1
|
Peng X, Sun B, Tang C, Shi C, Xie X, Wang X, Jiang D, Li S, Jia Y, Wang Y, Tang H, Zhong S, Piao M, Cui X, Zhang S, Wang F, Wang Y, Na R, Huang R, Jiang Y, Zhang W, Xu J, Lin K, Guo J, Pan Z, Wang K, Zhao Q, Liu H, Yu B, Ji Y, Zhang J, Li S, Tian J. HMOX1-LDHB interaction promotes ferroptosis by inducing mitochondrial dysfunction in foamy macrophages during advanced atherosclerosis. Dev Cell 2025; 60:1070-1086.e8. [PMID: 39731912 DOI: 10.1016/j.devcel.2024.12.011] [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: 01/20/2024] [Revised: 08/27/2024] [Accepted: 12/04/2024] [Indexed: 12/30/2024]
Abstract
Advanced atherosclerosis is the pathological basis for acute cardiovascular events, with significant residual risk of recurrent clinical events despite contemporary treatment. The death of foamy macrophages is a main contributor to plaque progression, but the underlying mechanisms remain unclear. Bulk and single-cell RNA sequencing demonstrated that massive iron accumulation in advanced atherosclerosis promoted foamy macrophage ferroptosis, particularly in low expression of triggering receptor expressed on myeloid cells 2 (TREM2low) foamy macrophages. This cluster exhibits metabolic characteristics with low oxidative phosphorylation (OXPHOS), increasing ferroptosis sensitivity. Mechanically, upregulated heme oxygenase 1 (HMOX1)-lactate dehydrogenase B (LDHB) interaction enables Lon peptidase 1 (LONP1) to degrade mitochondrial transcription factor A (TFAM), leading to mitochondrial dysfunction and ferroptosis. Administration of the mitochondria-targeted reactive oxygen species (ROS) scavenger MitoTEMPO (mitochondrial-targeted TEMPO) or LONP1 inhibitor bortezomib restored mitochondrial homeostasis in foamy macrophages and alleviated atherosclerosis. Collectively, our study elucidates the cellular and molecular mechanism of foamy macrophage ferroptosis, offering potential therapeutic strategies for advanced atherosclerosis.
Collapse
Affiliation(s)
- Xiang Peng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Bin Sun
- College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Chaohui Tang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Chengyu Shi
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Xianwei Xie
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China
| | - Xueyu Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Dingsheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuo Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China
| | - Ying Jia
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yani Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China
| | - Huifang Tang
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang 421001, China
| | - Shan Zhong
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Minghui Piao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Xiuru Cui
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Shenghao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China
| | - Fan Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China
| | - Yan Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China
| | - Ruisi Na
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China; Heilongjiang Province Key Laboratory of Research on Molecular Targeted Anti-Tumor Drugs, Harbin 150081, China
| | - Renping Huang
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yanan Jiang
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin 150081, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin 150081, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Kaiyang Lin
- Department of Cardiology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Junli Guo
- Key Laboratory of Emergency and Trauma of Ministry of Education, Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences, Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Zhenwei Pan
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin 150081, China
| | - Kun Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Huibin Liu
- Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China.
| | - Yong Ji
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China.
| | - Jian Zhang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| | - Shuijie Li
- College of Pharmacy, Harbin Medical University, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China; Heilongjiang Province Key Laboratory of Research on Molecular Targeted Anti-Tumor Drugs, Harbin 150081, China.
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin 150086, China; The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin 150081, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150080, China.
