury, cerebral ischemia-reperfusion injury, and neurodegenerative diseases. In this paper, the literature on PCr in three databases, Web of Sciences, SciFinder, and PubMed, was summarized and analyzed, and the research progress of PCr in recent years was reviewed, hoping to provide help for the expansion of its application in clinical therapy.
Collapse
Affiliation(s)
- Jing Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China
| | - Huizhong Ma
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China
| | - Haohao Guo
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China
| | - Yuan Chen
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China
| | - Yufeng Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China; Natural Products Pharmaceutical Engineering Technology Research Center of Liaoning Province, Shenyang 110036, China.
| |
Collapse
|
12
|
Zhang W, Shu Z, Huang P, Cheng H, Ji J, Wei D, Ren L. Adenylate cyclase 1 knockdown attenuates pirarubicin-induced cardiotoxicity. Clin Exp Pharmacol Physiol 2024; 51:e13920. [PMID: 39227014 DOI: 10.1111/1440-1681.13920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/02/2024] [Accepted: 07/14/2024] [Indexed: 09/05/2024]
Abstract
This study aimed to investigate the effects and possible mechanisms of adenylate cyclase 1 (ADCY1) on pirarubicin-induced cardiomyocyte injury. HL-1 cells were treated with pirarubicin (THP) to induce intracellular toxicity, and the extent of damage to mouse cardiomyocytes was assessed using CCK-8, Edu, flow cytometry, ROS, ELISA, RT-qPCR and western blotting. THP treatment reduced the viability of HL-1 cells, inhibited proliferation, induced apoptosis and triggered oxidative stress. In addition, the RT-qPCR results revealed that ADCY1 expression was significantly elevated in HL-1 cells, and molecular docking showed a direct interaction between ADCY1 and THP. Western blotting showed that ADCY1, phospho-protein kinase A and GRIN2D expression were also significantly elevated. Knockdown of ADCY1 attenuated THP-induced cardiotoxicity, possibly by regulating the ADCY1/PKA/GRIN2D pathway.
Collapse
Affiliation(s)
- Wenqing Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Zhiyun Shu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Peng Huang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - HongYuan Cheng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jiahua Ji
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Dexian Wei
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
13
|
Li J, Wang H, Guo M, Guo Q, Li Y. Combination of Exogenous Spermidine and Phosphocreatine Efficiently Improved the Quality and Antioxidant Capacity of Cryopreserved Boar Sperm and Reduced Apoptosis-Like Changes. Mol Reprod Dev 2024; 91:e70003. [PMID: 39445630 DOI: 10.1002/mrd.70003] [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: 03/16/2024] [Revised: 09/24/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
The low resistance of boar sperm to cryopreservation dictates that addition antioxidants and energetic substances to the diluent to improve sperm quality is necessary. This study evaluated the effect of spermidine and phosphocreatine in combination on the quality, antioxidant capacity, and antiapoptotic-like changes capacity of cryopreserved boar sperm based on previous reports. The results showed that the combined application of spermidine and phosphocreatine significantly enhanced the motility, average path velocity, straight-line velocity, curvilinear velocity, beat cross frequency, acrosome integrity, plasma membrane integrity, mitochondrial activity, and DNA integrity compared with the control group (p < 0.05). In addition, the combined application of spermidine and phosphocreatine significantly enhanced the total antioxidant capacity, superoxide dismutase activity, glutathione peroxidase activity, and catalase activity while significantly decreasing malondialdehyde content and hydrogen peroxide content (p < 0.05). Western Blot analysis further showed that spermidine and phosphocreatine significantly decreased the expression of CASP3 and BAX and significantly enhanced the expression of BCL2 (p < 0.05); therefore, the combination of spermidine and phosphocreatine has potentially positive implications for improving the quality of cryopreserved boar sperm.
