1
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [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: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
2
|
Qiu Q, Sun Q, Yang J, Yuan Q, Wang P, Liu Q, Cui Z, Ma X, Li M. The molecular mechanism by which CTSB degrades FPN to disrupt macrophage iron homeostasis and promote the progression of atherosclerosis. Mol Cell Biochem 2025; 480:3889-3906. [PMID: 39960586 DOI: 10.1007/s11010-025-05228-9] [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: 11/25/2024] [Accepted: 02/05/2025] [Indexed: 05/23/2025]
Abstract
The incidence of atherosclerosis (AS) remains high, and iron-dependent cell death (termed ferroptosis) is thought to play a key role in the progression of AS. Studies have shown that cathepsin B (CTSB) is an important regulatory molecule in atherosclerosis. However, how CTSB regulates AS progression has not been reported, and whether it is related to ferroptosis is poorly studied. In the present study, we observed a significant upregulation of CTSB expression in two AS models, ApoE knockout mice and SD rats given a HFD. According to our findings, CTSB can promote development of the AS plaque region, while inhibition of CTSB showed a reduction of AS lesion area and lipid deposition. Single-cell transcriptome analysis of AS tissue from humans revealed that CTSB is primarily expressed in macrophages. Oxidized low-density lipoprotein (ox-LDL) significantly enhanced macrophage CTSB expression, and induced ferroptosis in vitro. Mechanistically, Ferroportin (FPN) is the binding target of CTSB. CTSB can negatively regulate the protein level of FPN and promote its degradation, which further leads to ferroptosis of macrophages. We confirmed that ferroptosis in macrophages could be effectively inhibited by knockdown or pharmacological inhibition of CTSB, which in turn slowed the progression of AS. In conclusion, our study suggests that CTSB disrupts iron homeostasis in macrophages by degrading FPN and induces ferroptosis, thereby exacerbating the development of AS. Targeting CTSB may become an important potential strategy for the treatment of AS.
Collapse
Affiliation(s)
- Quanli Qiu
- Jinzhou Medical University Graduate Training Base (PLA 960, Hospital), 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Qiyu Sun
- Jinzhou Medical University Graduate Training Base (PLA 960, Hospital), 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Jiaxin Yang
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Qingxin Yuan
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Ping Wang
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Qingwei Liu
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Zhenzhen Cui
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China
| | - Xiaowen Ma
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
| | - Min Li
- Jinzhou Medical University Graduate Training Base (PLA 960, Hospital), 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
- PLA 960, Hospital, 25 Shifan Road, Tianqiao District, Jinan, 250031, Shandong, China.
| |
Collapse
|
3
|
Liu X, Wu H, Ren Z, Wang Y, Yang G, Cong H, Yu B. Dual-Functional Antioxidant CIRAKLC Peptide Discovery: A Biomaterial-Integrated Strategy for ROS Scavenging and Enzyme Modulation in Oxidative Stress Therapeutics. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40377334 DOI: 10.1021/acsami.5c05089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Antioxidants play a pivotal role in activating endogenous defense mechanisms, neutralizing free radicals, and mitigating oxidative stress. In this study, an antioxidant peptide library was constructed based on amino acid structure-activity relationships, focusing on ROS scavenging and enzymatic modulation. Through comprehensive chemical and biological evaluations, we identified the peptide CIRAKLC (designated as CL7), which exhibited exceptional ROS-scavenging capacity and enzyme-modulating activity, demonstrating superior in vitro antioxidant efficacy. CL7 exhibited excellent biocompatibility and effectively protected HepG2 cells from H2O2-induced ROS-mediated oxidative damage through both preventive and reparative mechanisms. Furthermore, CL7 modulated key enzymes (CAT, MDA, SOD, ALT, and AST), displaying hepatoprotective, anti-inflammatory, and antiaging effects, along with significant photoprotective efficacy. Molecular docking revealed key hydrogen bonding and electrostatic interactions when the peptide binds to redox-related receptors (Nrf2/Keap1 and IKK-β). These findings underscore CL7's robust antioxidant capacity and multifunctionality, highlighting its broad therapeutic potential in oxidative stress management.
Collapse
Affiliation(s)
- Xin Liu
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Han Wu
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Zekai Ren
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Yumei Wang
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Ge Yang
- School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China
| | - Hailin Cong
- School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China
- College of Chemistry, Chemical Engineering and Materials Science, Zaozhuang University, Zaozhuang 277160, China
| | - Bing Yu
- College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| |
Collapse
|
4
|
Cao L, Chen J, Ni H, Gong X, Zang Z, Chang H. Kumquat Flavonoids Attenuate Atherosclerosis in ApoE -/- Mice by Inhibiting the Activation of NLRP3 Inflammasome through Upregulating MicroRNA-145. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40260463 DOI: 10.1021/acs.jafc.5c00295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Atherosclerosis (AS) is widely recognized as a consequence of chronic inflammation, with the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome playing a pivotal role in mediating this inflammatory response. Kumquat flavonoids (KFs), the primary active ingredients in kumquat, have demonstrated potential in modulating inflammation and may help prevent AS. Herein, this study aimed to explore the protective effects and underlying mechanisms of KFs on AS using an ApoE-/- mouse model fed a high-fat/cholesterol diet (HFCD) and the mouse aortic vascular smooth muscle cell (MOVAS) inflammation model induced by oxidized low-density lipoprotein (ox-LDL). Our results show that KFs significantly reduced serum lipid levels and suppressed the overproduction of inflammatory cytokines in ApoE-/- mice. Notably, KFs also decreased the area of atherosclerotic lesions and plaque formation in the aorta of ApoE-/- mice. Additionally, in vivo (mouse aortic tissue) and in vitro (MOVAS cells), KFs were found to inhibit the activation of NLRP3 inflammasome and simultaneously upregulate microRNA-145 (miR-145). In conclusion, our findings suggest that KFs exert their inhibitory effects on NLRP3 inflammasome through upregulating miR-145, thereby alleviating the progression of AS.
