1
|
Chen J, Li F, Luo WS, Zhu MF, Zhao NJ, Zhang ZH, Chen YF, Feng DX, Yang SY, Sun WJ. Therapeutic potential of Da Cheng Qi Decoction and its ingredients in regulating ferroptosis via the NOX2-GPX4 signaling pathway to alleviate and predict severe acute pancreatitis. Cell Signal 2025; 131:111733. [PMID: 40081545 DOI: 10.1016/j.cellsig.2025.111733] [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: 08/26/2024] [Revised: 01/27/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE This study aimed to elucidate the protective effects of Da Cheng Qi Decoction (DCQD) on severe acute pancreatitis (SAP) by targeting ferroptosis in pancreatic acinar cells and to establish a predictive signature and nomogram for acute pancreatitis (AP) risk assessment. METHODS We utilized microarray analysis to delineate gene expression patterns among 32 healthy controls and 87 AP patients stratified by severity. Employing SAP models and NOX2-deficient cells, we investigated the molecular underpinnings of ferroptosis. The impact of DCQD and the ferroptosis inhibitor Fer-1 on gene expression, oxidative stress, and inflammation was assessed. Machine learning algorithms identified differentially expressed genes (DEGs) sensitive to DCQD, SAP, and ferroptosis (DSNFGs), which were validated across multiple datasets. A predictive nomogram integrating DSNFGs was developed, and single-cell analysis provided a comprehensive view of the cellular dynamics. RESULTS The microarray analysis revealed upregulation of NOX2 and downregulation of GPX4 in AP, with expression patterns correlating with disease severity. DCQD ameliorated SAP-induced pancreatic acinar cell damage and ferroptosis by reducing inflammatory markers and enhancing GPX4 expression. NOX2 knockout mitigated ferroptosis in SAP models, suggesting a key role in the disease process. DCQD and Fer-1 differentially regulated the expression of ferroptosis-related genes, reduced reactive oxygen species (ROS) and high-mobility group box 1 (HMGB1) levels, and suppressed the inflammatory response in a SAP mouse model. The HPLC analysis of DCQD constituents indicated eight components (aloe-emodin, rhein, emodin, chrysophanol, naringin, hesperidin, magnolol, and honokiol) with the capacity to modulate ferroptosis. Venn analysis identified 48 DSNFGs, with a subset of five genes demonstrating significant predictive value. The developed nomogram, based on LASSO regression, showed high accuracy in validation cohorts. Single-cell RNA sequencing (scRNA-seq) and CellChat analysis uncovered heterogeneity and cell-cell communication networks in the pancreas during recovery from pancreatitis, implicating several signaling pathways. CONCLUSION DCQD and its eight ingredients exert its protective effect in SAP by inhibiting ferroptosis through the NOX2/GPX4 pathway. The DCQD-SAP-ferroptosis-related signature and nomogram offer a novel tool for AP risk assessment, prognosis prediction, and personalized therapeutic strategies in SAP management.
Collapse
Affiliation(s)
- Jian Chen
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Department of Public Health, International College, Krirk University, Bangkok, Thailand
| | - Fu Li
- Department of Hepatopancreatobiliary Surgery, Shuguang Hospital affliated to Shanghai University of Traditional Chinese Medincine, Shanghai 201203, China
| | - Wang-Sheng Luo
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421200, China
| | - Mei-Fang Zhu
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Neng-Jiang Zhao
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zhi-Hai Zhang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Ya-Feng Chen
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Dian-Xu Feng
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| | - Shu-Yu Yang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China.
| | - Wen-Jie Sun
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China; Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
2
|
Li T, Wang N, Yi D, Xiao Y, Li X, Shao B, Wu Z, Bai J, Shi X, Wu C, Qiu T, Yang G, Sun X, Zhang R. ROS-mediated ferroptosis and pyroptosis in cardiomyocytes: An update. Life Sci 2025; 370:123565. [PMID: 40113077 DOI: 10.1016/j.lfs.2025.123565] [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: 01/25/2025] [Revised: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
The cardiomyocyte is an essential component of the heart, communicating and coordinating with non-cardiomyocytes (endothelial cells, fibroblasts, and immune cells), and are critical for the regulation of structural deformation, electrical conduction, and contractile properties of healthy and remodeled myocardium. Reactive oxygen species (ROS) in cardiomyocytes are mainly produced by the mitochondrial oxidative respiratory chain, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), xanthine oxidoreductase (XOR), monoamine oxidase (MAO), and p66shc. Under physiological conditions, ROS are involved in the regulation of cardiac development and cardiomyocyte maturation, cardiac calcium handling, and excitation-contraction coupling. In contrast, dysregulation of ROS metabolism is involved in the development and progression of cardiovascular diseases (CVDs), including myocardial hypertrophy, hyperlipidemia, myocardial ischemia/reperfusion injury, arrhythmias and diabetic cardiomyopathy. Further oxidative stress induced by ROS dyshomeostasis was found to be the major reason for cardiomyocyte death in cardiac diseases, and in recent years, ferroptosis induced by oxidative stress have been considered to be fatal to cardiomyocytes. In addition, ROS is also a key trigger for the activation of pyroptosis, which induces and exacerbates the inflammatory response caused by various cardiac diseases and plays a critical role in CVDs. Therefore, in this review, the sources and destinations of ROS in cardiomyocytes will be systematically addressed, so as to reveal the molecular mechanisms by which ROS accumulation triggers cardiomyocyte ferroptosis and pyroptosis under pathological conditions, and provide a new concept for the research and treatment of heart-related diseases.
Collapse
Affiliation(s)
- Tao Li
- Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, PR China
| | - Ningning Wang
- Experimental Teaching Center of Public Health, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China; Global Health Research Center, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Dongxin Yi
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Yuji Xiao
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China; Bishan Hospital of Chongqing Medical University, Chongqing 402760, PR China
| | - Xiao Li
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Bing Shao
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Ziyi Wu
- School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Jie Bai
- Experimental Teaching Center of Public Health, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Xiaoxia Shi
- Experimental Teaching Center of Public Health, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Chenbing Wu
- Experimental Teaching Center of Public Health, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Tianming Qiu
- Global Health Research Center, Dalian Medical University, Dalian, Liaoning 116044, PR China; Occupational and Environmental Health Department, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Guang Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Xiance Sun
- Global Health Research Center, Dalian Medical University, Dalian, Liaoning 116044, PR China; Occupational and Environmental Health Department, School of Public Health, Dalian Medical University, Dalian, Liaoning 116044, PR China
| | - Rongfeng Zhang
- Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, PR China.