Collapse
Affiliation(s)
- Jingchun Li
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Hechuan Wang
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Minghui Guo
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Qing Guo
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Yanbing Li
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| |
Collapse
|
14
|
Zhou C, Yang Y, Hu L, Meng X, Guo X, Lei M, Ren Z, Chen Q, Ouyang C, Yang X. Effects of miR-143 regulation on cardiomyocytes apoptosis in doxorubicin cardiotoxicity based on integrated bioinformatics analysis. Toxicol In Vitro 2023; 93:105662. [PMID: 37597758 DOI: 10.1016/j.tiv.2023.105662] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/26/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
This study aimed to investigate the effect of miRNAs involving oxidative stress response in doxorubicin (DOX)-induced cardiotoxicity based on the data from Gene Expression Omnibus (GEO) database and experimental results via integrated bioinformatics analysis. MiRNA expression profiles of DOX-induced cardiotoxicity in rat myocardial tissues and adult rat cardiomyocytes (ARC) were extracted from GEO datasets (GSE36239). Differential expression miRNA (DEMs) were separately captured in rat myocardial tissues and in ARC, and intersected between rat myocardial tissues and ARC via Venny 2.1. Subsequently, Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) analyzed 46 target genes of miR-143, one of 6 DEMs, and HIF-1 and PI3K-Akt signaling pathway were significantly enriched. Further experimental results showed DOX-induced oxidative stress downregulated the expression of miR-143, and then promoted target gene Bbc3 expression and H9c2 apoptosis, the intervention of phosphocreatine (PCr) or N-acetyl-L-cystine (NAC) alleviated oxidative stress, apoptosis and Bbc3 expression, upregulated miR-143 in DOX-induced cardiotoxicity in vivo and in vitro. Our findings elucidated the regulatory network between miR-143 and oxidative stress in DOX-induced cardiotoxicity, and might unveiled a potential biomarker and molecular mechanisms, which could be helpful to the diagnosis and treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Chi Zhou
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Yayuan Yang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Ling Hu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Department of Pharmacy, University of South China Affiliated Changsha Central Hospital, Changsha 410004, PR China
| | - Xiangwen Meng
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China.
| | - Xiying Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Min Lei
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Zhanhong Ren
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China.
| | - Qingjie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Changhan Ouyang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China.
| | - Xiaosong Yang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning 437100, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China.
| |
Collapse
|
15
|
Yang J, Sun P, Xu X, Liu X, Lan L, Yi M, Xiao C, Ni R, Fan Y. TAK1 Improves Cognitive Function via Suppressing RIPK1-Driven Neuronal Apoptosis and Necroptosis in Rats with Chronic Hypertension. Aging Dis 2023; 14:1799-1817. [PMID: 37196118 PMCID: PMC10529759 DOI: 10.14336/ad.2023.0219] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/19/2023] [Indexed: 05/19/2023] Open
Abstract
Chronic hypertension is a major risk factor for cognitive impairment, which can promote neuroinflammation and neuronal loss in the central nervous system. Transforming growth factor β-activated kinase 1 (TAK1) is a key molecular component in determining cell fate and can be activated by inflammatory cytokines. This study aimed to investigate the role of TAK1 in mediating neuronal survival in the cerebral cortex and hippocampus under chronic hypertensive conditions. To that end, we used stroke-prone renovascular hypertension rats (RHRSP) as chronic hypertension models. Adeno-associated virus (AAV) designed to overexpress or knock down TAK1 expression were injected into the lateral ventricles of rats and the subsequent effects on cognitive function and neuronal survival under chronic hypertensive conditions were assessed. We found that, TAK1 knockdown in RHRSP markedly increased neuronal apoptosis and necroptosis and induced cognitive impairment, which could be reversed by Nec-1s, an inhibitor of receptor interacting protein kinase 1 (RIPK1). In contrast, overexpression of TAK1 in RHRSP significantly suppressed neuronal apoptosis and necroptosis and improved cognitive function. Further knockdown of TAK1 in sham-operated rats received similar phenotype with RHRSP. The results have been verified in vitro. In this study, we provide in vivo and in vitro evidence that TAK1 improves cognitive function by suppressing RIPK1-driven neuronal apoptosis and necroptosis in rats with chronic hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yuhua Fan
