1
|
Xia Y, Yang Q, Zhang L, Chen K, Yu X, Li Y, Ge J, Xie C, Shen Y, Tong J. Blue light induced ferroptosis in retinal damage via iron overload-associated oxidative stress. J Environ Sci (China) 2025; 155:221-234. [PMID: 40246460 DOI: 10.1016/j.jes.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2025]
Abstract
The issue of light pollution has garnered increased attention recently, largely due to the widespread use of electronic devices. Blue light (BL) holds the highest energy level among visible light and has been extensively researched for its potential to cause damage to the retina. Ferroptosis, a recently identified form of programmed cell death form, has been linked to retinal diseases. However, the connection between BL-induced retinal damage and ferroptosis remains elusive. This study aims to investigate the involvement of ferroptosis in retinal damage under BL exposure and its underlying mechanism. In this study, a mouse retinal damage model and cultured ARPE-19 cells exposed to BL were employed. Various techniques including Haematoxylin-eosin staining, fundus photography, immunostaining, and transmission electron microscopy were employed to examine retinal structure and morphology changes resulting from BL exposure. To identify ferroptosis levels in vitro, we employed DCFH-DA, C11-BODIPY 581/591, and FeRhoNox™-1 probes. Additionally, real-time PCR and western blotting techniques were used to uncover potential targets in BL-induced ferroptosis. Our study showed that BL exposure can result in iron overload, oxidative stress, evidenced by increased markers TFR1, ACSL4, HO-1 and decreased expression level of SOD2, CAT and ferroptosis-associated gene of GPX4. Interestingly, we found that Deferoxamine mesylate, a compound capable of chelating excess Fe2+ caused by BL, effectively mitigated lipid peroxidation, and alleviated retinal damage both in vivo and in vitro. The discoveries will advance our knowledge of BL-induced retinal damage.
Collapse
Affiliation(s)
- Yutong Xia
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China
| | - Qianjie Yang
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China
| | - Liyue Zhang
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China
| | - Kuangqi Chen
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China; Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Jinan 250299, China; School of Ophthalmology, Shandong First Medical University, Jinan 250118, China
| | - Xin Yu
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China
| | - Yanqing Li
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China
| | - Jiayun Ge
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China
| | - Chen Xie
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China.
| | - Ye Shen
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China.
| | - Jianping Tong
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
2
|
Li MH, Yang Y, Dong QQ, Tao H, Lu C, Yang JJ. Novel epitranscriptomic and epigenetic therapeutic strategies and targets for ferroptosis in liver fibrosis. Eur J Pharmacol 2025; 996:177344. [PMID: 40015597 DOI: 10.1016/j.ejphar.2025.177344] [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: 11/17/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
Liver fibrosis is characterized by an excessive accumulation of extracellular matrix (ECM) and the activation of hepatic stellate cells (HSCs), which are influenced by epitranscriptomic and epigenetic factors. Recent advancements in epigenetic and epitranscriptomic research have revealed new opportunities for therapeutic interventions, particularly through the regulation of ferroptosis, a type of programmed cell death that is specifically linked to iron-dependent lipid peroxidation. In the context of liver fibrosis, a progressive scarring process that can progress to cirrhosis and ultimately end-stage liver disease, targeting these regulatory mechanisms to modulate ferroptosis presents a promising therapeutic strategy. This review aims to consolidate current knowledge on the epigenetic and epitranscriptomic control of ferroptosis and investigate its potential implications for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ming-Hui Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yang Yang
- Department of General Surgery, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
| | - Qi-Qi Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Chao Lu
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
3
|