| |
Collapse
|
3
|
Famurewa AC, Akhigbe RE, George MY, Adekunle YA, Oyedokun PA, Akhigbe TM, Fatokun AA. Mechanisms of ferroptotic and non-ferroptotic organ toxicity of chemotherapy: protective and therapeutic effects of ginger, 6-gingerol and zingerone in preclinical studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4747-4778. [PMID: 39636404 PMCID: PMC11985630 DOI: 10.1007/s00210-024-03623-5] [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: 10/04/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Chemotherapy (CT) is one of the flagship options for the treatment of cancers worldwide. It involves the use of cytotoxic anticancer agents to kill or inhibit the proliferation of cancer cells. However, despite its clinical efficacy, CT triggers side effect toxicities in several organs, which may impact cancer patient's quality of life and treatment outcomes. While the side effect toxicity is consistent with non-ferroptotic mechanisms involving oxidative stress, inflammation, mitochondrial impairment and other aberrant signalling leading to apoptosis and necroptosis, recent studies show that ferroptosis, a non-apoptotic, iron-dependent cell death pathway, is also involved in the pathophysiology of CT organ toxicity. CT provokes organ ferroptosis via system Xc-/GPX-4/GSH/SLC7A11 axis depletion, ferritinophagy, iron overload, lipid peroxidation and upregulation of ferritin-related proteins. Cisplatin (CP) and doxorubicin (DOX) are common CT drugs indicated to induce ferroptosis in vitro and in vivo. Studies have explored natural preventive and therapeutic strategies using ginger rhizome and its major bioactive compounds, 6-gingerol (6G) and zingerone (ZG), to combat mechanisms of CT side effect toxicity. Ginger extract, 6G and ZG mitigate non-ferroptotic oxidative inflammation, apoptosis and mitochondrial dysfunction mechanisms of CT side effect toxicity, but their effects on CT-induced ferroptosis remain unclear. Systematic investigations are, therefore, needed to unfold the roles of ginger, 6G and ZG on ferroptosis involved in CT side effect toxicity, as they are potential natural agents for the prevention of CT toxicity. This review reveals the ferroptotic and non-ferroptotic toxicity mechanisms of CT and the protective mechanisms of ginger, 6G and ZG against CT-induced, ferroptotic and non-ferroptotic organ toxicities.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike, Ikwo, Nigeria.
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
| | - Roland E Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Yemi A Adekunle
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Precious A Oyedokun
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Tunmise M Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Breeding and Genetics Unit, Department of Agronomy, Osun State University, Osogbo, Osun State, Nigeria
| | - Amos A Fatokun
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| |
Collapse
|
4
|
Zhang L, Wang S, Zhang Y, Zhang X, Xi J, Wu J, Fang J, Zhao H, Zhang B. Troglitazone as a Novel Nrf2 Activator to Attenuate Oxidative Stress and Exert Neuroprotection. ACS Chem Neurosci 2025; 16:1604-1616. [PMID: 40135498 DOI: 10.1021/acschemneuro.5c00163] [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] [Indexed: 03/27/2025] Open
Abstract
Nuclear factor erythroid 2 related factor 2 (Nrf2) is closely associated with neurodegenerative diseases, and the Nrf2-mediated activation of antioxidant response elements (AREs) brings about validated strategies for treating neurodegenerative diseases. Here, we discovered that troglitazone, a clinical medication for diabetes mellitus, could serve as a Nrf2 activator to rescue neuronal damages both in vitro and in vivo. The mechanism of troglitazone action involves binding with kelch-like ECH-associated protein 1 (Keap1) and the activation of Nrf2. This process leads to the migration of Nrf2 to the cell nucleus and transactivates the AREs. Troglitazone exhibits significant alleviation of oxidative stress in PC12 cells induced by hydrogen peroxide or 6-hydroxydopamine (6-OHDA). In vivo studies indicate that troglitazone could rescue the motor activity and neurodevelopmental deficiency in zebrafish induced by 6-OHDA. Additionally, mass spectrometry imaging demonstrates that troglitazone could cross the zebrafish blood-brain barrier, supporting the application of troglitazone in treating neurodegenerative diseases. Overall, this work reveals that the novel Nrf2 activator troglitazone has potential therapeutic value for neurodegeneration and provides a foundation for its repurposing.
Collapse
Affiliation(s)
- Linjie Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Shuang Wang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Yanxia Zhang
- Center of Analysis and Testing, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xiaopeng Zhang
- School of Information Science and Engineering, Lanzhou University, Lanzhou 730000, China
| | - Junmin Xi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jun Wu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Haiyu Zhao
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
5
|
Liu X, Feng J, Guo M, Chen C, Zhao T, Sun X, Zhang Y. Resetting the aging clock through epigenetic reprogramming: Insights from natural products. Pharmacol Ther 2025; 270:108850. [PMID: 40221101 DOI: 10.1016/j.pharmthera.2025.108850] [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: 05/01/2024] [Revised: 12/04/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Epigenetic modifications play a critical role in regulating gene expression under various physiological and pathological conditions. Epigenetic modifications reprogramming is a recognized hallmark of aging and a key component of the aging clock used to differentiate between chronological and biological age. The potential for prospective diagnosis and regulatory capabilities position epigenetic modifications as an emerging drug target to extend longevity and alleviate age-related organ dysfunctions. In the past few decades, numerous preclinical studies have demonstrated the therapeutic potential of natural products in various human diseases, including aging, with some advancing to clinical trials and clinical application. This review highlights the discovery and recent advancements in the aging clock, as well as the potential use of natural products as anti-aging therapeutics by correcting disordered epigenetic reprogramming. Specifically, the focus is on the imbalance of histone modifications, alterations in DNA methylation patterns, disrupted ATP-dependent chromatin remodeling, and changes in RNA modifications. By exploring these areas, new insights can be gained into aging prediction and anti-aging interventions.
Collapse
Affiliation(s)
- Xin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Jing Feng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Madi Guo
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Chen Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Tong Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Xiuxiu Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Yong Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology, College of Pharmacy, and Department of Cardiology, the Second Affiliated Hospital, Harbin Medical University, Harbin 150081, China; State Key Laboratory -Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China.
| |
Collapse
|
6
|
Yan D, Wei Y, Ye X, Chen M, Wen S, Yao Z, Li R, Gao F, Zheng C, Gao H, You J. Colon-Targeted Hydrogel Microsphere System Encapsulating Oleic Acid-Emodin for Crohn's Disease Treatment via Ferroptosis Inhibition. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 39985760 DOI: 10.1021/acsami.4c22525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2025]
Abstract
Crohn's disease (CD) is a relapsing, systemic inflammatory disease that primarily affects the gastrointestinal tract and is often accompanied by extraintestinal manifestations and associated immune disorders. However, current pharmacological treatments for CD encounter several challenges, such as a lack of precise drug targeting and inadequate retention of drugs in the inflamed colon, along with low bioavailability. Herein, we utilized oleic acid (OA) as a solvent to enhance the bioavailability and solubility of emodin. Simultaneously, we encapsulated OA-emodin (OAE) into hydrogel microspheres (HMs) composed of hyaluronic acid (HA) and calcium alginate (CA) to develop a colon-targeted drug delivery system (HM@OAE) for CD therapy. The pH responsiveness of CA enabled HM@OAE to bypass the stomach and specifically target the colon, where it released OAE following oral administration. In addition, in vitro studies demonstrated that HM@OAE significantly reduced the secretion of proinflammatory cytokines, decreased reactive oxygen species levels, and restrained ferroptosis by upregulating GPX4 and SLC7A11 expression while downregulating ACSL4 expression. Furthermore, to confirm these findings in a live organism, an in vivo study was conducted using a dextran sulfate sodium-induced colitis mouse model. This study validated the therapeutic efficacy of HM@OAE, significantly alleviating colonic inflammation and restoring intestinal epithelial integrity. These results suggest that HM@OAE is a promising clinical candidate for CD treatment.