Collapse
Affiliation(s)
- Linhai Cao
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Junli Chen
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing 100094, China
| | - Hongxia Ni
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Xiaoxiao Gong
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Ziyan Zang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Hui Chang
- College of Food Science, Southwest University, Chongqing 400715, China
| |
Collapse
|
5
|
Huang Z, Peng Z, Huang D, Zhou Z. Virtual Screening of Kelch-like ECH-Associated Protein 1-Nuclear Factor Erythroid 2-Related Factor 2 (Keap1-Nrf2) Inhibitors and In Vitro Validation. Molecules 2025; 30:1815. [PMID: 40333848 PMCID: PMC12029559 DOI: 10.3390/molecules30081815] [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: 02/11/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
The transcription factor erythroid 2-related factor 2 (Nrf2) is a central regulator of cellular defense mechanisms against oxidative stress and inflammation. Keap1 (Kelch-like ECH-associated protein 1) regulates Nrf2 activity by ubiquitination-mediated cytoplasmic retention, thereby suppressing its nuclear translocation and subsequent transcriptional activation of genes encoding phase II detoxifying enzymes. Using a structure-based virtual screening approach, we screened ~16,000 natural compounds to identify Keap1-Nrf2 PPI inhibitors. Nine compounds were identified based on their high binding affinities and favorable interactions with Keap1, primarily through non-covalent interactions. To validate the binding stability of these inhibitors, molecular dynamics (MD) simulations were performed, confirming the robustness of the Keap1-inhibitor complexes over time. Subsequent in vitro assays on human epithelial keratinocyte cells (HaCaT) revealed that six of these compounds notably upregulated Nrf2 mRNA expression, regis tering increases from 23% to 50% in comparison to the control. Notably, chebulinic acid emerged as the most potent compound, demonstrating the greatest elevation in Nrf2 expression. Penetration studies further showed that chebulinic acid, when encapsulated in silk fibroin, achieved a 0.14% penetration rate after 24 h though it could not penetrate into the stratum corneum alone. This result highlighted the potential of chebulinic acid in the use of anti-aging skincare formulations. Collectively, our findings affirmed that molecular docking is a reliable and effective approach for the identification of novel anti-aging agents targeting the Keap1-Nrf2 pathway.
Collapse
Affiliation(s)
- Zhengwan Huang
- Bloomage Biotechnology Corporation Limited, Shanghai 200100, China; (Z.H.); (D.H.)
| | - Zhengang Peng
- Bloomage Biotechnology Corporation Limited, Shanghai 200100, China; (Z.H.); (D.H.)
| | - Dandan Huang
- Bloomage Biotechnology Corporation Limited, Shanghai 200100, China; (Z.H.); (D.H.)
| | - Zhongyu Zhou
- Guangdong Provincial Key Laboratory of Applied Botany & Key Laboratory of National Forestry and Grassland Administration on Plant Conservation and Utilization in Southern China, South China Botanical Garden, Guangzhou 510650, China
| |
Collapse
|
6
|
Dai R, Qian Y, Liu S, Zou X, Sun S, Sun Z. Growth Arrest-specific 1 Inhibits Keap1/Nrf2 Signaling Transduction in the Activation of the Ferroptosis Program in Retinal Müller Cells. FRONT BIOSCI-LANDMRK 2025; 30:27954. [PMID: 40152386 DOI: 10.31083/fbl27954] [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: 11/07/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Diabetes retinopathy (DR) represents a microvascular disease in diabetes. Growth arrest-specific 1 (GAS1) is differentially expressed in rat retinal Müller cells under high glucose (HG) conditions, and its promotion of ferroptosis contributes to retinal cell death. However, the influence of GAS1 in DR is elusive. Herein, we aimed to investigate the effect and potential mechanism based on GAS1-mediated ferroptosis on DR. METHODS After HG treatment, the differentially expressed genes in rat retinal Müller cells were analyzed by transcriptome sequencing followed by Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses; finally, GAS1 was selected. The effects of GAS1 knockdown/overexpression and nuclear factor erythroid 2-related factor (Nrf2) silencing on viability, apoptosis, lipid peroxidation, Fe2+, and oxidative stress levels in HG-induced/transfected Müller cells were measured by Cell Counting Kit-8 (CCK-8) assay, flow cytometry, and commercial reagent kits. The potential effects of GAS1 and Nrf2, especially on GAS1, Nrf2, and Kelch-like ECH-associated protein 1 (Keap1) expressions in cells, were determined by quantitative real-time polymerase chain reaction (qRT-PCR) or Western blot. RESULTS HG treatment decreased cell viability and glutathione (GSH) levels and increased apoptosis, lipid reactive oxygen species (ROS), glutathione disulfide (GSSG), malondialdehyde (MDA), oxidative stress, and Fe2+ levels in Müller cells (p < 0.01). HG treatment also upregulated GAS1, Keap1, and total Nrf2 expressions while downregulating nuclear Nrf2 in Müller cells (p < 0.001). GAS1 downregulation enhanced cell viability, GSH levels, and nuclear Nrf2 expression while reducing the levels of apoptosis, lipid ROS, GSSG, MDA, Fe2+, Keap1, and total Nrf2 in HG-treated Müller cells (p < 0.001), whereas GAS1 overexpression had the opposite effects. Additionally, Nrf2 silencing reversed the impact of GAS1 overexpression in HG-treated Müller cells (p < 0.05). CONCLUSION GAS1 inhibits Keap1/Nrf2 signaling transduction in activating ferroptosis in retinal Müller cells; thus, this study can aid in setting the stage for novel treatment methods against DR.