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| |
Collapse
|
16
|
Sheng SY, Li JM, Hu XY, Wang Y. Regulated cell death pathways in cardiomyopathy. Acta Pharmacol Sin 2023; 44:1521-1535. [PMID: 36914852 PMCID: PMC10374591 DOI: 10.1038/s41401-023-01068-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
Heart disease is a worldwide health menace. Both intractable primary and secondary cardiomyopathies contribute to malignant cardiac dysfunction and mortality. One of the key cellular processes associated with cardiomyopathy is cardiomyocyte death. Cardiomyocytes are terminally differentiated cells with very limited regenerative capacity. Various insults can lead to irreversible damage of cardiomyocytes, contributing to progression of cardiac dysfunction. Accumulating evidence indicates that majority of cardiomyocyte death is executed by regulating molecular pathways, including apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Importantly, these forms of regulated cell death (RCD) are cardinal features in the pathogenesis of various cardiomyopathies, including dilated cardiomyopathy, diabetic cardiomyopathy, sepsis-induced cardiomyopathy, and drug-induced cardiomyopathy. The relevance between abnormity of RCD with adverse outcome of cardiomyopathy has been unequivocally evident. Therefore, there is an urgent need to uncover the molecular and cellular mechanisms for RCD in order to better understand the pathogenesis of cardiomyopathies. In this review, we summarize the latest progress from studies on RCD pathways in cardiomyocytes in context of the pathogenesis of cardiomyopathies, with particular emphasis on apoptosis, necroptosis, ferroptosis, autophagy, and pyroptosis. We also elaborate the crosstalk among various forms of RCD in pathologically stressed myocardium and the prospects of therapeutic applications targeted to various cell death pathways.
Collapse
Affiliation(s)
- Shu-Yuan Sheng
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Jia-Min Li
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Xin-Yang Hu
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China
| | - Yibin Wang
- Department of Cardiology, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, 310009, China.
- Signature Program in Cardiovascular and Metabolic Diseases, DukeNUS Medical School and National Heart Center of Singapore, Singapore, Singapore.
| |
Collapse
|
17
|
Wang W, Zhong X, Fang Z, Li J, Li H, Liu X, Yuan X, Huang W, Huang Z. Cardiac sirtuin1 deficiency exacerbates ferroptosis in doxorubicin-induced cardiac injury through the Nrf2/Keap1 pathway. Chem Biol Interact 2023; 377:110469. [PMID: 37030624 DOI: 10.1016/j.cbi.2023.110469] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 03/04/2023] [Accepted: 03/31/2023] [Indexed: 04/10/2023]
Abstract
Doxorubicin (DOX), a broad-spectrum chemotherapeutic agent for various cancers, has limited clinical application because of its serious cardiotoxicity, which is due to different mechanisms, including cardiac ferroptosis and oxidative stress. Some drugs, such as berberine or dioscin, show efficacy in impeding DOX-induced cardiotoxicity by activating Sirtuin 1 (Sirt1). However, there is no direct evidence to clarify the role of Sirt1 in DOX-induced cardiomyopathy and its underlying role in cardiac ferroptosis. In this study, C57BL/6 and cardiac-specific Sirt1-/- knockout mice were used as a DOX-induced cardiotoxicity model. We found that cardiac Sirt1 was downregulated, oxidative stress was increased and ferroptosis were obviously enhanced, as reflected by decreased Glutathione peroxidase 4 (GPX4) and increased Heme oxygenase 1 (Hmox-1), exposure to DOX treatment in mice and H9c2 cells compared with the control. And Sirt1 activation was resistant to cardiac injury induced by DOX, as observed the improvement of cardiac dysfunction, and the reduction of cardiac fibrosis. However, cardiac Sirt1 deficiency aggravated Dox-induced cardiac dysfunction and cardiac remodeling, further downregulated GPX4, upregulated Hmox-1 expression and increased ROS level. In addition, Sirt1-siRNA exacerbated DOX-induced cardiotoxicity in H9c2 cells, which is similar to the results obtained in vivo. Furthermore, DOX decrease Nrf2 translocation from the cytosol to the nucleus, and Sirt1 deficiency further restrain the process, as well as the downstream Keap1 pathways, in DOX-induced cardiotoxicity. This study provides direct evidence that Sirt1 plays a protective role in DOX-induced cardiotoxicity by mediating ferroptosis reduction via the Nrf2/Keap1 pathway.