Collapse
Affiliation(s)
- Danxi Yan
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Yingqi Wei
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xijie Ye
- Department of Anorectal, Dongguan Hospital of Integrated Chinese and Western Medicine, Dongguan 523820, China
| | - Mingxia Chen
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Shuyi Wen
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Zhongxuan Yao
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Renkai Li
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Fei Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Chao Zheng
- The Affiliated Dongguan Songshan Lake Central Hospital of Guangdong Medical University, Dongguan 523808, China
| | - Huichang Gao
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Jieshu You
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| |
Collapse
|
7
|
Hao J, Li X, Shi S, Zhang H, Zhu H, Wu J, Gao M, Zhang B. Application of a near-infrared viscosity-responsive fluorescent probe for lysosomal targeting in fatty liver mice. Bioorg Chem 2025; 155:108162. [PMID: 39823757 DOI: 10.1016/j.bioorg.2025.108162] [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: 11/26/2024] [Revised: 01/01/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Viscosity is a fundamental property in biological systems, influencing organelle function and molecular diffusion. Abnormal viscosity is associated with diseases such as metabolic disorders, neurodegeneration, and cancer. Lysosomes, central to cellular degradation and recycling, are sensitive to viscosity changes, which can disrupt enzymatic activity and cellular homeostasis. Monitoring lysosomal viscosity provides essential information on lysosomal health, helping to uncover underlying mechanisms in various diseases. Recognizing the need for effective monitoring of lysosomal viscosity changes in living cells, we have developed a near-infrared (NIR) viscosity-responsive fluorescent probe, VFLyso, specifically designed for lysosomal targeting. Based on the twisted intramolecular charge transfer (TICT) mechanism, VFLyso exhibits strong NIR fluorescence, a fast response, and a notable fluorescence response to viscosity variations (F/F0 = 65.5-fold), along with excellent selectivity and stability under physiological conditions. Our studies demonstrated that VFLyso could accurately monitor lysosomal viscosity changes in both cell cultures and animal models, including zebrafish and mouse models of fatty liver. This work not only provides a powerful tool for real-time monitoring of lysosomal viscosity but also offers valuable insights into the role of viscosity in disease progression, paving the way for potential diagnostic applications in related disorders.
Collapse
Affiliation(s)
- Junlei Hao
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai Minzu University, Xining 810007 Qinghai, China; State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xiao Li
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai Minzu University, Xining 810007 Qinghai, China
| | - Suntao Shi
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Haijuan Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Hailiang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jiang Wu
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai Minzu University, Xining 810007 Qinghai, China.
| | - Mingyong Gao
- The Third Clinical Institute Affiliated to Wenzhou Medical University/Wenzhou People's Hospital/The Third Affiliated Hospital of Shanghai University, Wenzhou, China.
| | - Baoxin Zhang
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, College of Pharmacy, Qinghai Minzu University, Xining 810007 Qinghai, China; State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
8
|
Famurewa AC, Prabhune NM, Prabhu S. Natural product mitigation of ferroptosis in platinum-based chemotherapy toxicity: targeting the underpinning oxidative signaling pathways. J Pharm Pharmacol 2025; 77:1-17. [PMID: 39485898 DOI: 10.1093/jpp/rgae132] [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: 04/21/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVES Platinum-based anticancer chemotherapy (PAC) represents a cornerstone in cancer treatment, retaining its status as the gold standard therapy. However, PAC's efficacy is countered by significant toxicities, such as nephrotoxicity, ototoxicity, and neurotoxicity. Recent studies have linked these toxicities to ferroptosis, characterized by iron accumulation, reactive oxygen species generation, and lipid peroxidation. This review explores the mechanisms underlying PAC-induced toxicities, focusing on the involvement of ferroptosis with three major PAC drugs-cisplatin, carboplatin, and oxaliplatin. Further, we provide a comprehensive analysis of the natural product mitigation of PAC-induced ferroptotic toxicity. KEY FINDINGS The mechanistic role of ferroptosis in cisplatin- and oxaliplatin-induced toxicities has been investigated, while studies on carboplatin-induced ferroptotic toxicities are lacking. Natural compounds targeting molecular pathways of ferroptosis have been explored to mitigate PAC-induced ferroptotic toxicity. CONCLUSION While ferroptosis in cisplatin- and oxaliplatin-induced toxicities has been investigated, there remains a notable dearth of studies examining its involvement in carboplatin-induced toxicities. Hence, further exploration is warranted to define the role of ferroptosis in carboplatin-induced toxicities, and its further mitigation. Moreover, in-depth mechanistic evaluation is necessary to establish natural products evaluated against PAC-induced ferroptosis, as PAC adjuvants.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ikwo 482103, Ebonyi State, Nigeria
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Nupura Manish Prabhune
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sudharshan Prabhu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
9
|
Tayal R, Mannan A, Singh S, Dhiman S, Singh TG. Unveiling the Complexities: Exploring Mechanisms of Anthracyclineinduced Cardiotoxicity. Curr Cardiol Rev 2025; 21:42-77. [PMID: 39484769 DOI: 10.2174/011573403x322928241021100631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 11/03/2024] Open
Abstract
The coexistence of cancer and heart disease, both prominent causes of illness and death, is further exacerbated by the detrimental impact of chemotherapy. Anthracycline-induced cardiotoxicity is an unfortunate side effect of highly effective therapy in treating different types of cancer; it presents a significant challenge for both clinicians and patients due to the considerable risk of cardiotoxicity. Despite significant progress in understanding these mechanisms, challenges persist in identifying effective preventive and therapeutic strategies, rendering it a subject of continued research even after three decades of intensive global investigation. The molecular targets and signaling pathways explored provide insights for developing targeted therapies, emphasizing the need for continued research to bridge the gap between preclinical understanding and clinical applications. This review provides a comprehensive exploration of the intricate mechanisms underlying anthracycline-induced cardiotoxicity, elucidating the interplay of various signaling pathways leading to adverse cellular events, including cardiotoxicity and death. It highlights the extensive involvement of pathways associated with oxidative stress, inflammation, apoptosis, and cellular stress responses, offering insights into potential and unexplored targets for therapeutic intervention in mitigating anthracycline-induced cardiac complications. A comprehensive understanding of the interplay between anthracyclines and these complexes signaling pathways is crucial for developing strategies to prevent or mitigate the associated cardiotoxicity. Further research is needed to outline the specific contributions of these pathways and identify potential therapeutic targets to improve the safety and efficacy of anthracycline-based cancer treatment. Ultimately, advancements in understanding anthracycline-induced cardiotoxicity mechanisms will facilitate the development of more efficacious preventive and treatment approaches, thereby improving outcomes for cancer patients undergoing anthracycline-based chemotherapy.