Li Y, Zhang L, Zhang Q, Zhang Y, Pan S, Zhao H, Zhang L. HSPB1 suppresses oxLDL-induced vascular smooth muscle cell ferroptosis by inhibiting DPP4. Arch Biochem Biophys 2025; 768:110400. [PMID: 40132776 DOI: 10.1016/j.abb.2025.110400] [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: 12/17/2024] [Revised: 02/25/2025] [Accepted: 03/22/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Atherosclerosis is the major pathological basis of cardiovascular diseases. Vascular smooth muscle cell (VSMC) dysfunction and death induced by oxidized low-density lipoprotein (oxLDL) play a key role in atherosclerosis. Ferroptosis is a novel iron-dependent lipid peroxidation regulated cell death, which is implicated in atherosclerosis. However, whether oxLDL induces VSMC ferroptosis and the specific mechanism is unclear. METHODS To determine the effects of oxLDL on VSMC ferroptosis, LDH activity, MDA and Fe2+ content, glutathione peroxidase 4 (GPX4) expression and GPX enzyme activity were assayed. The level of lipid peroxidation was detected by C11 BODIPY fluorescence staining. RT-qPCR and Western blot were used to detect the mRNA and protein expressions of heat shock protein B1 (HSPB1), dipeptidyl peptidase 4 (DPP4) and nuclear factor kappa-B (NF-κB). The siRNAs, plasmids and Val-boropro were utilized to explore the roles of HSPB1/NF-κB/DPP4 in oxLDL-induced VSMC ferroptosis. RESULTS oxLDL increased LDH activity, Fe2+ content, lipid peroxidation and MDA content in VSMCs, which were inhibited by ferroptosis inhibitors Lip-1 and DFO. Moreover, oxLDL reduced GPX4 protein expression and GPX enzyme activity, indicating that oxLDL induces VSMC ferroptosis. Notably, HSPB1 inhibited oxLDL-induced VSMC ferroptosis by reducing the accumulation of Fe2+ and lipid peroxidation and increasing GPX4 expression and activity. In addition, HSPB1 suppressed oxLDL-induced VSMC ferroptosis by inhibiting DPP4 through NF-κB. Furthermore, Val-boropro could rescue oxLDL-induced ferroptosis in VSMCs with HSPB1 knockdown by inhibiting DPP4. CONCLUSIONS This study reveals for the first time that HSPB1 suppresses oxLDL-induced VSMC ferroptosis by inhibiting DPP4 through NF-κB, providing new strategies for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yi Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qi Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuke Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shuang Pan
- Department of Physiology, School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Huanhuan Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
4
|
Wen J, Li L, Ou D, Li J, Yang Y, Duan L, Zhang X, Zhu Y, Hao J, Tong Y. Higenamine protects against doxorubicin-induced heart failure by attenuating ferroptosis via modulating the Nrf2/GPX4 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156670. [PMID: 40220414 DOI: 10.1016/j.phymed.2025.156670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/24/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Higenamine (HG), a benzylisoquinoline alkaloid in Aconiti Lateralis Radix Praeparata (ALRP), has cardioprotective effects. Prior research indicated its potential anti-heart failure (HF) function, yet the molecular mechanism remained elusive. PURPOSE This study aimed to explore the underlying mechanism of HG against doxorubicin (DOX)-induced HF via an integrated approach involving gut microbiota, untargeted metabolomics, network pharmacology, and molecular biology. METHODS DOX was employed to induce HF in rats and H9c2 cardiomyocytes injury models. Cardiac injury was assessed using hemodynamic indices, cardiac injury biomarkers, and oxidative stress markers. Cell counting kit-8 (CCK-8) method and high-content analysis were used to investigate the effects of HG on the cell proliferation, morphology and mitochondrial function of H9c2 cardiomyocytes. 16S rDNA sequencing analysis, untargeted metabolomics, and network pharmacology were performed to identify the multi-target and multi-pathway mechanisms of HG in treating HF. Furthermore, reverse transcription quantitative polymerase chain reaction (RT-qPCR), immunohistochemistry, and Western Blotting was used to investigate its intervention on the nuclear factor erythroid 2-related factor 2 (Nrf2)/glutathione peroxidase 4 (GPX4) ferroptosis pathway. RESULTS HG alleviated DOX-mediated myocardial injury by enhancing cardiac and mitochondrial function, reducing oxidative stress levels, and promoting cell proliferation. Effects of HG on changes in the gut microbiota of rats is characterized by a low abundance of Firmicutes and Proteobacteria, along with a high abundance of Bacteroidetes and Actinobacteria, indicating an improvement in DOX-induced dysbiosis. Untargeted metabolomics combined with network pharmacology showed that HG exerted anti-HF effects by regulating eight metabolites, eight pathways, and interacting with ferroptosis-related targets. Molecular biology studies revealed its cardioprotective effects via regulating the Nrf2/GPX4 ferroptosis pathway. CONCLUSION HG could inhibit ferroptosis and protect against HF by regulating the Nrf2/GPX4-mediated "mitochondrial-ferroptosis" pathway, offering a potential treatment strategy for HF.