Collapse
Affiliation(s)
- Weiqi Wang
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Xin Zhong
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Zimin Fang
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Hebo Li
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Xuesheng Liu
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Xindi Yuan
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Weijian Huang
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China
| | - Zhouqing Huang
- Department of Cardiology, The First Affiliated Hospital of WenZhou Medical College, 2 Fuxue Road, WenZhou, ZheJiang, 325000, PR China.
| |
Collapse
|
18
|
Chen Y, Lin W, Chen P, Ye B, Luo W, Wang X, Huang W, Wu G, Liang G. Tabersonine alleviates obesity-induced cardiomyopathy by binding to Transforming growth factor activated kinase 1 (TAK1) and inhibiting TAK1-mediated inflammation. Phytother Res 2023; 37:860-871. [PMID: 36420902 DOI: 10.1002/ptr.7666] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/27/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022]
Abstract
Obesity-induced cardiomyopathy (OIC) is an increasingly serious global disease caused by obesity. Chronic inflammation greatly contributes to the pathogenesis of OIC. This study aimed to explore the role and mechanism of tabersonine (Tab), a natural alkaloid with antiinflammatory activity, in the treatment of OIC. High fat diet (HFD)-induced obese mice were administered with Tab. The results showed that Tab significantly inhibit inflammation, myocardial fibrosis, and hypertrophy to prevent heart dysfunction, without the alteration of body weight and hyperlipidemia, in HFD-induced obese mice. H9c2 cells and primary cardiomyocytes stimulated by palmitic acid (PA) were used to explore the molecular mechanism and target of Tab. We examined the effect of Tab on key proteins involved in HFD/PA-induced inflammatory signaling pathway and found that Tab significantly inhibits TAK1 phosphorylation in cardiomyocytes. We further detected the direct interaction between Tab and TAK1 at the cellular, animal, and molecular levels. We found that Tab directly binds to TAK1 to inhibit TAK1 phosphorylation, which then blocks TAK1-TAB2 interaction and then NF-κB pro-inflammatory pathway in cultured cardiomyocytes. Our results indicate that Tab is a potential agent for the treatment of OIC, and TAK1 is an effective therapeutic target for this disease.
Collapse
Affiliation(s)
- Yanghao Chen
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wante Lin
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Pan Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Gaojun Wu
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Guang Liang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
19
|
Bi Y, Xu H, Wang X, Zhu H, Ge J, Ren J, Zhang Y. FUNDC1 protects against doxorubicin-induced cardiomyocyte PANoptosis through stabilizing mtDNA via interaction with TUFM. Cell Death Dis 2022; 13:1020. [PMID: 36470869 PMCID: PMC9723119 DOI: 10.1038/s41419-022-05460-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
Doxorubicin (DOX) is an effective anthracycline chemotherapeutic anticancer drug with its life-threatening cardiotoxicity severely limiting its clinical application. Mitochondrial damage-induced cardiomyocyte death is considered an essential cue for DOX cardiotoxicity. FUN14 domain containing 1 (FUNDC1) is a mitochondrial membrane protein participating in the regulation of mitochondrial integrity in multiple diseases although its role in DOX cardiomyopathy remains elusive. Here, we examined whether PANoptosis, a novel type of programmed cell death closely associated with mitochondrial damage, was involved in DOX-induced heart injury, and FUNDC1-mediated regulation of cardiomyocyte PANoptosis, if any. FUNDC1 was downregulated in heart tissues in patients with dilated cardiomyopathy (DCM) and DOX-challenged mice. FUNDC1 deficiency aggravated DOX-induced cardiac dysfunction, mitochondrial injury, and cardiomyocyte PANoptosis. Further examination revealed that FUNDC1 countered cytoplasmic release of mitochondrial DNA (mtDNA) and activation of PANoptosome through interaction with mitochondrial Tu translation elongation factor (TUFM), a key factor in the translational expression and repair of mitochondrial DNA, via its 96-133 amino acid domain. TUFM intervention reversed FUNDC1-elicited protection against DOX-induced mtDNA cytosolic release and cardiomyocyte PANoptosis. Our findings shed light toward a beneficial role of FUNDC1 in DOX cardiotoxicity and cardiomyocyte PANoptosis, thus offering therapeutic promises in DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Yaguang Bi