Collapse
Affiliation(s)
- Rohit Tayal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | |
Collapse
|
10
|
Ouyang F, Li Y, Wang H, Liu X, Tan X, Xie G, Zeng J, Zeng G, Luo Q, Zhou H, Chen S, Hou K, Fang J, Zhang X, Zhou L, Li Y, Gao A. Aloe Emodin Alleviates Radiation-Induced Heart Disease via Blocking P4HB Lactylation and Mitigating Kynurenine Metabolic Disruption. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406026. [PMID: 39494721 PMCID: PMC11653682 DOI: 10.1002/advs.202406026] [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: 06/01/2024] [Revised: 09/22/2024] [Indexed: 11/05/2024]
Abstract
Aloe emodin is an anthraquinone of traditional Chinese medicine monomer, which plays a protective action in cardiovascular diseases. However, the regulatory mechanisms of aloe emodin in the protection of radiation-induced heart damage (RIHD) are unclear. As a novel post-translational modification, lactylation is considered as a critical mediator in inflammatory cascade and cardiac injury. Here, using a cross of differential omics and 4D label-free lactylation omics, protein disulfide-isomerase (P4HB) is identified as a novel target for lactylation, and aloe emodin inhibits the binding of lactate to the K311 site of P4HB. Aloe emodin stabilizes kynurenine metabolism through inhibition of aspartate aminotransferase (GOT2) accumulation on damaged mitochondria. Mechanistically, aloe emodin inhibits phosphorylated glycogen synthase kinase 3B (p-GSK3B) transcription in the nucleus to repress the interaction of prostaglandin G/H synthase 2 (PTGS2) with SH3 domain of SH3 domain-containing GRB2-like protein B1 (SH3GLB1), thereby disrupting the functions of mitochondrial complexes and reducing SH3GLB1-mediated mitoROS accumulation, eventually suppressing calcium-binding and coiled-coil domain-containing protein 2 (NDP52)-induced mitophagy. This study unveils the regulatory role of aloe emodin in RIHD alleviation through PTGS2/SH3GLB1/NDP52 axis, indicates aloe emodin stabilizes GOT2-mediated kynurenine metabolism through P4HB lactylation. Collectively, this study provides novel insights into the regulatory mechanisms underlying the protective role of aloe emodin in cardiac injury, and opens new avenues for therapeutic strategies of aloe emodin in preventing RIHD by regulating lactylation.
Collapse
Affiliation(s)
- Fan Ouyang
- Department of Cardiovascular MedicineZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Yaling Li
- Department of Cardiovascular MedicineZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Haoming Wang
- Department of Cardiovascular MedicineZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Xiangyang Liu
- Department of Cardiovascular MedicineZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Xiaoli Tan
- Zhuzhou Clinical CollegeJishou UniversityJishouHunan416000P. R. China
| | - Genyuan Xie
- Zhuzhou Clinical CollegeJishou UniversityJishouHunan416000P. R. China
| | - Junfa Zeng
- Department of Critical Care MedicineHengyang Medical SchoolThe Second Affiliated HospitalUniversity of South ChinaHengyangHunan421001P. R. China
| | - Gaofeng Zeng
- Clinical Research InstituteThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Department of Assisted Reproductive CentreZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
| | - Qiong Luo
- Clinical Research Center for Arteriosclerotic Disease in Hunan ProvinceHengyangHunan421001P. R. China
| | - Hong Zhou
- Department of RadiologyHengyang Medical SchoolThe First Affiliated HospitalUniversity of South ChinaHengyangHunan421001P. R. China
| | - Siming Chen
- Clinical Research Center for Arteriosclerotic Disease in Hunan ProvinceHengyangHunan421001P. R. China
| | - Kai Hou
- Department of Cardiovascular MedicineThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Jinren Fang
- Department of Cardiovascular MedicineThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Xia Zhang
- Department of Ultrasound MedicineHengyang Medical SchoolThe Second Affiliated HospitalUniversity of South ChinaHengyangHunan421001P. R. China
| | - Linlin Zhou
- Zhuzhou Clinical CollegeJishou UniversityJishouHunan416000P. R. China
| | - Yukun Li
- Department of Assisted Reproductive CentreZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
- Department of Cardiovascular MedicineThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Anbo Gao
- Clinical Research InstituteThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Department of Assisted Reproductive CentreZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunan412000P. R. China
- Department of Cardiovascular MedicineThe Second Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal CancerHengyangHunan421001P. R. China
| |
Collapse
|
11
|
Famurewa AC, George MY, Ukwubile CA, Kumar S, Kamal MV, Belle VS, Othman EM, Pai SRK. Trace elements and metal nanoparticles: mechanistic approaches to mitigating chemotherapy-induced toxicity-a review of literature evidence. Biometals 2024; 37:1325-1378. [PMID: 39347848 DOI: 10.1007/s10534-024-00637-7] [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] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
Anticancer chemotherapy (ACT) remains a cornerstone in cancer treatment, despite significant advances in pharmacology over recent decades. However, its associated side effect toxicity continues to pose a major concern for both oncology clinicians and patients, significantly impacting treatment protocols and patient quality of life. Current clinical strategies to mitigate ACT-induced toxicity have proven largely unsatisfactory, leaving a critical unmet need to block toxicity mechanisms without diminishing ACT's therapeutic efficacy. This review aims to document the molecular mechanisms underlying ACT toxicity and highlight research efforts exploring the protective effects of trace elements (TEs) and their nanoparticles (NPs) against these mechanisms. Our literature review reveals that the primary driver of ACT toxicity is redox imbalance, which triggers oxidative inflammation, apoptosis, endoplasmic reticulum stress, mitochondrial dysfunction, autophagy, and dysregulation of signaling pathways such as PI3K/mTOR/Akt. Studies suggest that TEs, including zinc, selenium, boron, manganese, and molybdenum, and their NPs, can potentially counteract ACT-induced toxicity by inhibiting oxidative stress-mediated pathways, including NF-κB/TLR4/MAPK/NLRP3, STAT-3/NLRP3, Bcl-2/Bid/p53/caspases, and LC3/Beclin-1/CHOP/ATG6, while also upregulating protective signaling pathways like Sirt1/PPAR-γ/PGC-1α/FOXO-3 and Nrf2/HO-1/ARE. However, evidence regarding the roles of lncRNA and the Wnt/β-catenin pathway in ACT toxicity remains inconsistent, and the impact of TEs and NPs on ACT efficacy is not fully understood. Further research is needed to confirm the protective effects of TEs and their NPs against ACT toxicity in cancer patients. In summary, TEs and their NPs present a promising avenue as adjuvant agents for preventing non-target organ toxicity induced by ACT.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike Ikwo, Abakaliki, Ebonyi, Nigeria.