Collapse
Affiliation(s)
- Jianxia Wen
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Lu Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Dinglin Ou
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Jianling Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yi Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Liting Duan
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming, China
| | - Xinghai Zhang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yichan Zhu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Junjie Hao
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming, China.
| | - Yuling Tong
- School of Medicine and Food, Sichuan Vocational College of Health and Rehabilitation, Zigong, China.
| |
Collapse
|
5
|
Xian X, Zhao X, Zhou X, Liu H, Li C, Wu X, Chen Y, Ye K, Yang H, Li M, Yan J, Zhang X. Honokiol attenuates oxidative stress and vascular calcification via the upregulation of heme oxygenase-1 in chronic kidney disease. Toxicol Appl Pharmacol 2025; 499:117318. [PMID: 40194744 DOI: 10.1016/j.taap.2025.117318] [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: 01/22/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
Vascular calcification (VC) is a common complication of chronic kidney disease (CKD), with oxidative stress identified as a key contributor to VC progression. Honokiol (HKL), a biphenolic compound derived from plants, has been found to be effective in treating various models of cardiovascular disease through the mitigation of oxidative stress. However, its effects on VC remain unexplored. To elucidate the effects of HKL on VC, a CKD rat model, a vitamin D3-overload-induced mouse model of vascular calcification, and a high-phosphate-induced human vascular smooth muscle cell (VSMC) calcification model were established. Calcification levels were assessed using alizarin red staining, calcium quantification, and western blotting of osteogenic markers. Oxidative stress was assessed by measuring reactive oxygen species. Furthermore, transcriptome sequencing was employed to identify molecules and pathways affected by HKL. HKL was found to significantly reduce calcification in both in vivo and in vitro models. It also mitigated oxidative stress induced by high phosphate in human VSMCs. Mechanistically, HKL upregulated heme oxygenase-1 (HMOX-1), thereby inhibiting oxidative stress and reducing calcification. Pharmacological inhibition of HMOX-1 counteracted the protective effect of HKL against vascular calcification. In summary, the findings suggest that HKL ameliorates VC by upregulating HMOX-1 and decreasing oxidative stress.
Collapse
MESH Headings
- Oxidative Stress/drug effects
- Animals
- Vascular Calcification/drug therapy
- Vascular Calcification/enzymology
- Vascular Calcification/prevention & control
- Vascular Calcification/pathology
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/enzymology
- Biphenyl Compounds/pharmacology
- Biphenyl Compounds/therapeutic use
- Heme Oxygenase-1/metabolism
- Heme Oxygenase-1/genetics
- Lignans/pharmacology
- Lignans/therapeutic use
- Humans
- Male
- Up-Regulation/drug effects
- Mice
- Rats
- Mice, Inbred C57BL
- Rats, Sprague-Dawley
- Disease Models, Animal
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Antioxidants/pharmacology
- Cells, Cultured
- Allyl Compounds
- Phenols
Collapse
Affiliation(s)
- Xuemin Xian
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Xin Zhao
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Xingchen Zhou
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Hanfang Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Changxi Li
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Xinquan Wu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Yuhang Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Keyue Ye
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Hongwei Yang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Mingxi Li
- Department of Pathophysiology, Zhongshan Medical School, Sun Yat-Sen University, Guangzhou, China
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China.