Collapse
Affiliation(s)
- Rongfeng Dai
- Department of Endocrinology, the Third People's Hospital of Changzhou, 213001 Changzhou, Jiangsu, China
| | - Yu Qian
- Department of Endocrinology, the Third People's Hospital of Changzhou, 213001 Changzhou, Jiangsu, China
| | - Siqi Liu
- Department of Endocrinology, the Third People's Hospital of Changzhou, 213001 Changzhou, Jiangsu, China
| | - Xi Zou
- Department of Ophthalmology, the Third People's Hospital of Changzhou, 213001 Changzhou, Jiangsu, China
| | - Shanshan Sun
- Department of Ophthalmology, the Third People's Hospital of Changzhou, 213001 Changzhou, Jiangsu, China
| | - Zhuo Sun
- Department of Ophthalmology, the Third People's Hospital of Changzhou, 213001 Changzhou, Jiangsu, China
| |
Collapse
|
7
|
Zhang DD. Thirty years of NRF2: advances and therapeutic challenges. Nat Rev Drug Discov 2025:10.1038/s41573-025-01145-0. [PMID: 40038406 DOI: 10.1038/s41573-025-01145-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2025] [Indexed: 03/06/2025]
Abstract
Over the last 30 years, NRF2 has evolved from being recognized as a transcription factor primarily involved in redox balance and detoxification to a well-appreciated master regulator of cellular proteostasis, metabolism and iron homeostasis. NRF2 plays a pivotal role in diverse pathologies, including cancer, and metabolic, inflammatory and neurodegenerative disorders. It exhibits a Janus-faced duality, safeguarding cellular integrity in normal cells against environmental insults to prevent disease onset, whereas in certain cancers, constitutively elevated NRF2 levels provide a tumour survival advantage, promoting progression, therapy resistance and metastasis. Advances in understanding the mechanistic regulation of NRF2 and its roles in human pathology have propelled the investigation of NRF2-targeted therapeutic strategies. This Review dissects the mechanistic intricacies of NRF2 signalling, its cross-talk with biological processes and its far-reaching implications for health and disease, highlighting key discoveries that have shaped innovative therapeutic approaches targeting NRF2.
Collapse
Affiliation(s)
- Donna D Zhang
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
- University of Florida Health Cancer Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
8
|
Chen Y, Xu B, Lin Q, Zhu X, Lv Y, Bai X, Weng X, Du J, Li M, Zhu Y, Mou J, Wang M, Wang Y, Luo X, Xu C. Spermine delivered by ZIF90 nanoparticles alleviates atherosclerosis by targeted inhibition of macrophage ferroptosis in plaque. J Nanobiotechnology 2025; 23:165. [PMID: 40038689 PMCID: PMC11877808 DOI: 10.1186/s12951-025-03271-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Nowadays, emerging evidence have suggested that the ferroptosis of macrophages could contribute to the progression of atherosclerosis (AS). Meanwhile, Spermine (Sp) could serve as an endogenous small molecule exhibiting a wide range of cardiovascular protective effects. METHODS Zeolitic imidazolate framework-90 (ZIF90) nanoparticles were synthesized and utilized to create a novel delivery nanosystem encapsulated with Sp (CD16/32-ZIF90@Sp). The efficacy of CD16/32-ZIF90@Sp in protecting against AS and ferroptosis was evaluated in ApoE-/- mice and macrophages, with a focus on assessing potential adverse effects in vivo. RESULTS CD16/32-ZIF90@Sp exhibited reliable and stable delivery of Sp within acidic environments and ATP sensitivity. CD16/32-ZIF90@Sp effectively reduced the cytotoxicity of Sp. As is evidenced by in vitro and vivo experiments, CD16/32-ZIF90@Sp showed precise targeting of macrophages within atherosclerotic plaques and ox-LDL-activated macrophages. Furthermore, treatment with CD16/32-ZIF90@Sp effectively attenuated the progression of AS and the ferroptosis of macrophage within plaque in ApoE-/- mice without causing significant side effects. Mechanistically, we found that CD16/32-ZIF90@Sp inhibited ferroptosis via improving mitochondrial function and upregulating the expression level of GPX4/xCT. CONCLUSION Our study demonstrated that CD16/32-modified ZIF90 nanoparticles could effectively target macrophages within atherosclerotic plaques, leading to the inhibition of atherosclerotic plaque progression in ApoE-/- mice. These effects were attributed to the enhancement of mitochondrial function and the inhibition of macrophage ferroptosis, with limited side effects.
Collapse
Affiliation(s)
- Yuwu Chen
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Biyi Xu
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Quan Lin
- Department of Thoracic Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150001, China
| | - Xinxin Zhu
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Ying Lv
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Xiaoxuan Bai
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Xiuzhu Weng
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Jie Du
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Man Li
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Yuxiao Zhu
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Junke Mou
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Mengyang Wang
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China
| | - Yuehong Wang
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China.
- Department of Pathophysiology, Harbin Medical University, Harbin, 150001, China.
| | - Xing Luo
- Department of Cardiology, 2, Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, People's Republic of China.