- Centre for Natural Products Discovery, School of P harmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Cletus A Ukwubile
- Department of Pharmacognosy, Faculty of Pharmacy, University of Maiduguri, Bama Road, Maiduguri, Borno, Nigeria
| | - Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mehta V Kamal
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vijetha S Belle
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Eman M Othman
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
- Cancer Therapy Research Center, Department of Biochemistry-I, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
- Department of Bioinformatics, University of Würzburg, Am Hubland, 97074, BiocenterWürzburg, Germany
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
12
|
Tan M, Yin Y, Chen W, Zhang J, Jin Y, Zhang Y, Zhang L, Jiang T, Jiang B, Li H. Trimetazidine attenuates Ischemia/Reperfusion-Induced myocardial ferroptosis by modulating the Sirt3/Nrf2-GSH system and reducing Oxidative/Nitrative stress. Biochem Pharmacol 2024; 229:116479. [PMID: 39134283 DOI: 10.1016/j.bcp.2024.116479] [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: 03/05/2024] [Revised: 07/11/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024]
Abstract
Ferroptosis is a newly defined mode of cellular demise. The increasing investigation supports that ferroptosis is a crucial factor in the complex mechanisms of myocardial ischemia-reperfusion (I/R) injury. Hence, targeting ferroptosis is a novel strategy for treating myocardial injury. Although evidence suggests that trimetazidine (TMZ) is potentially efficacious against myocardial injury, the exact mechanism of this efficacy is yet to be fully elucidated. This study aimed to determine whether TMZ can act as a ferroptosis resistor and affect I/R-mediated myocardial injury. To this end, researchers have constructed in vitro and in vivo models of I/R using H9C2 cardiomyocytes, primary cardiomyocytes, and SD rats. Here, I/R mediated the onset of ferroptosis in vitro and in vivo, as reflected by excessive iron aggregation, GSH depletion, and the increase in lipid peroxidation. TMZ largely reversed this alteration and attenuated cardiomyocyte injury. Mechanistically, we found that TMZ upregulated the expression of Sirt3. Therefore, we used si-Sirt3 and 3-TYP to interfere with Sirt3 action in vitro and in vivo, respectively. Both si-Sirt3 and 3-TYP partly mitigated the inhibitory effect of TMZ on I/R-mediated ferroptosis and upregulated the expression of Nrf2 and its downstream target, GPX4-SLC7A11. These results indicate that TMZ attenuates I/R-mediated ferroptosis by activating the Sirt3-Nrf2/GPX4/SLC7A11 signaling pathway. Our study offers insights into the mechanism underlying the cardioprotective benefits of TMZ and establishes a groundwork for expanding its potential applications.
Collapse
Affiliation(s)
- Mingyue Tan
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Yunfei Yin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Weixiang Chen
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Jun Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Yifeng Jin
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Yue Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Lei Zhang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| | - Bin Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| | - Hongxia Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
13
|
Li Y, Yan J, Yang P. The mechanism and therapeutic strategies in doxorubicin-induced cardiotoxicity: Role of programmed cell death. Cell Stress Chaperones 2024; 29:666-680. [PMID: 39343295 PMCID: PMC11490929 DOI: 10.1016/j.cstres.2024.09.001] [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: 07/15/2024] [Revised: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024] Open
Abstract
Doxorubicin (DOX) is the most commonly used anthracycline anticancer agent, while its clinical utility is limited by harmful side effects like cardiotoxicity. Numerous studies have elucidated that programmed cell death plays a significant role in DOX-induced cardiotoxicity (DIC). This review summarizes several kinds of programmed cell death, including apoptosis, pyroptosis, necroptosis, autophagy, and ferroptosis. Furthermore, oxidative stress, inflammation, and mitochondrial dysfunction are also important factors in the molecular mechanisms of DIC. Besides, a comprehensive understanding of specific signal pathways of DIC can be helpful to its treatment. Therefore, the related signal pathways are elucidated in this review, including sirtuin deacetylase (silent information regulator 2 [Sir2]) 1 (SIRT1)/nuclear factor erythroid 2-related factor 2, SIRT1/Klotho, SIRT1/Recombinant Sestrin 2, adenosine monophosphate-activated protein kinase, AKT, and peroxisome proliferator-activated receptor. Heat shock proteins function as chaperones, which play an important role in various stressful situations, especially in the heart. Thus, some of heat shock proteins involved in DIC are also included. Hence, the last part of this review focuses on the therapeutic research based on the mechanisms above.
Collapse
Affiliation(s)
- Yanzhao Li
- Department of Second Clinical Medical College, Southern Medical University, Guangzhou, China.
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Wu J, Ding Z, Tu J, Osama A, Nie Q, Cai W, Zhang B. Unveiling the anticancer potential of plumbagin: targeting pyruvate kinase M2 to induce oxidative stress and apoptosis in hepatoma cells. RSC Med Chem 2024:d4md00519h. [PMID: 39363929 PMCID: PMC11446330 DOI: 10.1039/d4md00519h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
Pyruvate kinase M2 (PKM2), a crucial enzyme in the glycolysis pathway, is commonly documented as being overexpressed in cancer cells. Inhibiting PKM2, a strategy to mitigate cancer cell-dependent glycolysis, has demonstrated efficacy in anticancer treatment. In this study, plumbagin, which was originally extracted from the plant Plumbago zeylanica L., was discovered as a novel PKM2 inhibitor and it could bind to PKM2 to inhibit the enzymatic activity. Treatment with plumbagin in HepG2 cells resulted in the decrease of PKM2 expression, which in turn reduced the protein kinase function. The mRNA levels of its downstream genes, such as LDHA and MYC, were suppressed. Additionally, plumbagin downregulated the expression of intracellular antioxidant proteins, which induced oxidative stress and mitochondrial damage, ultimately triggering apoptosis. Moreover, plumbagin also reduced the migration and proliferation of HepG2 cells. This study offered valuable insights into the molecular mechanism of plumbagin and advocated for the exploration of PKM2 inhibitors as viable possibilities for anticancer therapeutics.