| | - Xiuli Zhang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China.
| |
Collapse
|
6
|
Zhang M, Sha Y, Wang J, Qi H, Shi P, Liu Y, Jiang M, Ba L, Liu Y, Cao Y, Zhang Q, Sun H. Inhibition of ULK1 attenuates ferroptosis-mediated cardiac hypertrophy via HMGA2/METTL14/SLC7A11 axis in mice. Eur J Pharmacol 2025; 995:177416. [PMID: 39993699 DOI: 10.1016/j.ejphar.2025.177416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 01/24/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
UNC-51-like kinase 1 (ULK1), a primary serine/threonine kinase, is implicated in diverse pathophysiological processes. Previous findings have linked ULK1-dependent autophagy to cardiac hypertrophy. Our study further explored the functional role and molecular mechanisms of ULK1 in non-autophagic signaling pathways. Notably, ULK1 expression was significantly elevated in both transverse aortic constriction (TAC)-induced hypertrophic mouse hearts and Angiotensin II (Ang II)-treated cardiomyocytes, suggesting an increased sensitivity to hypertrophic stimuli potentially mediated by ULK1-induced ferroptosis in hypertrophic cardiomyocytes. Treatment with the ferroptosis inhibitor ferrostatin-1 (Fer-1) effectively reduced ULK1-induced cardiomyocyte hypertrophy and ferroptosis. Proteomic analysis identified the upregulation of transcription factor high mobility group A2 (HMGA2) as a key mechanism in this ferroptotic process. Elevated HMGA2 levels exacerbated ferroptosis, evidenced by increased cell death, lipid peroxidation, ROS production, and reduced GPX4 expression. Furthermore, HMGA2 was shown to promote cardiomyocyte ferroptosis via binding to methyltransferase-like 14 (METTL14), which in turn enhanced ferroptosis in cardiomyocytes through solute carrier family 7 member 11 (SLC7A11) m6A modification. In vivo, a delivery system using neutrophil membrane (NM)-coated mesoporous silica nanoparticles (MSN) was developed to inhibit cardiac hypertrophy, underscoring the therapeutic potential of targeting ULK1. Overall, this study demonstrates that ULK1 promotes cardiac hypertrophy through HMGA2/METTL14/SLC7A11 axis-mediated cardiomyocyte ferroptosis, suggesting a novel therapeutic approach for cardiac hypertrophy.
Collapse
Affiliation(s)
- Meitian Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yuetong Sha
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Jiaxin Wang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yongsheng Liu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Man Jiang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Lina Ba
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yuhang Liu
- Department of Physiology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Qianhui Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China.
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China.
| |
Collapse
|
7
|
Jiang Y, Jiang K, Sun P, Liu Y, Nie H. Oroxylin A ameliorates non-alcoholic fatty liver disease by modulating oxidative stress and ferroptosis through the Nrf2 pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2025:159628. [PMID: 40368273 DOI: 10.1016/j.bbalip.2025.159628] [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: 01/09/2025] [Revised: 04/26/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent and progressive liver disorder posing a global health challenge. Oroxylin A, a naturally occurring flavonoid, with a broad spectrum of pharmacological activities. This study aimed to explore the therapeutic potential of oroxylin A and unravel its molecular mechanisms in mitigating high-fat diet (HFD)-induced NAFLD in murine models. Wild-type (WT) and nuclear factor erythroid 2-related factor 2 knockout (Nrf2-/-) mice were administered a HFD to generate in vivo models, while free fatty acids-treated HepG2 cells served as the in vitro model. To investigate the effects of oroxylin A, serum and liver biochemical markers, hepatic histology, lipid metabolism, and oxidative stress were assessed in a NAFLD mouse model. The underlying mechanisms of oroxylin A were further explored through Western blotting, immunohistochemistry, and immunofluorescence analysis. Oroxylin A mitigated hepatic steatosis and injury by reducing liver index, AST, ALT, TG, and TC levels, improving histology, and restoring lipid metabolism. Glucose and insulin tolerance tests demonstrated improved glucose homeostasis and insulin sensitivity. Moreover, oroxylin A suppressed inflammation, apoptosis, and fibrosis, while enhancing antioxidant defenses, and improving mitochondrial function. Mechanistically, oroxylin A activated the Keap1/Nrf2/GPX4/SLC7A11 axis, upregulating Nrf2 and HO-1. These effects were abolished in Nrf2-/- mice. In vitro results were consistent, and molecular docking, dynamics simulations, and CETSA confirmed its direct Keap1 binding. Oroxylin A protects against NAFLD by modulating the Nrf2 pathway, reducing oxidative stress and ferroptosis, making it a promising candidate for clinical NAFLD therapy.