| |
Collapse
|
2
|
Yuan Z, Chen Y, Xin Y, Zhang Y, Dong Z, Wang J, Wang X, Yang G, Li S. Key role of the CSE/transsulfuration pathway in macrophage phenotypic change under iron overload. J Trace Elem Med Biol 2025; 88:127611. [PMID: 39914135 DOI: 10.1016/j.jtemb.2025.127611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/22/2024] [Accepted: 01/31/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND Iron homeostasis has a significant impact on the phenotypic transformation of macrophages and is implicated in various diseases. In this study, we evaluated the effect of cystathionine-gamma-lyase (CSE)/transsulfuration pathway in iron-overload induced macrophage phenotype change. METHODS The biochemical parameters, such as qRT-PCR, western blot, fluorescence staining, were assessed both in vitro and in vivo. RESULTS Iron overload disrupts iron metabolism and alters the expression of genes involved in iron transport, resulting in the polarization of macrophages towards the M1 phenotype and an alternating activation state of M2. Meanwhile, excessive iron led to an increase in lipid peroxidation levels and disrupted cysteine metabolism. By utilizing erastin to inhibit SLC7A11 activity and block exogenous cysteine uptake, we were able to observe the exacerbation of the proinflammatory state in macrophages under conditions of cysteine deprivation. The CSE/transsulfuration pathway, serves as the primary route for endogenous cysteine synthesis. In the presence of iron overload, the expression of CSE was upregulated and further enhanced by cysteine deprivation. Deletion of CSE in CSE-knockout mice exacerbated the inflammatory transition of iron-overloaded macrophages by impacting cysteine metabolism and ferritinophagy. CONCLUSION The CSE/transsulfuration pathway regulated macrophage phenotype change under iron-overload, which may offer novel insights into potential therapeutic strategies for iron overload-related disorders.
Collapse
Affiliation(s)
- Zhaoji Yuan
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong 250062, China
| | - Yuxuan Chen
- Department of Cell Biology, Shandong University, Jinan, Shandong 250012, China
| | - Yijun Xin
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong 250062, China
| | - Yong Zhang
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Zihao Dong
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Jianxu Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xiangdong Wang
- Department of Cell Biology, Shandong University, Jinan, Shandong 250012, China
| | - Guang Yang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Siying Li
- Metabolism and Disease Research Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong 250062, China.
| |
Collapse
|
3
|
Guo Q, Qian C, Wang X, Qian ZM. Transferrin receptors. Exp Mol Med 2025; 57:724-732. [PMID: 40263550 PMCID: PMC12045970 DOI: 10.1038/s12276-025-01436-x] [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: 10/31/2024] [Accepted: 01/17/2025] [Indexed: 04/24/2025] Open
Abstract
The transferrin receptor (TfR) is one of the key proteins involved in cellular iron uptake. TfR-mediated endocytosis of transferrin-bound iron is the major pathway for iron acquisition by most cells in the body. Over the past three decades, the studies on TfR have made significant progress, and also, our knowledge on cell iron uptake has greatly been improved. Here we focus on recent advances in the studies on TfR and a brief discussion of the structures and functions of four different types of TfR, namely TfR1 (transferrin receptor 1), TfR2 (transferrin receptor 2), TfR3 (glyceraldehyde-3-phosphate dehydrogenase) and TfR4 (cubilin). These proteins work in different cells or organs and at different times, ensuring that cells and tissues get the iron they need. Their normal expression and function are fundamental to the body's iron homeostasis.
Collapse
Affiliation(s)
- Qian Guo
- Laboratory of Drug Delivery, School of Medicine, Shanghai University, Shanghai, China.
| | - Christopher Qian
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xinyu Wang
- Laboratory of Drug Delivery, School of Medicine, Shanghai University, Shanghai, China
| | - Zhong-Ming Qian
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Van den Branden A, Opdebeeck B, Adriaensen S, Evenepoel P, Vanden Berghe T, Verhulst A. Intravenous iron treatment fuels chronic kidney disease-induced arterial media calcification in rats. J Pathol 2025; 265:172-183. [PMID: 39610372 PMCID: PMC11717497 DOI: 10.1002/path.6375] [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: 04/22/2024] [Revised: 09/09/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024]
Abstract
Arterial media calcification is a severe cardiovascular complication commonly manifesting in patients with chronic kidney disease (CKD). Patients with CKD frequently undergo intravenous iron therapy to address iron deficiency. Iron is suggested to be sequestered in vascular cells, potentially leading to oxidative (lipid) stress and cell death, which are recognized as key contributors to arterial calcification. The objective of this study was to investigate the effect of intravenous iron administration on CKD-induced arterial media calcification. Therefore, adenine-induced CKD rats were treated intravenously with iron and checked for arterial iron deposition and calcification, as well as for ferritin and lipid peroxidation markers. Additionally, arterial sections from patients with CKD who were dialysis dependent were analyzed for these parameters. This study showed that intravenous iron administration in CKD rats led to arterial iron deposition and a lipid peroxidation signature. CKD-induced arterial calcification was increased upon iron treatment and correlated with arterial iron accumulation and lipid peroxidation markers. Patients with CKD who were dialysis dependent showed arterial iron accumulation and elevated lipid peroxidation, but a direct correlation with arterial calcification was lacking. Taken together, iron treatment is suggested as a potential contributor to the calcification process, instead of being a predominant factor, thereby emphasizing the complexity of arterial calcification as a multifactorial disease. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Astrid Van den Branden