- grid.8547.e0000 0001 0125 2443Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 200032 Shanghai, China ,National Clinical Research Center for Interventional Medicine, 200032 Shanghai, China
| | - Haixia Xu
- grid.8547.e0000 0001 0125 2443Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 200032 Shanghai, China ,grid.440642.00000 0004 0644 5481Department of Cardiology, Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu China
| | - Xiang Wang
- grid.8547.e0000 0001 0125 2443Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 200032 Shanghai, China ,National Clinical Research Center for Interventional Medicine, 200032 Shanghai, China
| | - Hong Zhu
- grid.16821.3c0000 0004 0368 8293Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, 200125 Shanghai, China
| | - Junbo Ge
- grid.8547.e0000 0001 0125 2443Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 200032 Shanghai, China ,National Clinical Research Center for Interventional Medicine, 200032 Shanghai, China
| | - Jun Ren
- grid.8547.e0000 0001 0125 2443Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 200032 Shanghai, China ,National Clinical Research Center for Interventional Medicine, 200032 Shanghai, China ,grid.34477.330000000122986657Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195 USA
| | - Yingmei Zhang
- grid.8547.e0000 0001 0125 2443Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 200032 Shanghai, China ,National Clinical Research Center for Interventional Medicine, 200032 Shanghai, China
| |
Collapse
|
20
|
The regulation of necroptosis and perspectives for the development of new drugs preventing ischemic/reperfusion of cardiac injury. Apoptosis 2022; 27:697-719. [DOI: 10.1007/s10495-022-01760-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 12/11/2022]
|
21
|
Zhou L, Han Y, Yang Q, Xin B, Chi M, Huo Y, Guo C, Sun X. Scutellarin attenuates doxorubicin-induced oxidative stress, DNA damage, mitochondrial dysfunction, apoptosis and autophagy in H9c2 cells, cardiac fibroblasts and HUVECs. Toxicol In Vitro 2022; 82:105366. [PMID: 35470029 DOI: 10.1016/j.tiv.2022.105366] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/03/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023]
Abstract
Studies on doxorubicin (DOX)-induced cardiotoxicity have mainly focused on cardiomyocytes (CMs), but it is unclear whether there are differences in the toxicity degree of DOX to CMs, cardiac fibroblasts (CFs) and endothelial cells (ECs). We used H9c2 cells, rat primary isolated CFs and human umbilical vein endothelial cells (HUVECs) to systematically research the cytotoxicity of DOX. Scutellarin (SCU) is a natural polyphenolic flavonoid that exerts a cardioprotective effect. In the present study, we explored the protective effects of SCU on DOX-induced cytotoxicity in H9c2 cells, CFs and HUVECs. The results showed that DOX decreased cell viability and increased the apoptosis rate, whereas DOX had a greater killing effect on H9c2 cells compared to CFs and HUVECs. DOX significantly elevated oxidative stress, but the malondialdehyde (MDA) levels in H9c2 cells were higher after DOX treatment. In all three cell types, DOX induced DNA damage and mitochondrial dysfunction, it activated apoptosis by activation of Bax/ Bcl-2 and it induced autophagy by inhibiting the Akt/ mTOR pathway. Pretreatment with different concentrations of SCU reversed these phenomena in a dose-dependent manner. Collectively, these results revealed that there were slight differences in DOX-induced cytotoxicity among H9c2 cells, CFs and HUVECs. Furthermore, the cardioprotective effect of SCU may be attributed to attenuation of DOX-induced oxidative stress, DNA damage, mitochondrial dysfunction, apoptosis and autophagy.