Collapse
Affiliation(s)
- Yuzi Jiang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China
| | - Kangwei Jiang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China
| | - Peilin Sun
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China
| | - Yuan Liu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China
| | - Hongming Nie
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| |
Collapse
|
8
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Mechanism of ferroptosis in heart failure: The role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and intervention strategies. Ageing Res Rev 2025:102770. [PMID: 40360081 DOI: 10.1016/j.arr.2025.102770] [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: 12/03/2024] [Revised: 04/29/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
The ferroptosis of cardiomyocytes has been recognized as the core pathological mechanism of heart failure. During the evolution of cardiovascular diseases, the accumulation of angiotensin II and advanced glycation end products can lead to the excessive activation of the RAGE/TLR4-JNK1/2 pathway, which subsequently triggers ferritinophagy, clockophagy, and enhanced p53 activity, ultimately leading to cardiomyocyte ferroptosis. It is evident that deeply unraveling the specific mechanisms in this field and comprehensively evaluating potential drugs and therapeutic strategies targeting this pathway is crucial for improving the status of cardiomyocyte ferroptosis. However, our current understanding of this pathway's specific molecular biological mechanisms in the process of cardiomyocyte ferroptosis remains limited. In light of this, this paper first comprehensively reviews the historical context of ferroptosis research, compares the similarities and differences between ferroptosis and other standard modes of cell death, elucidates the core mechanisms of ferroptosis and its close connection with heart failure, aiming to establish a basic cognitive framework for readers on ferroptosis and its role in heart failure. Subsequently, the paper delves into the pivotal role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and its intricate molecular biological regulatory network. Furthermore, it systematically integrates various therapeutic approaches aimed at inhibiting RAGE, TLR4, and JNK1/2 activity to alleviate cardiomyocyte ferroptosis, encompassing RNA interference technology, gene knockout techniques, small molecule inhibitors, natural active ingredients, as well as traditional Chinese and Western medicines, with the ultimate goal of forging new avenues and strategies for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| |
Collapse
|
9
|
Li Q, Li C, Liu X, Guo Z, Li X, Zhang X. The key role of Piezo1 channels in ferroptosis after spinal cord injury and the therapeutic potential of Piezo1 inhibitors. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:132-140. [PMID: 40339662 DOI: 10.1016/j.pbiomolbio.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/07/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Ferroptosis has been confirmed to be one of the key mechanisms of neuronal injury and dysfunction after spinal cord injury (SCI). Mechanical stresses such as deformation, compression, and stretching not only directly cause physical damage to spinal cord tissue at the moment of SCI, but also promote the development of ferroptosis through various pathways. However, the mechanism of ferroptosis after SCI remains unclear, which hinders the development of therapeutic methods. OBJECTIVE This article aims to review the key mechanisms by which mechanical stress affects ferroptosis after SCI, including its impact on the structure and function of the endoplasmic reticulum (ER) and mitochondria, its role in triggering inflammatory responses, and its activation of mechanosensitive channels. Special emphasis is placed on the role of Piezo1 channels, which are key factors in cell mechanosensation and ion homeostasis regulation. The review explores how Piezo1 channels are upregulated by mechanical stress after SCI and participate in the ferroptosis process by mediating ion flow and other mechanisms. CONCLUSIONS Inhibiting Piezo1 channels may be a potential therapeutic strategy for SCI. This review summarizes the therapeutic potential of Piezo1 inhibitors by sorting out existing studies, hoping to provide a theoretical basis for effective therapeutic strategies targeting ferroptosis after SCI.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinyu Liu