- Cell Death Signaling Lab, Department of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpAntwerpBelgium
| | - Britt Opdebeeck
- Cell Death Signaling Lab, Department of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpAntwerpBelgium
| | - Saar Adriaensen
- Cell Death Signaling Lab, Department of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpAntwerpBelgium
| | - Pieter Evenepoel
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and TransplantationKU LeuvenLeuvenBelgium
| | - Tom Vanden Berghe
- Cell Death Signaling Lab, Department of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpAntwerpBelgium
| | - Anja Verhulst
- Cell Death Signaling Lab, Department of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpAntwerpBelgium
| |
Collapse
|
5
|
Shen MQ, Guo Q, Li W, Qian ZM. Apolipoprotein E deficiency leads to the polarization of splenic macrophages towards M1 phenotype by increasing iron content. Genes Immun 2024; 25:381-388. [PMID: 39103538 DOI: 10.1038/s41435-024-00290-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/07/2024]
Abstract
Apolipoprotein E (ApoE) plays a crucial role in iron homeostasis in the body, while macrophages are the principal cells responsible for handling iron in mammals. However, it is unknown whether ApoE can affect the functional subtypes and the iron handling capacity of splenic macrophages (SM). Here, we investigated the effects of ApoE deficiency (ApoE-/-) on the polarization and iron content of SM and its potential mechanisms. ApoE-/- was found to induce a significant increase in the expressions of M1 marker genes CD86, IL-1β, IL-6, IL-12, TNF-α and iNOS and a reduction in M2 marker genes CD206, Arg-1, IL-10 and Ym-1 in SM of mice aged 28 weeks, Meanwhile, ApoE-/- caused a significant increase in iron content and expression of ferritin, transferrin receptor 1 (TfR1), iron regulatory protein 1 (IRP1) and heme oxygenase-1 (HO-1) and a reduction in ferroportin1 (Fpn1) in spleen and/or SM of mice aged 28 weeks. It was concluded that ApoE-/- can increase iron content through increased iron uptake mediated by TfR/ IRPs and decreased iron release mediated by Fpn1, leading to polarization of the SM to M1 phenotype.
Collapse
Affiliation(s)
- Meng-Qi Shen
- Institute of Translational and Precision Medicine, Nantong University, Nantong, Jiangsu, China
- School of Health Medicine, Nantong Polytechnic College, Nantong, China
| | - Qian Guo
- School of Medicine, Shanghai University, Shanghai, China.
| | - Wei Li
- Institute of Translational and Precision Medicine, Nantong University, Nantong, Jiangsu, China
| | - Zhong-Ming Qian
- Institute of Translational and Precision Medicine, Nantong University, Nantong, Jiangsu, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Chen Y, Cui Y, Li M, Xia M, Xiang Q, Mao Y, Li H, Chen J, Zeng W, Zheng X, Peng J, Dai X, Tang Z. A novel mechanism of ferroptosis inhibition-enhanced atherosclerotic plaque stability: YAP1 suppresses vascular smooth muscle cell ferroptosis through GLS1. FASEB J 2024; 38:e23850. [PMID: 39091212 DOI: 10.1096/fj.202401251r] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/05/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Abstract
Atherosclerosis is a leading cause of cardiovascular diseases (CVDs), often resulting in major adverse cardiovascular events (MACEs), such as myocardial infarction and stroke due to the rupture or erosion of vulnerable plaques. Ferroptosis, an iron-dependent form of cell death, has been implicated in the development of atherosclerosis. Despite its involvement in CVDs, the specific role of ferroptosis in atherosclerotic plaque stability remains unclear. In this study, we confirmed the presence of ferroptosis in unstable atherosclerotic plaques and demonstrated that the ferroptosis inhibitor ferrostatin-1 (Fer-1) stabilizes atherosclerotic plaques in apolipoprotein E knockout (Apoe-/-) mice. Using bioinformatic analysis combining RNA sequencing (RNA-seq) with single-cell RNA sequencing (scRNA-seq), we identified Yes-associated protein 1 (YAP1) as a potential key regulator of ferroptosis in vascular smooth muscle cells (VSMCs) of unstable plaques. In vitro, we found that YAP1 protects against oxidized low-density lipoprotein (oxLDL)-induced ferroptosis in VSMCs. Mechanistically, YAP1 exerts its anti-ferroptosis effects by regulating the expression of glutaminase 1 (GLS1) to promote the synthesis of glutamate (Glu) and glutathione (GSH). These findings establish a novel mechanism where the inhibition of ferroptosis promotes the stabilization of atherosclerotic plaques through the YAP1/GLS1 axis, attenuating VSMC ferroptosis. Thus, targeting the YAP1/GLS1 axis to suppress VSMC ferroptosis may represent a novel strategy for preventing and treating unstable atherosclerotic plaques.