- Department of Pathophysiology, Harbin Medical University, Harbin, 150001, China.
| | - Changqing Xu
- Department of Pathophysiology, Harbin Medical University, Harbin, 150001, China
| |
Collapse
|
9
|
Liu C, Pan J, Bao Q. Ferroptosis in senescence and age-related diseases: pathogenic mechanisms and potential intervention targets. Mol Biol Rep 2025; 52:238. [PMID: 39960579 DOI: 10.1007/s11033-025-10338-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/04/2025] [Indexed: 05/09/2025]
Abstract
As the global population continues to age, the prevalence of age-related diseases is increasing, significantly influencing social and economic development, the stability of social security systems, and progress in medical technology. Ferroptosis, a recently discovered form of programmed cell death driven by iron-dependent lipid peroxidation, has emerged as a key area of research. Studies have revealed a strong association between ferroptosis and senescence. In this article, we systematically summarize the molecular mechanisms and associated signaling pathways underlying ferroptosis, emphasizing its pivotal role in the onset and progression of age-related diseases. By providing new perspectives, we aim to advance understanding of the pathogenesis of age-related diseases and guide the development of effective intervention strategies.
Collapse
Affiliation(s)
- Chang Liu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Pan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Qi Bao
- Zhejiang University School of Medicine, Hangzhou, China.
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
10
|
Li W, Li Y, Xiao L, Xie Z, Peng J, Huang W, Li X, Meng Y. Micheliolide attenuates sepsis-induced acute lung injury by suppressing mitochondrial oxidative stress and PFKFB3-driven glycolysis. J Transl Med 2025; 23:181. [PMID: 39953547 PMCID: PMC11829335 DOI: 10.1186/s12967-024-05906-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/19/2024] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Sepsis is a potentially fatal condition with a significant risk of death. Acute lung injury (ALI) is a life-threatening complication of sepsis, and the inflammatory response plays a critical role in sepsis-induced ALI. The protective effects of micheliolide (MCL) against renal fibrosis and leukemia have been demonstrated, but the precise underlying mechanisms remain unclear. METHODS In vitro, lipopolysaccharides (LPS) and interferon-gamma (IFN-γ) were used to stimulate RAW264.7 cells and bone marrow-derived macrophages (BMDMs) to investigate the protective effect of MCL on sepsis-induced ALI. Cecal ligation and puncture (CLP) models were constructed in mice to induce ALI in vivo. The expression of inflammatory factors, macrophage polarization markers, and the glycolysis-related enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) were measured in vivo. Mitochondrial function, oxidative stress, and mitochondrial-related proteins were evaluated in vitro. RESULTS MCL inhibited CLP-induced ALI, as evidenced by improvements in proinflammatory factor levels, lung wet/dry ratios, and histopathological findings. In vitro, MCL treatment significantly suppressed LPS + IFN-γ-induced M1-type polarization of RAW264.7 cells and BMDMs, as well as the production of inflammatory factors and oxidative stress. Mechanistic experiments revealed that MCL suppresses PFKFB3-driven glycolysis to reduce inflammation and activates the mitochondrial unfolded protein response (UPRmt) to alleviate mitochondrial stress. However, the therapeutic effect of MCL was diminished when PFKFB3 was overexpressed in cells. CONCLUSION This study is the first to demonstrate that MCL attenuates sepsis-induced ALI by reducing M1-type macrophage polarization. Its therapeutic effect is closely related to the suppression of oxidative stress and PFKFB3-driven glycolysis.
Collapse
Affiliation(s)
- Wenhan Li
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Yuhan Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Linjie Xiao
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Zhanzhan Xie
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Jun Peng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Wenhui Huang
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| | - Ying Meng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| |
Collapse
|
11
|
Fan S, Wang K, Zhang T, Deng D, Shen J, Zhao B, Fu D, Chen X. Mechanisms and Therapeutic Potential of GPX4 in Pain Modulation. Pain Ther 2025; 14:21-45. [PMID: 39503961 PMCID: PMC11751247 DOI: 10.1007/s40122-024-00673-8] [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: 08/14/2024] [Accepted: 10/04/2024] [Indexed: 01/23/2025] Open
Abstract
Pain, a complex symptom encompassing both sensory and emotional dimensions, constitutes a significant global public health issue. Oxidative stress is a pivotal factor in the complex pathophysiology of pain, with glutathione peroxidase 4 (GPX4) recognized as a crucial antioxidant enzyme involved in both antioxidant defense mechanisms and ferroptosis pathways. This review systematically explores GPX4's functions across various pain models, including neuropathic, inflammatory, low back, and cancer-related pain. Specifically, the focus includes GPX4's physiological roles, antioxidant defense mechanisms, regulation of ferroptosis, involvement in signal transduction pathways, and metabolic regulation. By summarizing current research, we highlight the potential of GPX4-targeted therapies in pain management.
Collapse
Affiliation(s)
- Shiwen Fan
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, 832002, China
| | - Kaixin Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Tianhao Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Daling Deng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Jiwei Shen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Bowen Zhao
- Department of Anesthesiology, First Affiliated Hospital of Shihezi University, Shihezi, 832002, China
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Daan Fu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| | - Xiangdong Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
12
|
Lai K, Chen Z, Lin S, Ye K, Yuan Y, Li G, Song Y, Ma H, Mak TW, Xu Y. The IDH1-R132H mutation aggravates cisplatin-induced acute kidney injury by promoting ferroptosis through disrupting NDUFA1 and FSP1 interaction. Cell Death Differ 2025; 32:242-255. [PMID: 39306640 PMCID: PMC11802792 DOI: 10.1038/s41418-024-01381-8] [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: 07/28/2024] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 02/08/2025] Open
Abstract
The IDH1-R132H mutation is implicated in the development of various tumors. Whether cisplatin, a common chemotherapeutic agent, induces more significant renal toxicity in individuals with the IDH1-R132H mutation remains unclear. In this study, we observed that the IDH1-R132H mutation exacerbates mitochondrial lipid peroxidation and dysfunction in renal tubules, rendering the kidneys more susceptible to cisplatin-induced ferroptosis. The IDH1-R132H mutation increases methylation of the Ndufa1 promoter, thereby suppressing NDUFA1 transcription and translation. This suppression disrupts NDUFA1's interaction with FSP1, reducing its resistance to cisplatin-induced tubular epithelial cell death. As a consequence, ROS accumulates, lipid peroxidation occurs, and ferroptosis is triggered, thereby promoting acute kidney injury. In summary, this study elucidates a novel mechanism underlying cisplatin-induced nephrotoxicity and provides valuable insights for the development of personalized treatment strategies for tumor patients carrying the IDH1-R132H mutation.