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Zixuan Guo
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinxin Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
10
|
Zhu Z, Liu H, Feng L, Lu L, Zhu J, Liang Q, Lan Z, Ye Y, Wang S, Chen A, Yan J. Loss of ADAMTS5 promotes vascular calcification via versican/integrin β1/FAK signal. Atherosclerosis 2025; 404:119190. [PMID: 40215897 DOI: 10.1016/j.atherosclerosis.2025.119190] [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: 12/02/2024] [Revised: 03/29/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Extracellular matrix (ECM) proteases have been closely linked to the pathogenesis of vascular calcification. A disintegrin and metalloprotease with thrombospondin motifs-5 (ADAMTS5) is an ECM-degrading enzyme involved in ECM remodeling. Versican, a critical ECM component in the arteries, can be proteolytically cleaved by ADAMTS5 and activates integrin β1. However, whether ADAMTS5 is involved in the regulation of the pathogenesis of vascular calcification remains unclear. This study investigates the regulatory role of ADAMTS5 in vascular calcification and its mechanistic link to versican-integrin β1/FAK signaling. METHODS AND RESULTS Western blot, immunofluorescence, and immunohistochemistry analysis revealed that ADAMTS5 expression was significantly downregulated in rat and human vascular smooth muscle cells (VSMCs), as well as in rat and human arteries during vascular calcification. In addition, both pharmacological inhibition of ADAMTS5 and knockdown of ADAMTS5 by siRNA significantly aggravated mineral deposition in rat and human VSMCs under osteogenic conditions. Moreover, adenovirus-mediated ADAMTS5 overexpression markedly attenuated calcification of VSMCs and aortic calcification in rats with chronic kidney disease. Furthermore, inhibition of ADAMTS5 promoted aortic calcification in VitD3-overloaded mice. Mechanistically, overexpression of ADAMTS5 significantly reduced versican protein levels, and inhibited integrin β1 and FAK phosphorylation in rat VSMCs, but increased versikine protein levels. Moreover, either knockdown of versican or pharmacological inhibition of FAK phosphorylation repressed VSMC calcification mediated by loss of ADAMTS5. CONCLUSIONS We have demonstrated for the first time that ADAMTS5 deficiency promotes versican accumulation and activates integrin β1/FAK signaling. These findings suggest ADAMTS5 as a potential therapeutic target for vascular calcification.
Collapse
MESH Headings
- Animals
- Versicans/metabolism
- Humans
- ADAMTS5 Protein/genetics
- ADAMTS5 Protein/metabolism
- ADAMTS5 Protein/deficiency
- Signal Transduction
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Vascular Calcification/enzymology
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Integrin beta1/metabolism
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Male
- Cells, Cultured
- Disease Models, Animal
- Focal Adhesion Kinase 1/metabolism
- Mice, Inbred C57BL
- Rats, Sprague-Dawley
- Rats
- Phosphorylation
- Mice
- Aortic Diseases/enzymology
- Aortic Diseases/genetics
- Aortic Diseases/pathology
Collapse
Affiliation(s)
- Zhenyu Zhu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China; Department of Cardiology, Tongde Hospital of Zhejiang Province, PR China
| | - Hao Liu
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, PR China
| | - Liyun Feng
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Lihe Lu
- Department of Pathophysiolgy, Zhongshan Medical School, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Jiahui Zhu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510665, PR China
| | - Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - Siyi Wang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China
| | - An Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China.