Collapse
MESH Headings
- Ferroptosis
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- YAP-Signaling Proteins/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Humans
- Male
- Mice, Inbred C57BL
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Mice, Knockout
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Phenylenediamines/pharmacology
- Cyclohexylamines/pharmacology
- Apolipoproteins E/metabolism
- Apolipoproteins E/genetics
Collapse
Affiliation(s)
- Yanyu Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Yuting Cui
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
- Department of Pathophysiology, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Man Li
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Mengdie Xia
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Qiong Xiang
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Yu Mao
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Hengjuan Li
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Jialin Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Wen Zeng
- Shaoyang Branch of Key Laboratory for Arteriosclerology of Hunan Province, The Central Hospital of Shaoyang, Shaoyang, China
| | - Xilong Zheng
- Department of Biochemistry & Molecular Biology and Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Juan Peng
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
- Shaoyang Branch of Key Laboratory for Arteriosclerology of Hunan Province, The Central Hospital of Shaoyang, Shaoyang, China
| | - Xiaoyan Dai
- Clinical Research Institute, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhihan Tang
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, Institute of Cardiovascular Disease, University of South China, Hengyang, China
- Shaoyang Branch of Key Laboratory for Arteriosclerology of Hunan Province, The Central Hospital of Shaoyang, Shaoyang, China
| |
Collapse
|
7
|
Long Z, Luo Y, Yu M, Wang X, Zeng L, Yang K. Targeting ferroptosis: a new therapeutic opportunity for kidney diseases. Front Immunol 2024; 15:1435139. [PMID: 39021564 PMCID: PMC11251909 DOI: 10.3389/fimmu.2024.1435139] [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: 05/19/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Ferroptosis is a form of non-apoptotic regulated cell death (RCD) that depends on iron and is characterized by the accumulation of lipid peroxides to lethal levels. Ferroptosis involves multiple pathways including redox balance, iron regulation, mitochondrial function, and amino acid, lipid, and glycometabolism. Furthermore, various disease-related signaling pathways also play a role in regulating the process of iron oxidation. In recent years, with the emergence of the concept of ferroptosis and the in-depth study of its mechanisms, ferroptosis is closely associated with various biological conditions related to kidney diseases, including kidney organ development, aging, immunity, and cancer. This article reviews the development of the concept of ferroptosis, the mechanisms of ferroptosis (including GSH-GPX4, FSP1-CoQ1, DHODH-CoQ10, GCH1-BH4, and MBOAT1/2 pathways), and the latest research progress on its involvement in kidney diseases. It summarizes research on ferroptosis in kidney diseases within the frameworks of metabolism, reactive oxygen biology, and iron biology. The article introduces key regulatory factors and mechanisms of ferroptosis in kidney diseases, as well as important concepts and major open questions in ferroptosis and related natural compounds. It is hoped that in future research, further breakthroughs can be made in understanding the regulation mechanism of ferroptosis and utilizing ferroptosis to promote treatments for kidney diseases, such as acute kidney injury(AKI), chronic kidney disease (CKD), diabetic nephropathy(DN), and renal cell carcinoma. This paves the way for a new approach to research, prevent, and treat clinical kidney diseases.