Collapse
Affiliation(s)
- Kunmei Lai
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhimin Chen
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Siyi Lin
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Keng Ye
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ying Yuan
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Guoping Li
- Department of Pathology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yankun Song
- Department of Pathology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Huabin Ma
- Central Laboratory, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Tak W Mak
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SA, China.
| | - Yanfang Xu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Research Center for Metabolic Chronic Kidney Disease, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
13
|
Shi N, Wang JW, Su G, Ma G, Huang FQ, Jin SJ, Xie HM, Ge WX, Song JP, Luan X, Zhang L, Qi LW. N -Acetyl-Tryptophan in Acute Kidney Injury after Cardiac Surgery. J Am Soc Nephrol 2025:00001751-990000000-00537. [PMID: 39847454 DOI: 10.1681/asn.0000000626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
Key Points
N-acetyl-tryptophan (NAT) was elevated in plasma of patients with cardiac surgery–associated AKI compared with those who do not develop AKI after surgery.NAT exhibited kidney-protective effects in ischemia-reperfusion–, cisplatin-, and unilateral ureteral obstruction–induced kidney injury mouse models and epithelial cellular models.NAT interacted with Kelch-like ECH-associated protein 1 at 483 and 508 sites, resulting in Nrf2 nuclear translocation and proteasome genes transcription, respectively.
Background
Cardiac surgery–associated AKI is a common serious complication after cardiac surgery. Currently, there are no specific pharmacological therapies. Our understanding of its pathophysiology remains preliminary.
Methods
A total of 2504 patients with and without AKI after cardiac surgery were enrolled. High-performance liquid chromatography coupled with mass spectrometry was used for untargeted analysis of metabolites in plasma, identifying significant differential metabolites. Subsequently, a liquid chromatography–tandem mass spectrometry–based approach using isotope-labeled standard addition was performed for targeted analysis of the metabolic marker N-acetyl-tryptophan (NAT). The function of NAT was determined using different kidney injury mouse models and epithelial cellular models. Transcriptome sequencing, surface plasmon resonance, and protein mutation were used to explore the mechanism of NAT on the kidney.
Results
We identified a total of 32 differential metabolites related to AKI occurrence on the basis of a cohort of 1042 patients. Among them, NAT was elevated in plasma of patients with cardiac surgery–associated AKI compared with those who did not develop AKI after cardiac surgery. The higher level of NAT in plasma was confirmed by accurate targeted quantification. NAT exhibited kidney-protective effects in ischemia-reperfusion–, cisplatin-, and unilateral ureteral obstruction–induced kidney injury mouse models. Mechanistically, NAT exerted kidney-protective effects by interacting with Kelch-like ECH-associated protein 1 at 483 and 508 sites, resulting in Nrf2 nuclear translocation and the transcription of proteasome genes, respectively.
Conclusions
NAT plays a key role in kidney protection.
Collapse
Affiliation(s)
- Ning Shi
- Pukou Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ji-Wen Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gengchen Su
- School of Medicine, Tsinghua University, Beijing, China
| | - Gaoxiang Ma
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Feng-Qing Huang
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, China
| | - Si-Jia Jin
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, China
| | - Hua-Mei Xie
- Pukou Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wen-Xin Ge
- Pukou Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jiang-Ping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaodong Luan
- Center for Drug Research and Evaluation, Institute of clinical medicine, Peking Union Medical College Hospital, Beijing, China
| | - Lei Zhang
- Pukou Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lian-Wen Qi
- Pukou Hospital of Chinese Medicine, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
14
|
Wang Z, Jin X, Yong X. Identification of ferroptosis-related LncRNAs as potential targets for improving immunotherapy in glioblastoma. Comput Methods Biomech Biomed Engin 2025:1-13. [PMID: 39743840 DOI: 10.1080/10255842.2024.2448556] [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: 05/15/2024] [Revised: 10/22/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025]
Abstract
The effect of ferroptosis-related long non-coding RNAs (lncRNAs) in predicting immunotherapy response to glioblastoma (GBM) remains obscure. This study established a 11-lncRNAs prognostic signature. Differential gene expression analysis, univariate and multivariate Cox regression analyses and the least absolute shrinkage and selection operator (LASSO) regression algorithm were used to identify prognostic ferroptosis-related genes and establish a nomogram model of risk score. Kaplan-Meier survival plots and receiver operating characteristic (ROC) curve analysis were used to evaluate the prognostic accuracy of the model in the TCGA-GBM cohort. To verify the expression of these signatures, we analyzed the expression levels of three lncRNAs (AGAP2-AS1, OSMR-AS1, UNC5B-AS1) in LN229 and U87 cells. The ROC analysis showed that the area under curve (AUC) of this signature is 0.814, suggesting that it has a promising performance on GBM prognostic prediction. Kaplan-Meier analysis showed that the survival rate of GBM patients in high-risk group was significantly lower than low-risk group, and the performance of this signature on GBM prognostic prediction was superior to conventional clinicopathological factors. Further qRT-PCR experiment also confirmed our prediction of lncRNA signatures. These ferroptosis-related lncRNAs might be therapeutic targets for glioblastoma, and targeting these lncRNAs can also improve the efficacy of immunotherapy, especially immune checkpoint inhibitors. Mechanistically, these findings might attribute to N6-methyladenosine (m6A) mRNA modification on lncRNAs.