Collapse
|
22
|
Li Y, Hao H, Yu H, Yu L, Ma H, Zhang H. Ginsenoside Rg2 Ameliorates Myocardial Ischemia/Reperfusion Injury by Regulating TAK1 to Inhibit Necroptosis. Front Cardiovasc Med 2022; 9:824657. [PMID: 35391841 PMCID: PMC8981204 DOI: 10.3389/fcvm.2022.824657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/11/2022] [Indexed: 11/13/2022] Open
Abstract
Necroptosis contribute to the pathogenesis of myocardial ischemia/reperfusion (MI/R) injury. Ginsenoside Rg2 has been reported to have cardioprotective effects against MI/R injury; however, the underlying mechanism remains unclear. This work aimed to investigate the effect of ginsenoside Rg2 on necroptosis induced by MI/R and to explore the mechanism. In this study, hypoxia/reoxygenation (H/R) injury model was established in H9c2 cells. In vivo, male C57/BL6 mice were subjected to myocardial ischemia 30 min/reperfusion 4 h. Rg2 (50 mg/kg) or vehicle was intravenously infused 5 min before reperfusion. Cardiac function and the signaling pathway involved in necroptosis were investigated. Compared with H/R group, Rg2 significantly inhibited H/R-induced cardiomyocyte death. Rg2 treatment effectively inhibited the phosphorylation of RIP1, RIP3, and MLKL in H/R cardiomyocytes, and inhibited RIP1/RIP3 complex (necrosome) formation. In mice, Rg2 treatment manifested significantly lower ischemia/reperfusion (I/R)-induced myocardial necroptosis, as evidenced by decrease in phosphorylation of RIP1, RIP3, and MLKL, inhibited lactate dehydrogenase (LDH) release and Evans blue dye (EBD) penetration. Mechanically, an increased level of tumor necrosis factor α (TNFα), interleukin (IL)-1β, IL-6, and MCP-1 were found in MI/R hearts, and Rg2 treatment significantly inhibit the expression of these factors. We found that TNFα-induced phosphorylation of RIP1, RIP3, and MLKL was negatively correlated with transforming growth factor-activated kinase 1 (TAK1) phosphorylation, and inhibition of TAK1 phosphorylation led to necroptosis enhancement. More importantly, Rg2 treatment significantly increased TAK1 phosphorylation, enhanced TAK1 binding to RIP1 while inhibiting RIP1/RIP3 complex, ultimately reducing MI/R-induced necroptosis. These findings highlight a new mechanism of Rg2-induced cardioprotection: reducing the formation of RIP1/RIP3 necrosome by regulating TAK1 phosphorylation to block necroptosis induced by MI/R.