Collapse
Affiliation(s)
- Zhiyong Long
- Department of Physical Medicine and Rehabilitation, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanfang Luo
- Department of Nephrology, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Min Yu
- Department of Physical Medicine and Rehabilitation, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyan Wang
- Department of Nephrology, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Liuting Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
8
|
Tan H, Liu L, Qi Y, Zhang D, Zhi Y, Li Y, Zhang H, Liu J. Atorvastatin Attenuates Endothelial Cell Injury in Atherosclerosis Through Inhibiting ACSL4-Mediated Ferroptosis. Cardiovasc Ther 2024; 2024:5522013. [PMID: 39742023 PMCID: PMC11211010 DOI: 10.1155/2024/5522013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/27/2024] [Accepted: 05/18/2024] [Indexed: 01/03/2025] Open
Abstract
Objective: This study is aimed at investigating the effects of atorvastatin (ATV) on endothelial cell injury in atherosclerosis (AS) through inhibiting acyl-CoA synthetase long-chain family member 4 (ACSL4)-mediated ferroptosis. Methods: Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to establish an in vitro model of AS. The cell viability, lactate dehydrogenase (LDH) release, apoptosis, and expression levels of apoptotic proteins were assessed. The levels of inflammatory factors and adhesion molecules were determined by ELISA and Western blot, respectively. Cellular iron content, lipid peroxidation, glutathione (GSH) levels, and lipid reactive oxygen species (ROS) were measured. ACSL4 overexpression was performed to investigate its role in ATV-mediated protection against ferroptosis. Results: ATV alleviated ox-LDL-induced HUVEC damage by restoring cell viability, reducing LDH levels, and inhibiting apoptosis. It also attenuated inflammation and adhesion by decreasing the levels of inflammatory factors TNF-α, IL-6, and IL-8, as well as adhesion molecules ICAM-1 and VCAM-1. ATV inhibited ferroptosis by regulating iron content, malondialdehyde (MDA) levels, ROS levels, and ACSL4 protein expression. Overexpression of ACSL4 (oe-ACSL4) hindered the protective effects of ATV on cell viability, antiapoptotic protein expression, LDH levels, apoptosis, and inflammatory factors. Conclusion: Our findings suggest that ATV attenuates endothelial cell injury in AS by inhibiting ACSL4-mediated ferroptosis. These results provide insights into the potential therapeutic strategies for the treatment of AS.
Collapse
Affiliation(s)
- Huilian Tan
- Department of CardiologyThe First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ling Liu
- Department of CardiologyThe First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yanchao Qi
- Department of CardiologyThe First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dahong Zhang
- Department of CardiologyThe First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yanchun Zhi
- Department of CardiologyThe First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yu Li
- Department of CardiologyThe First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huimin Zhang
- Department of CardiologyThe First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jun Liu
- Department of CardiologyThe First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
9
|
Haase T, Ludwig A, Stach A, Mohtashamdolatshahi A, Hauptmann R, Mundhenk L, Kratz H, Metzkow S, Kader A, Freise C, Mueller S, Stolzenburg N, Radon P, Liebl M, Wiekhorst F, Hamm B, Taupitz M, Schnorr J. Repeated Injection of Very Small Superparamagnetic Iron Oxide Particles (VSOPs) in Murine Atherosclerosis: A Safety Study. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:773. [PMID: 38727367 PMCID: PMC11085881 DOI: 10.3390/nano14090773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
Citrate-coated electrostatically stabilized very small superparamagnetic iron oxide particles (VSOPs) have been successfully tested as magnetic resonance angiography (MRA) contrast agents and are promising tools for molecular imaging of atherosclerosis. Their repeated use in the background of pre-existing hyperlipidemia and atherosclerosis has not yet been studied. This study aimed to investigate the effect of multiple intravenous injections of VSOPs in atherosclerotic mice. Taurine-formulated VSOPs (VSOP-T) were repeatedly intravenously injected at 100 µmol Fe/kg in apolipoprotein E-deficient (ApoE KO) mice with diet-induced atherosclerosis. Angiographic imaging was carried out by in vivo MRI. Magnetic particle spectrometry was used to detect tissue VSOP content, and tissue iron content was quantified photometrically. Pathological changes in organs, atherosclerotic plaque development, and expression of hepatic iron-related proteins were evaluated. VSOP-T enabled the angiographic imaging of heart and blood vessels with a blood half-life of one hour. Repeated intravenous injection led to VSOP deposition and iron accumulation in the liver and spleen without affecting liver and spleen pathology, expression of hepatic iron metabolism proteins, serum lipids, or atherosclerotic lesion formation. Repeated injections of VSOP-T doses sufficient for MRA analyses had no significant effects on plaque burden, steatohepatitis, and iron homeostasis in atherosclerotic mice. These findings underscore the safety of VSOP-T and support its further development as a contrast agent and molecular imaging tool.