Collapse
Affiliation(s)
- Zhaochen Wang
- Department of Neurosurgery, Northwest University Xi'an No1 Hospital, Xi'an, China
| | - Xiao Jin
- The Personnel Department, Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoli Yong
- Department of Neurology, Chang'An Hospital, Economic and Technological Development District, Xi'an, China
| |
Collapse
|
15
|
Jin X, Chen L, Yang Y, Tan R, Jiang C. Adverse Effects of Nrf2 in Different Organs and the Related Diseases. Antioxid Redox Signal 2024. [PMID: 39723588 DOI: 10.1089/ars.2024.0586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Significance: Under normal physiological conditions, Nrf2 undergoes ubiquitination and subsequent proteasome degradation to maintain its basal activity. Oxidative stress can trigger Nrf2 activation, prompting its translocation to the nucleus where it functions as a transcription factor, activating various antioxidant pathways, and conferring antioxidant properties. Recent Advances: While extensive research has shown Nrf2's protective role in various diseases, emerging evidence suggests that Nrf2 activation can also produce harmful effects. Critical Issues: This review examines the pathological contexts in which Nrf2 assumes different roles, emphasizing the mechanisms and conditions that result in adverse outcomes. Future Directions: Persistent Nrf2 activation may have deleterious consequences, necessitating further investigation into the specific conditions and mechanisms through which Nrf2 exerts its harmful effects. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Xuemei Jin
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
- Department of Preventive Medicine, School of Medicine, Yanbian University, Yanji, China
| | - Long Chen
- Department of Orthopedics, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Yuelan Yang
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
- Department of Nephrology, The First Clinical College of Guangdong Medical University, Zhanjiang, China
| | - Rongshao Tan
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Chunjie Jiang
- Department of Clinical Nutrition, Guangzhou Institute of Disease-Oriented Nutritional Research, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
16
|
Zhang L, Li J, Kou Y, Shen L, Wang H, Wang Y, Ma R, Wu T, Yang X, Gu Y, Yi L. Mechanisms and treatment of atherosclerosis: focus on macrophages. Front Immunol 2024; 15:1490387. [PMID: 39569201 PMCID: PMC11576186 DOI: 10.3389/fimmu.2024.1490387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/09/2024] [Indexed: 11/22/2024] Open
Abstract
Macrophages are the basic mediators and coordinators of various types of chronic inflammation and play a crucial role in the formation and development of atherosclerosis (AS). In the complex microenvironment of atherosclerotic plaques, macrophages of different sources are exposed to different signal stimuli and thus polarized into various subpopulations. Various types of macrophages with predominantly M1 and M2 phenotypes also play different regulatory roles in the initiation and progression of AS. Lipid-lowering drugs, mainly statins, are widely used in clinical practice, but the adverse reactions are obvious and there is a lack of personalized treatment. Emerging targeted macrophage and Traditional Chinese medicine (TCM)-related therapies can regulate the cellular microenvironment, inhibit the polarization of M1 macrophages, and promote the activation of M2 macrophages, providing new ideas for the prevention and treatment of AS.
Collapse
Affiliation(s)
- LingNa Zhang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - JiaWei Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - YuShun Kou
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - LuFan Shen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Hong Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - YiYuan Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Ruiling Ma
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Tao Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Xin Yang
- First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - YuanHui Gu
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Lin Yi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Chronic Disease Laboratory, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
17
|
Chen X, Yang Z, Liao M, Zhao Q, Lu Y, Li Q, Liu S, Li S, Chen J, He Y. Ginkgo Flavone Aglycone Ameliorates Atherosclerosis via Inhibiting Endothelial Pyroptosis by Activating the Nrf2 Pathway. Phytother Res 2024; 38:5458-5473. [PMID: 39322309 DOI: 10.1002/ptr.8321] [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: 04/25/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/27/2024]
Abstract
Natural antioxidants have been shown to be effective against atherosclerosis. Ginkgo flavone aglycone (GA) has strong antioxidant properties and can protect against endothelial damage. However, the mechanisms by which GA protects against atherosclerosis remain largely unexplored. This study hopes to find the anti-atherosclerotic mechanism of GA. ApoE-/- mice fed a high-fat diet were used for modeling atherosclerosis. The efficacy of GA on mice with atherosclerosis was evaluated based on the following indicators: Oil Red O staining, Masson staining, lipid content, and apoptosis. Transmission electron microscopy, Western blot, immunofluorescence staining, and propidium iodide staining were used to analyze the effects of GA on ox-LDL-treated human aortic endothelial cells. GA activated Nrf2 by promoting the nuclear translocation of Nrf2, thereby inhibiting endothelial pyroptosis. GA prevented endothelial pyroptosis suppressed oxidative stress, and inhibited the development of atherosclerosis in ApoE-/- mice fed high-fat diets. At the cellular level, GA suppressed ox-LDL-induced pyroptosis of HAECs by reducing reactive oxygen species (ROS) levels and inhibiting NLRP3 inflammasome. Furthermore, siRNA targeting Nrf2 or ML385, an Nrf2 inhibitor, reversed these effects. GA liberated Nrf2 from Keap1 sequestration, enhanced the nuclear translocation of Nrf2 and the transcription of downstream antioxidant proteins, reinforced the antioxidant defense system, and inhibited oxidative stress, thereby preventing endothelial cell pyroptosis, and attenuating the progression of atherosclerosis. This study indicated that GA mitigated endothelial pyroptosis by modulating Keap1/Nrf2 interactions, shedding light on the potential mechanisms underlying the protective effects of natural antioxidants against atherosclerosis.