Collapse
Affiliation(s)
- Jun Wu
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University Lanzhou 730000 China
| | - Zhenjiang Ding
- Beijing Key Laboratory of the Innovative Development of Functional Staple and Nutritional Intervention for Chronic Diseases, China National Research Institute of Food and Fermentation Industries Beijing 100015 China
| | - Jingwen Tu
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University Lanzhou 730000 China
| | - Alsiddig Osama
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University Lanzhou 730000 China
| | - Qiuying Nie
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University Lanzhou 730000 China
| | - Wenqing Cai
- Regor Therapeutics Inc 1206 Zhangjiang Road, Building C, Pu Dong New District Shanghai 201210 China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University Lanzhou 730000 China
| |
Collapse
|
15
|
Lu LQ, Li MR, Huang LL, Che YX, Qi YN, Luo XJ, Peng J. Micafungin protects mouse heart against doxorubicin-induced oxidative injury via suppressing MALT1-dependent k48-linked ubiquitination of Nrf2. Chem Biol Interact 2024; 400:111179. [PMID: 39089415 DOI: 10.1016/j.cbi.2024.111179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Oxidative stress contributes greatly to doxorubicin (DOX)-induced cardiotoxicity. Down-regulation of nuclear factor erythroid 2-related factor 2 (Nrf2) is a key factor in DOX-induced myocardial oxidative injury. Recently, we found that mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1)-dependent k48-linked ubiquitination was responsible for down-regulation of myocardial Nrf2 in DOX-treated mice. Micafungin, an antifungal drug, was identified as a potential MALT1 inhibitor. This study aims to explore whether micafungin can reduce DOX-induced myocardial oxidative injury and if its anti-oxidative effect involves a suppression of MALT1-dependent k48-linked ubiquitination of Nrf2. To establish the cardiotoxicity models in vivo and in vitro, mice were treated with a single dose of DOX (15 mg/kg, i.p.) and cardiomyocytes were incubated with DOX (1 μM) for 24 h, respectively. Using mouse model of DOX-induced cardiotoxicity, micafungin (10 or 20 mg/kg) was shown to improve cardiac function, concomitant with suppression of oxidative stress, mitochondrial dysfunction, and cell death in a dose-dependent manner. Similar protective roles of micafungin (1 or 5 μM) were observed in DOX-treated cardiomyocytes. Mechanistically, micafungin weakened the interaction between MALT1 and Nrf2, decreased the k48-linked ubiquitination of Nrf2 while elevated the protein levels of Nrf2 in both DOX-treated mice and cardiomyocytes. Furthermore, MALT1 overexpression counteracted the cardioprotective effects of micafungin. In conclusion, micafungin reduces DOX-induced myocardial oxidative injury via suppression of MALT1, which decreases the k48-linked ubiquitination of Nrf2 and elevates Nrf2 protein levels. Thus, micafungin may be repurposed for treating DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Li-Qun Lu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Lin-Lu Huang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Yan-Xi Che
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Ya-Nan Qi
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China; Hunan Provincial Key Laboratory of Cardiovascular Research, School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China.
| |
Collapse
|
16
|
Wen J, Li L, Yang Y, Ou D, Yang J, Xie J, Du W, Tong Y. Phytochemicals targeting ferroptosis in cardiovascular diseases: Recent advances and therapeutic perspectives. Phytother Res 2024; 38:4386-4405. [PMID: 38973263 DOI: 10.1002/ptr.8278] [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: 01/02/2024] [Revised: 06/03/2024] [Accepted: 06/08/2024] [Indexed: 07/09/2024]
Abstract
Ferroptosis is a form of iron-dependent regulatory cell death that is related to the pathogenesis and progression of various cardiovascular diseases, such as arrhythmia, diabetic cardiomyopathy, myocardial infarction, myocardial ischemia/reperfusion injury, and heart failure. This makes it a promising therapeutic target for cardiovascular diseases. It is interesting that a significant number of cardiovascular disease treatment drugs derived from phytochemicals have been shown to target ferroptosis, thus producing cardioprotective effects. This study offers a concise overview of the initiation and control mechanisms of ferroptosis. It discusses the core regulatory factors of ferroptosis as potential new therapeutic targets for various cardiovascular diseases, elucidating how ferroptosis influences the progression of cardiovascular diseases. In addition, this review systematically summarizes the regulatory effects of phytochemicals on ferroptosis, emphasizing their potential mechanisms and clinical applications in treating cardiovascular diseases. This study provides a reference for further elucidating the molecular mechanisms of phytochemicals in treating cardiovascular diseases. This may accelerate their application in the treatment of cardiovascular diseases and is worth further research in this field.
Collapse
Affiliation(s)
- Jianxia Wen
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Lu Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yi Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Dinglin Ou
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Junjie Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Jiachen Xie
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Wenya Du
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yuling Tong
- School of Medicine and Food, Sichuan Vocational College of Health and Rehabilitation, Zigong, China
| |
Collapse
|
17
|
Yi X, Wang Q, Zhang M, Shu Q, Zhu J. Ferroptosis: A novel therapeutic target of natural products against doxorubicin-induced cardiotoxicity. Biomed Pharmacother 2024; 178:117217. [PMID: 39079260 DOI: 10.1016/j.biopha.2024.117217] [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: 05/07/2024] [Revised: 07/13/2024] [Accepted: 07/26/2024] [Indexed: 08/25/2024] Open
Abstract
Doxorubicin (DOX), a commonly used chemotherapy drug, is hindered due to its tendency to induce cardiotoxicity (DIC). Ferroptosis, a novel mode of programmed cell death, has received substantial attention for its involvement in DIC. Recently, natural product-derived ferroptosis regulator emerged as a potential strategy for treating DIC. In this review, a comprehensive search was conducted across PubMed, Web of Science, Google Scholar, and ScienceDirect databases to gather relevant articles on the use of natural products for treating DIC in relation to ferroptosis. The available papers were carefully reviewed to summarize the therapeutic effects and underlying mechanisms of natural products in modulating ferroptosis for DIC treatment. It was found that ferroptosis plays an important role in DIC pathogenesis, with dysregulated expression of ferroptosis-related proteins strongly implicated in the condition. Natural products, such as flavonoids, polyphenols, terpenoids, and quinones can act as GPX4 activators, Nrf2 agonists, and lipid peroxidation inhibitors, thereby enhancing cell viability, attenuating myocardial fibrosis, improving cardiac function, and suppressing ferroptosis in both in vitro and in vivo models of DIC. This review demonstrates a strong correlation between DOX-induced cardiac ferroptosis and key proteins, such as GPX4, Keap1, Nrf2, AMPK, and HMOX1. Natural products are likely to exert therapeutic effects against DIC by modulating the activity of these proteins.