Collapse
Affiliation(s)
- Tobias Haase
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Antje Ludwig
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10117 Berlin, Germany
| | - Anke Stach
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Charitéplatz 1, 10117 Berlin, Germany
| | - Azadeh Mohtashamdolatshahi
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Ralf Hauptmann
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Lars Mundhenk
- Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, 14163 Berlin, Germany;
| | - Harald Kratz
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Susanne Metzkow
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Avan Kader
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Christian Freise
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Susanne Mueller
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
- Charité 3R|Replace, Reduce, Refine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Nicola Stolzenburg
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Patricia Radon
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany; (P.R.); (M.L.); (F.W.)
| | - Maik Liebl
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany; (P.R.); (M.L.); (F.W.)
| | - Frank Wiekhorst
- Physikalisch-Technische Bundesanstalt, Abbestr. 2-12, 10587 Berlin, Germany; (P.R.); (M.L.); (F.W.)
| | - Bernd Hamm
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Matthias Taupitz
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Jörg Schnorr
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.L.); (A.M.); (R.H.); (H.K.); (S.M.); (A.K.); (C.F.); (S.M.); (N.S.); (B.H.); (M.T.); (J.S.)
- Department of Radiology, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
10
|
Xu X, Xu XD, Ma MQ, Liang Y, Cai YB, Zhu ZX, Xu T, Zhu L, Ren K. The mechanisms of ferroptosis and its role in atherosclerosis. Biomed Pharmacother 2024; 171:116112. [PMID: 38171246 DOI: 10.1016/j.biopha.2023.116112] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024] Open
Abstract
Ferroptosis is a newly identified form of non-apoptotic programmed cell death, characterized by the iron-dependent accumulation of lethal lipid reactive oxygen species (ROS) and peroxidation of membrane polyunsaturated fatty acid phospholipids (PUFA-PLs). Ferroptosis is unique among other cell death modalities in many aspects. It is initiated by excessive oxidative damage due to iron overload and lipid peroxidation and compromised antioxidant defense systems, including the system Xc-/ glutathione (GSH)/glutathione peroxidase 4 (GPX4) pathway and the GPX4-independent pathways. In the past ten years, ferroptosis was reported to play a critical role in the pathogenesis of various cardiovascular diseases, e.g., atherosclerosis (AS), arrhythmia, heart failure, diabetic cardiomyopathy, and myocardial ischemia-reperfusion injury. Studies have identified dysfunctional iron metabolism and abnormal expression profiles of ferroptosis-related factors, including iron, GSH, GPX4, ferroportin (FPN), and SLC7A11 (xCT), as critical indicators for atherogenesis. Moreover, ferroptosis in plaque cells, i.e., vascular endothelial cell (VEC), macrophage, and vascular smooth muscle cell (VSMC), positively correlate with atherosclerotic plaque development. Many macromolecules, drugs, Chinese herbs, and food extracts can inhibit the atherogenic process by suppressing the ferroptosis of plaque cells. In contrast, some ferroptosis inducers have significant pro-atherogenic effects. However, the mechanisms through which ferroptosis affects the progression of AS still need to be well-known. This review summarizes the molecular mechanisms of ferroptosis and their emerging role in AS, aimed at providing novel, promising druggable targets for anti-AS therapy.