Collapse
Affiliation(s)
- Yao Li
- Clinical Medical College of Air Force, Anhui Medical University, Hefei, China
| | - Hao Hao
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi’an, China
| | - Haozhen Yu
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Heng Ma
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi’an, China
| | - Haitao Zhang
- Clinical Medical College of Air Force, Anhui Medical University, Hefei, China
- Department of Cardiology, PLA Air Force Medical Center, Beijing, China
| |
Collapse
|
23
|
Adameova A, Horvath C, Abdul-Ghani S, Varga ZV, Suleiman MS, Dhalla NS. Interplay of Oxidative Stress and Necrosis-like Cell Death in Cardiac Ischemia/Reperfusion Injury: A Focus on Necroptosis. Biomedicines 2022; 10:biomedicines10010127. [PMID: 35052807 PMCID: PMC8773068 DOI: 10.3390/biomedicines10010127] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Extensive research work has been carried out to define the exact significance and contribution of regulated necrosis-like cell death program, such as necroptosis to cardiac ischemic injury. This cell damaging process plays a critical role in the pathomechanisms of myocardial infarction (MI) and post-infarction heart failure (HF). Accordingly, it has been documented that the modulation of key molecules of the canonical signaling pathway of necroptosis, involving receptor-interacting protein kinases (RIP1 and RIP3) as well as mixed lineage kinase domain-like pseudokinase (MLKL), elicit cardioprotective effects. This is evidenced by the reduction of the MI-induced infarct size, alleviation of myocardial dysfunction, and adverse cardiac remodeling. In addition to this molecular signaling of necroptosis, the non-canonical pathway, involving Ca2+/calmodulin-dependent protein kinase II (CaMKII)-mediated regulation of mitochondrial permeability transition pore (mPTP) opening, and phosphoglycerate mutase 5 (PGAM5)–dynamin-related protein 1 (Drp-1)-induced mitochondrial fission, has recently been linked to ischemic heart injury. Since MI and HF are characterized by an imbalance between reactive oxygen species production and degradation as well as the occurrence of necroptosis in the heart, it is likely that oxidative stress (OS) may be involved in the mechanisms of this cell death program for inducing cardiac damage. In this review, therefore, several observations from different studies are presented to support this paradigm linking cardiac OS, the canonical and non-canonical pathways of necroptosis, and ischemia-induced injury. It is concluded that a multiple therapeutic approach targeting some specific changes in OS and necroptosis may be beneficial in improving the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 83232 Bratislava, Slovakia;
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 81438 Bratislava, Slovakia
- Correspondence:
| | - Csaba Horvath
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 83232 Bratislava, Slovakia;
| | - Safa Abdul-Ghani
- Department of Physiology, Faculty of Medicine, Al-Quds University, Abu Dis P.O. Box 89, Palestine;
| | - Zoltan V. Varga
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary;
| | - M. Saadeh Suleiman
- Faculty of Health Sciences, Bristol Heart Institute, The Bristol Medical School, University of Bristol, Bristol BS8 1TH, UK;
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Center, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada;
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
24
|
Kabel AM, Salama SA, Adwas AA, Estfanous RS. Targeting Oxidative Stress, NLRP3 Inflammasome, and Autophagy by Fraxetin to Combat Doxorubicin-Induced Cardiotoxicity. Pharmaceuticals (Basel) 2021; 14:1188. [PMID: 34832970 PMCID: PMC8621693 DOI: 10.3390/ph14111188] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 01/24/2023] Open
Abstract
Doxorubicin belongs to the class of anthracycline antibiotics that is widely used in the treatment protocols of a wide range of malignancies. The major deleterious effect of doxorubicin use is the possible occurrence of cardiotoxicity. This study aimed to delineate the possible effects of targeting oxidative stress, NLRP3 inflammasome, and autophagy by fraxetin on doxorubicin-induced cardiac dysfunction in rats. In a model of doxorubicin-induced cardiotoxicity, the effects of different doses of fraxetin were assessed by determination of biochemical, histopathological, immunohistochemical, and electron microscopic changes. Fraxetin, in a dose-dependent manner, was found to have the ability to mitigate the harmful effects of oxidative stress and inflammation on myocardial muscles with significant decrease in NLRP3 inflammasome, augmentation of autophagy, and amelioration of the apoptotic signaling pathways. In addition, fraxetin, in a dose-dependent manner, had the ability to combat the echocardiographic, histopathological, immunohistochemical, and electron microscopic changes induced by doxorubicin in cardiomyocytes. As a result, fraxetin may be put into consideration as a new adjuvant line of therapy on the way to mitigate doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ahmed M. Kabel
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| | - Samir A. Salama
- Division of Biochemistry, Department of Pharmacology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Almokhtar A. Adwas
- Department of Pharmacology, Faculty of Medicine, Sabratha University, Sabratha P.O. Box 250, Libya;
| | - Remon S. Estfanous
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| |
Collapse
|