Collapse
Affiliation(s)
- Xingyi Chen
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Zhuan Yang
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Meijuan Liao
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Qing Zhao
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yuan Lu
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Qin Li
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shijing Liu
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shiliang Li
- Department of Vascular Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jiyu Chen
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yan He
- Clinical Trials Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| |
Collapse
|
18
|
Han H, Zhang G, Zhang X, Zhao Q. Nrf2-mediated ferroptosis inhibition: a novel approach for managing inflammatory diseases. Inflammopharmacology 2024; 32:2961-2986. [PMID: 39126567 DOI: 10.1007/s10787-024-01519-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 06/28/2024] [Indexed: 08/12/2024]
Abstract
Inflammatory diseases, including psoriasis, atherosclerosis, rheumatoid arthritis, and ulcerative colitis, are characterized by persistent inflammation. Moreover, the existing treatments for inflammatory diseases only provide temporary relief by controlling symptoms, and treatments of unstable and expensive. Therefore, new therapeutic solutions are urgently needed to address the underlying causes or symptoms of inflammatory diseases. Inflammation frequently coincides with a high level of (reactive oxygen species) ROS activation, serving as a fundamental element in numerous physiological and pathological phenotypes that can result in serious harm to the organism. Given its pivotal role in inflammation, oxidative stress, and ferroptosis, ROS represents a focal node for investigating the (nuclear factor E2-related factor 2) Nrf2 pathway and ferroptosis, both of which are intricately linked to ROS. Ferroptosis is mainly triggered by oxidative stress and involves iron-dependent lipid peroxidation. The transcription factor Nrf2 targets several genes within the ferroptosis pathway. Recent studies have shown that Nrf2 plays a significant role in three key ferroptosis-related routes, including the synthesis and metabolism of glutathione/glutathione peroxidase 4, iron metabolism, and lipid processes. As a result, ferroptosis-related treatments for inflammatory diseases have attracted much attention. Moreover, drugs targeting Nrf2 can be used to manage inflammatory conditions. This review aimed to assess ferroptosis regulation mechanism and the role of Nrf2 in ferroptosis inhibition. Therefore, this review article may provide the basis for more research regarding the treatment of inflammatory diseases through Nrf2-inhibited ferroptosis.
Collapse
Affiliation(s)
- Hang Han
- College of Pharmacy, Chongqing Medical University, Chongqing, Chongqing, 400016, China
| | - Guojiang Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, Chongqing, 400016, China
| | - Xiao Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, Chongqing, 400016, China.
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, Chongqing, 400016, China.
| |
Collapse
|
19
|
Wu Q, Yao J, Xiao M, Zhang X, Zhang M, Xi X. Targeting Nrf2 signaling pathway: new therapeutic strategy for cardiovascular diseases. J Drug Target 2024; 32:874-883. [PMID: 38753446 DOI: 10.1080/1061186x.2024.2356736] [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: 04/02/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally, with oxidative stress (OS) identified as a primary contributor to their onset and progression. Given the elevated incidence and mortality rates associated with CVDs, there is an imperative need to investigate novel therapeutic strategies. Nuclear factor erythroid 2-related factor 2 (Nrf2), ubiquitously expressed in the cardiovascular system, has emerged as a promising therapeutic target for CVDs due to its role in regulating OS and inflammation. This review aims to delve into the mechanisms and actions of the Nrf2 pathway, highlighting its potential in mitigating the pathogenesis of CVDs.
Collapse
Affiliation(s)
- Qi Wu
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| | - Jiangting Yao
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| | - Mengyun Xiao
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| | - Xiawei Zhang
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| | - Mengxiao Zhang
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Xinting Xi
- School of Medical Imaging, Bengbu Medical University, Bengbu, China
| |
Collapse
|
20
|
Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X, Huang S, Liu C. Macrophages in cardiovascular diseases: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:130. [PMID: 38816371 PMCID: PMC11139930 DOI: 10.1038/s41392-024-01840-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/02/2024] [Accepted: 04/21/2024] [Indexed: 06/01/2024] Open
Abstract
The immune response holds a pivotal role in cardiovascular disease development. As multifunctional cells of the innate immune system, macrophages play an essential role in initial inflammatory response that occurs following cardiovascular injury, thereby inducing subsequent damage while also facilitating recovery. Meanwhile, the diverse phenotypes and phenotypic alterations of macrophages strongly associate with distinct types and severity of cardiovascular diseases, including coronary heart disease, valvular disease, myocarditis, cardiomyopathy, heart failure, atherosclerosis and aneurysm, which underscores the importance of investigating macrophage regulatory mechanisms within the context of specific diseases. Besides, recent strides in single-cell sequencing technologies have revealed macrophage heterogeneity, cell-cell interactions, and downstream mechanisms of therapeutic targets at a higher resolution, which brings new perspectives into macrophage-mediated mechanisms and potential therapeutic targets in cardiovascular diseases. Remarkably, myocardial fibrosis, a prevalent characteristic in most cardiac diseases, remains a formidable clinical challenge, necessitating a profound investigation into the impact of macrophages on myocardial fibrosis within the context of cardiac diseases. In this review, we systematically summarize the diverse phenotypic and functional plasticity of macrophages in regulatory mechanisms of cardiovascular diseases and unprecedented insights introduced by single-cell sequencing technologies, with a focus on different causes and characteristics of diseases, especially the relationship between inflammation and fibrosis in cardiac diseases (myocardial infarction, pressure overload, myocarditis, dilated cardiomyopathy, diabetic cardiomyopathy and cardiac aging) and the relationship between inflammation and vascular injury in vascular diseases (atherosclerosis and aneurysm). Finally, we also highlight the preclinical/clinical macrophage targeting strategies and translational implications.