Collapse
Affiliation(s)
- Xi Xu
- College of Nursing, Anhui University of Chinese Medicine, Hefei 230012, Anhui, PR China
| | - Xiao-Dan Xu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, PR China
| | - Meng-Qing Ma
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, PR China
| | - Yin Liang
- The First Clinical College, Guangdong Medical University, Zhanjiang 524000, Guangdong, PR China
| | - Yang-Bo Cai
- Division of Hepatobiliary and Pancreas Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570100, Hainan, PR China
| | - Zi-Xian Zhu
- Emergency and Trauma College, Hainan Medical University, Haikou 570100, Hainan, PR China
| | - Tao Xu
- College of Nursing, Anhui University of Chinese Medicine, Hefei 230012, Anhui, PR China
| | - Lin Zhu
- College of Nursing, Anhui University of Chinese Medicine, Hefei 230012, Anhui, PR China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, PR China.
| | - Kun Ren
- College of Nursing, Anhui University of Chinese Medicine, Hefei 230012, Anhui, PR China; Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou 570100, Hainan, PR China.
| |
Collapse
|
11
|
Halliwell B, Watt F, Minqin R. Iron and atherosclerosis: Lessons learned from rabbits relevant to human disease. Free Radic Biol Med 2023; 209:165-170. [PMID: 37852545 DOI: 10.1016/j.freeradbiomed.2023.10.383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023]
Abstract
The role of iron in promoting atherosclerosis, and hence the cardiovascular, neurodegenerative and other diseases that result from atherosclerosis, has been fiercely controversial. Many studies have been carried out on various rodent models of atherosclerosis, especially on apoE-knockout (apoE-/-) mice, which develop atherosclerosis more readily than normal mice. These apoE-/- mouse studies generally support a role for iron in atherosclerosis development, although there are conflicting results. The purpose of the current article is to describe studies on another animal model that is not genetically manipulated; New Zealand White (NZW) rabbits fed a high-cholesterol diet. This may be a better model than the apoE-/- mice for human atherosclerosis, although it has been given much less attention. Studies on NZW rabbits support the view that iron promotes atherosclerosis, although some uncertainties remain, which need to be resolved by further experimentation.
Collapse
Affiliation(s)
- Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Research Programme, National University of Singapore, Centre for Life Sciences, #05-01A, 28 Medical Drive, 117456, Singapore.
| | - Frank Watt
- Department of Physics, National University of Singapore, Faculty of Science, 2 Science Drive 3, Blk S12, Level 2, 117551, Singapore.
| | - Ren Minqin
- Department of Physics, National University of Singapore, Faculty of Science, 2 Science Drive 3, Blk S12, Level 2, 117551, Singapore.
| |
Collapse
|
12
|
Kontoghiorghes GJ. Iron Load Toxicity in Medicine: From Molecular and Cellular Aspects to Clinical Implications. Int J Mol Sci 2023; 24:12928. [PMID: 37629109 PMCID: PMC10454416 DOI: 10.3390/ijms241612928] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Iron is essential for all organisms and cells. Diseases of iron imbalance affect billions of patients, including those with iron overload and other forms of iron toxicity. Excess iron load is an adverse prognostic factor for all diseases and can cause serious organ damage and fatalities following chronic red blood cell transfusions in patients of many conditions, including hemoglobinopathies, myelodyspasia, and hematopoietic stem cell transplantation. Similar toxicity of excess body iron load but at a slower rate of disease progression is found in idiopathic haemochromatosis patients. Excess iron deposition in different regions of the brain with suspected toxicity has been identified by MRI T2* and similar methods in many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Based on its role as the major biological catalyst of free radical reactions and the Fenton reaction, iron has also been implicated in all diseases associated with free radical pathology and tissue damage. Furthermore, the recent discovery of ferroptosis, which is a cell death program based on free radical generation by iron and cell membrane lipid oxidation, sparked thousands of investigations and the association of iron with cardiac, kidney, liver, and many other diseases, including cancer and infections. The toxicity implications of iron in a labile, non-protein bound form and its complexes with dietary molecules such as vitamin C and drugs such as doxorubicin and other xenobiotic molecules in relation to carcinogenesis and other forms of toxicity are also discussed. In each case and form of iron toxicity, the mechanistic insights, diagnostic criteria, and molecular interactions are essential for the design of new and effective therapeutic interventions and of future targeted therapeutic strategies. In particular, this approach has been successful for the treatment of most iron loading conditions and especially for the transition of thalassemia from a fatal to a chronic disease due to new therapeutic protocols resulting in the complete elimination of iron overload and of iron toxicity.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, 3, Ammochostou Street, Limassol 3021, Cyprus
| |
Collapse
|