Collapse
Affiliation(s)
- Xiaojiao Yi
- Department of Pharmacy, Hangzhou Xixi Hospital, Hangzhou Sixth People's Hospital, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310023, China
| | - Qi Wang
- Department of Pharmacy, Hangzhou Xixi Hospital, Hangzhou Sixth People's Hospital, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310023, China
| | - Mengjie Zhang
- Department of Pharmacy, Hangzhou Xixi Hospital, Hangzhou Sixth People's Hospital, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310023, China
| | - Qi Shu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| | - Junfeng Zhu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| |
Collapse
|
18
|
Cai K, Jiang H, Zou Y, Song C, Cao K, Chen S, Wu Y, Zhang Z, Geng D, Zhang N, Liu B, Sun G, Tang M, Li Z, Zhang Y, Sun Y, Zhang Y. Programmed death of cardiomyocytes in cardiovascular disease and new therapeutic approaches. Pharmacol Res 2024; 206:107281. [PMID: 38942341 DOI: 10.1016/j.phrs.2024.107281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Cardiovascular diseases (CVDs) have a complex pathogenesis and pose a major threat to human health. Cardiomyocytes have a low regenerative capacity, and their death is a key factor in the morbidity and mortality of many CVDs. Cardiomyocyte death can be regulated by specific signaling pathways known as programmed cell death (PCD), including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, etc. Abnormalities in PCD can lead to the development of a variety of cardiovascular diseases, and there are also molecular-level interconnections between different PCD pathways under the same cardiovascular disease model. Currently, the link between programmed cell death in cardiomyocytes and cardiovascular disease is not fully understood. This review describes the molecular mechanisms of programmed death and the impact of cardiomyocyte death on cardiovascular disease development. Emphasis is placed on a summary of drugs and potential therapeutic approaches that can be used to treat cardiovascular disease by targeting and blocking programmed cell death in cardiomyocytes.
Collapse
Affiliation(s)
- Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Haoyue Jiang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China
| | - Bo Liu
- The first hospital of China Medical University, Department of cardiac surgery, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Man Tang
- Department of clinical pharmacology, College of Pharmacy, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| |
Collapse
|
19
|
Wang X, Lin Z, Tang X, Xie M, Li T, Zhou J. Matrine induces cardiotoxicity by promoting ferroptosis through the Nrf2 antioxidant system in H9c2 cells. Toxicol Lett 2024; 397:11-22. [PMID: 38723914 DOI: 10.1016/j.toxlet.2024.05.001] [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: 02/03/2024] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
Matrine (MT) has shown promising efficacy in various cancers and chronic hepatitis; however, its clinical application is limited because of its side effects. Our previous studies have indicated that MT can induce severe hepatotoxicity and nephrotoxicity. The current study aimed to investigate its cardiotoxicity and potential underlying mechanisms in H9c2 cells. Our results showed that MT induced H9c2 cell death and disrupted the cellular membrane integrity. Moreover, MT decreased glutathione (GSH) and cysteine (Cys) levels, and increased Fe2+, lipid peroxidation, reactive oxygen species (ROS), and MDA levels, ultimately leading to ferroptosis. Interestingly, these phenomena were alleviated by the ferroptosis inhibitor Fer-1, whereas MT-induced ferroptosis was exacerbated by the ferroptosis agonist RSL3. In addition, MT significantly reduced FTH, Nrf2, xCT, GPX4, and FSP1 protein levels and inhibited the transcriptional activity of Nrf2 while increasing TFR1 protein levels. Supplementation with Nrf2 agonist (Dimethyl fumarate, DMF) or selenium (Sodium selenite, SS) and CoQ10 alleviated MT-induced cytotoxic effects in H9c2 cells. These results suggest that ferroptosis, which is mediated by an imbalance in the Nrf2 antioxidant system, is involved in MT-induced cardiac toxicity.
Collapse
Affiliation(s)
- Xi Wang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Zixiong Lin
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Xinyi Tang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Minjuan Xie
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Ting Li
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun 336000, PR China.
| |
Collapse
|
20
|
Zhang L, Luo YL, Xiang Y, Bai XY, Qiang RR, Zhang X, Yang YL, Liu XL. Ferroptosis inhibitors: past, present and future. Front Pharmacol 2024; 15:1407335. [PMID: 38846099 PMCID: PMC11153831 DOI: 10.3389/fphar.2024.1407335] [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: 03/26/2024] [Accepted: 05/06/2024] [Indexed: 06/09/2024] Open
Abstract
Ferroptosis is a non-apoptotic mode of programmed cell death characterized by iron dependence and lipid peroxidation. Since the ferroptosis was proposed, researchers have revealed the mechanisms of its formation and continue to explore effective inhibitors of ferroptosis in disease. Recent studies have shown a correlation between ferroptosis and the pathological mechanisms of neurodegenerative diseases, as well as diseases involving tissue or organ damage. Acting on ferroptosis-related targets may provide new strategies for the treatment of ferroptosis-mediated diseases. This article specifically describes the metabolic pathways of ferroptosis and summarizes the reported mechanisms of action of natural and synthetic small molecule inhibitors of ferroptosis and their efficacy in disease. The paper also describes ferroptosis treatments such as gene therapy, cell therapy, and nanotechnology, and summarises the challenges encountered in the clinical translation of ferroptosis inhibitors. Finally, the relationship between ferroptosis and other modes of cell death is discussed, hopefully paving the way for future drug design and discovery.
Collapse
Affiliation(s)
- Lei Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yi Lin Luo
- School of Medicine, Yan’an University, Yan’an, China
| | - Yang Xiang
- College of Physical Education, Yan’an University, Yan’an, China
| | - Xin Yue Bai
- School of Medicine, Yan’an University, Yan’an, China
| | | | - Xin Zhang
- School of Medicine, Yan’an University, Yan’an, China
| | - Yan Ling Yang
- School of Medicine, Yan’an University, Yan’an, China
| | - Xiao Long Liu
- School of Medicine, Yan’an University, Yan’an, China
| |
Collapse
|
21
|
Liu J, Deng L, Qu L, Li X, Wang T, Chen Y, Jiang M, Zou W. Herbal medicines provide regulation against iron overload in cardiovascular diseases: Informing future applications. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117941. [PMID: 38387684 DOI: 10.1016/j.jep.2024.117941] [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: 03/27/2023] [Revised: 02/04/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Iron is an essential micronutrient for maintaining physiological activities, especially for highly active cardiomyocytes. Inappropriate iron overload or deficiency has a significant impact on the incidence and severity of cardiovascular diseases (CVD). Iron overload exerts potentially deleterious effects on doxorubicin (DOX) cardiomyopathy, atherosclerosis, and myocardial ischemia-reperfusion injury (MI/RI) by participating in lipid peroxides production. Notably, iron overload-associated cell death has been defined as a possible mechanism for ferroptosis. At present, some traditional herbal medicines and extracts have been included in the study of regulating iron overload and the subsequent therapeutic effect on CVD. AIM OF THE STUDY To give an outline of iron metabolism and ferroptosis in cardiomyocytes and to focus on herbal medicines and extracts to prevent iron overload in CVD. MATERIALS AND METHODS Literature information was systematically collected from ScienceDirect, PubMed, Google Scholar, Web of Science, China National Knowledge Infrastructure, WanFang data, as well as classic books and clinical reports. RESULTS After understanding the mechanism of iron overload on CVD, this paper reviews the therapeutic function of various herbal medicines in eliminating iron overload in CVD. These include Chinese herbal compound prescriptions (Salvia miltiorrhiza injection, Gegen Qinlian decoction, Tongxinluo, Banxia-Houpu decoction), plant extracts, phenylpropanoids, flavonoids, terpenoids, and polyphenols. Among them, flavonoids are considered to be the most promising compounds because of their prominent iron chelation. Mechanically, these herbal medicines act on the Nrf2 signaling pathway, AMPK signaling pathway, and KAT5/GPX4 signaling pathway, thereby attenuating iron overload and lipid peroxidation in CVD. CONCLUSION Our review provides up-to-date information on herbal medicines that exert cardiovascular protective effects by modulating iron overload and ferroptosis. These herbal medicines hold promise as a template for preventing iron overload in CVD.