Collapse
Affiliation(s)
- Runkai Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Hongrui Zhang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Botao Tang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yukun Luo
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Yufei Yang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Xin Zhong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China
| | - Sifei Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Shengkang Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Canzhao Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510280, China.
| |
Collapse
|
21
|
Tao Y, Zhao Q, Lu C, Yong W, Xu M, Wang Z, Leng X. Melatonin suppresses atherosclerosis by ferroptosis inhibition via activating NRF2 pathway. FASEB J 2024; 38:e23678. [PMID: 38780199 DOI: 10.1096/fj.202400427rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Melatonin (MLT), a conserved small indole compound, exhibits anti-inflammatory and antioxidant properties, contributing to its cardioprotective effects. Lipoprotein-associated phospholipase A2 (Lp-PLA2) is associated with atherosclerosis disease risk, and is known as an atherosclerosis risk biomarker. This study aimed to investigate the impact of MLT on Lp-PLA2 expression in the atherosclerotic process and explore the underlying mechanisms involved. In vivo, ApoE-/- mice were fed a high-fat diet, with or without MLT administration, after which the plaque area and collagen content were assessed. Macrophages were pretreated with MLT combined with ox-LDL, and the levels of ferroptosis-related proteins, NRF2 activation, mitochondrial function, and oxidative stress were measured. MLT administration significantly attenuated atherosclerotic plaque progression, as evidenced by decreased plaque area and increased collagen. Compared with those in the high-fat diet (HD) group, the levels of glutathione peroxidase 4 (GPX4) and SLC7A11 (xCT, a cystine/glutamate transporter) in atherosclerotic root macrophages were significantly increased in the MLT group. In vitro, MLT activated the nuclear factor-E2-related Factor 2 (NRF2)/SLC7A11/GPX4 signaling pathway, enhancing antioxidant capacity while reducing lipid peroxidation and suppressing Lp-PLA2 expression in macrophages. Moreover, MLT reversed ox-LDL-induced ferroptosis, through the use of ferrostatin-1 (a ferroptosis inhibitor) and/or erastin (a ferroptosis activator). Furthermore, the protective effects of MLT on Lp-PLA2 expression, antioxidant capacity, lipid peroxidation, and ferroptosis were decreased in ML385 (a specific NRF2 inhibitor)-treated macrophages and in AAV-sh-NRF2 treated ApoE-/- mice. MLT suppresses Lp-PLA2 expression and atherosclerosis processes by inhibiting macrophage ferroptosis and partially activating the NRF2 pathway.
Collapse
Affiliation(s)
- Yangyang Tao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qinglong Zhao
- Department of Interventional Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengbo Lu
- Department of Cardiology, The First Affiliated Hospital of Jiamusi University, jiamusi, China
| | - Weilin Yong
- Department of Medical Services, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingyuan Xu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhuo Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoping Leng
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
22
|
Li C, Liu R, Xiong Z, Bao X, Liang S, Zeng H, Jin W, Gong Q, Liu L, Guo J. Ferroptosis: a potential target for the treatment of atherosclerosis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:331-344. [PMID: 38327187 PMCID: PMC10984869 DOI: 10.3724/abbs.2024016] [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: 11/06/2023] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
Atherosclerosis (AS), the main contributor to acute cardiovascular events, such as myocardial infarction and ischemic stroke, is characterized by necrotic core formation and plaque instability induced by cell death. The mechanisms of cell death in AS have recently been identified and elucidated. Ferroptosis, a novel iron-dependent form of cell death, has been proven to participate in atherosclerotic progression by increasing endothelial reactive oxygen species (ROS) levels and lipid peroxidation. Furthermore, accumulated intracellular iron activates various signaling pathways or risk factors for AS, such as abnormal lipid metabolism, oxidative stress, and inflammation, which can eventually lead to the disordered function of macrophages, vascular smooth muscle cells, and vascular endothelial cells. However, the molecular pathways through which ferroptosis affects AS development and progression are not entirely understood. This review systematically summarizes the interactions between AS and ferroptosis and provides a feasible approach for inhibiting AS progression from the perspective of ferroptosis.
Collapse
Affiliation(s)
- Chengyi Li
- School of MedicineYangtze UniversityJingzhou434020China
| | - Ran Liu
- School of MedicineYangtze UniversityJingzhou434020China
| | - Zhenyu Xiong
- School of MedicineYangtze UniversityJingzhou434020China
| | - Xue Bao
- School of MedicineYangtze UniversityJingzhou434020China
| | - Sijia Liang
- Department of PharmacologyZhongshan School of MedicineSun Yat-Sen UniversityGuangzhou510120China
| | - Haotian Zeng
- Department of GastroenterologyShenzhen People’s HospitalThe Second Clinical Medical CollegeJinan UniversityShenzhen518000China
| | - Wei Jin
- Department of Second Ward of General PediatricsSuizhou Central HospitalHubei University of MedicineSuizhou441300China
| | - Quan Gong
- School of MedicineYangtze UniversityJingzhou434020China
| | - Lian Liu
- School of MedicineYangtze UniversityJingzhou434020China
| | - Jiawei Guo
- School of MedicineYangtze UniversityJingzhou434020China
| |
Collapse
|