Collapse
Affiliation(s)
- Jia Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Liangyan Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Liping Qu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Xiaofen Li
- School of Basic Medicine Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Tao Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yuanyuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Miao Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Wenjun Zou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
22
|
Liu J, Liu H, Deng L, Wang T, Li L, Chen Y, Qu L, Zou W. Protective Role of Dioscin against Doxorubicin-Induced Chronic Cardiotoxicity: Insights from Nrf2-GPX4 Axis-Mediated Cardiac Ferroptosis. Biomolecules 2024; 14:422. [PMID: 38672439 PMCID: PMC11047995 DOI: 10.3390/biom14040422] [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: 02/07/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Recent evidence suggests that ferroptosis, an iron-facilitated cell death with excessive lipid peroxidation, is a critical mechanism underlying doxorubicin (DOX)-induced cardiotoxicity (DIC). Although dioscin has been reported to improve acute DIC, direct evidence is lacking to clarify the role of dioscin in chronic DIC and its potential mechanism in cardiac ferroptosis. In this study, we used chronic DIC rat models and H9c2 cells to investigate the potential of dioscin to mitigate DIC by inhibiting ferroptosis. Our results suggest that dioscin significantly improves chronic DIC-induced cardiac dysfunction. Meanwhile, it significantly inhibited DOX-induced ferroptosis by reducing Fe2+ and lipid peroxidation accumulation, maintaining mitochondrial integrity, increasing glutathione peroxidase 4 (GPX4) expression, and decreasing acyl-CoA synthetase long-chain family 4 (ACSL4) expression. Through transcriptomic analysis and subsequent validation, we found that the anti-ferroptotic effects of dioscin are achieved by regulating the nuclear factor-erythroid 2-related factor 2 (Nrf2)/GPX4 axis and Nrf2 downstream iron metabolism genes. Dioscin further downregulates nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4) and upregulates expression of frataxin (FXN) and ATP-binding cassette B8 (ABCB8) to limit mitochondrial Fe2+ and lipid peroxide accumulation. However, Nrf2 inhibition diminishes the anti-ferroptotic effects of dioscin, leading to decreased GPX4 expression and increased lipid peroxidation. This study is a compelling demonstration that dioscin can effectively reduce DIC by inhibiting ferroptosis, which is dependent on the Nrf2/GPX4 pathway modulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Liping Qu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (J.L.); (H.L.); (L.D.); (T.W.); (L.L.); (Y.C.)
| | - Wenjun Zou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (J.L.); (H.L.); (L.D.); (T.W.); (L.L.); (Y.C.)
| |
Collapse
|
23
|
Nie A, Shen C, Zhou Z, Wang J, Sun B, Zhu C. Ferroptosis: Potential opportunities for natural products in cancer therapy. Phytother Res 2024; 38:1173-1190. [PMID: 38116870 DOI: 10.1002/ptr.8088] [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: 08/30/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/21/2023]
Abstract
Cancer cells often exhibit defects in the execution of cell death, resulting in poor clinical outcomes for patients with many cancer types. Ferroptosis is a newly discovered form of programmed cell death characterized by intracellular iron overload and lipid peroxidation in the cell membrane. Increasing evidence suggests that ferroptosis is closely associated with a wide variety of physiological and pathological processes, particularly in cancer. Notably, various bioactive natural products have been shown to induce the initiation and execution of ferroptosis in cancer cells, thereby exerting anticancer effects. In this review, we summarize the core regulatory mechanisms of ferroptosis and the multifaceted roles of ferroptosis in cancer. Importantly, we focus on natural products that regulate ferroptosis in cancer cells, such as terpenoids, polyphenols, alkaloids, steroids, quinones, and polysaccharides. The clinical efficacy, adverse effects, and drug-drug interactions of these natural products need to be evaluated in further high-quality studies to accelerate their application in cancer treatment.
Collapse
Affiliation(s)
- Anzheng Nie
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaozan Shen
- Department of Clinical Pharmacy, The Second People's Hospital of Huaihua, Huaihua, China
| | - Zheng Zhou
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Juan Wang
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bao Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chunsheng Zhu
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Kim MS, Choi HK, Park SH, Lee JI, Lee J. Poncirus trifoliata Aqueous Extract Protects Cardiomyocytes against Doxorubicin-Induced Toxicity through Upregulation of NAD(P)H Dehydrogenase Quinone Acceptor Oxidoreductase 1. Molecules 2023; 28:8090. [PMID: 38138580 PMCID: PMC10745630 DOI: 10.3390/molecules28248090] [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: 10/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Doxorubicin (DOX), an anthracycline-based chemotherapeutic agent, is widely used to treat various types of cancer; however, prolonged treatment induces cardiomyotoxicity. Although studies have been performed to overcome DOX-induced cardiotoxicity (DICT), no effective method is currently available. This study investigated the effects and potential mechanisms of Poncirus trifoliata aqueous extract (PTA) in DICT. Changes in cell survival were assessed in H9c2 rat cardiomyocytes and MDA-MB-231 human breast cancer cells. The C57BL/6 mice were treated with DOX to induce DICT in vivo, and alterations in electrophysiological characteristics, serum biomarkers, and histological features were examined. The PTA treatment inhibited DOX-induced decrease in H9c2 cell viability but did not affect the MDA-MB-231 cell viability. Additionally, the PTA restored the abnormal heart rate, R-R interval, QT interval, and ST segment and inhibited the decrease in serum cardiac and hepatic toxicity indicators in the DICT model. Moreover, the PTA administration protected against myocardial fibrosis and apoptosis in the heart tissue of mice with DICT. PTA treatment restored DOX-induced decrease in the expression of NAD(P)H dehydrogenase quinone acceptor oxidoreductase 1 in a PTA concentration-dependent manner. In conclusion, the PTA inhibitory effect on DICT is attributable to its antioxidant properties, suggesting the potential of PTA as a phytotherapeutic agent for DICT.
Collapse
Affiliation(s)
| | | | | | | | - Jangho Lee
- Korea Food Research Institute, Wanju 55365, Republic of Korea; (M.-S.K.); (H.-K.C.); (S.-H.P.); (J.-I.L.)
| |
Collapse
|