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, 510280, PR China.
| |
Collapse
|
11
|
Luo JQ, Wang L, Liao ZQ, Lu BX, Luo CY, He HY, Ou Yang ZH, Duan SB, He SH, Wei AY, Zhang HB. Adipose stem cells ameliorate erectile dysfunction in diabetes mellitus rats by attenuating ferroptosis through NRP1 with SLC7A11 interaction. Free Radic Biol Med 2025; 232:40-55. [PMID: 40020883 DOI: 10.1016/j.freeradbiomed.2025.02.041] [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: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Adipose stem cells (ADSCs) have garneVred increasing attention for their potential to treat diabetes mellitus erectile dysfunction (DMED), but the underlying molecular mechanisms remain unclear. The aim of this study was to identify and investigate the key cytokines and mechanisms by which ADSCs improve erectile function in DMED rats. METHODS We performed in vivo and in vitro assays, including rat erectile function assessment, cell co-culture, cytokine microarray screening and co-immunoprecipitation to investigate the role of ADSCs in improving erectile function in DMED rats. RESULTS Our analyses confirmed the occurrence of ferroptosis in the corpus cavernosum of DMED rats, while ADSCs treatment significantly restored erectile function and improved relevant indicators of ferroptosis. In vitro assays further indicated that corpus cavernosum smooth muscle cells (CCSMCs) co-cultured with ADSCs exhibited enhanced resistance to ferroptosis, with notably lower levels of cytoplasmic and lipid reactive oxygen species compared to the ferroptosis inducer Erastin-treated group. Mechanistic studies revealed that Neuropilin 1 (NRP1) may be a key molecule in ADSCs to improve erectile function in DMED rats. Furthermore, NRP1 in CCSMCs can interact with solute carrier family 7 member 11 (SLC7A11) to enhance the function of the glutamate-cysteine countertransport (Xc-) system and ferroptosis resistance in CCSMCs. CONCLUSION In conclusion, our findings indicate that NRP1 is a key molecule for ADSCs treatment to alleviate ferroptosis and improve erectile function in DMED rats, providing a promising target for DMED treatment and prognosis.
Collapse
Affiliation(s)
- Jun-Qi Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Li Wang
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zi-Qi Liao
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Bing-Xin Lu
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Cai-Yu Luo
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Hai-Yang He
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Zhi-Han Ou Yang
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Song-Bo Duan
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China
| | - Shu-Hua He
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China.
| | - An-Yang Wei
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China; Department of Urology, Foresea Life Insurance Guangzhou General Hospital, Guangzhou, Guangdong, 511340, China.
| | - Hai-Bo Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, North of Guangzhou Avenue 1838#, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
12
|
Liu F, Liang Q, Li L, Gong Y, Li M, Feng L, Chen A, Ye Y, Lan Z, Li Y, Ou JS, Lu L, Yan J. Thrombospondin-1 binds to integrin β3 to inhibit vascular calcification through suppression of NF-κB pathway. J Pathol 2025; 266:109-123. [PMID: 40084742 DOI: 10.1002/path.6417] [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: 06/16/2024] [Revised: 01/01/2025] [Accepted: 02/12/2025] [Indexed: 03/16/2025]
Abstract
Vascular calcification is an important risk factor related to all-cause mortality of cardiovascular events in patients with chronic kidney disease (CKD). Vascular extracellular matrix (ECM) proteins have been demonstrated to regulate vascular calcification. ECM protein thrombospondin 1 (THBS1/TSP-1) plays a critical role in the regulation of vascular diseases. However, whether THBS1 is involved in vascular calcification in CKD patients remains unclear. In this study, RNA sequencing datasets from the Gene Expression Omnibus (GEO) database GSE146638 showed that THBS1 was upregulated in the aortas of CKD rats. Enzyme-linked immunosorbent assay (elisa) revealed that serum THBS1 levels were increased in CKD patients with thoracic calcification. Western blotting and immunofluorescence analysis showed that THBS1 expression was increased in calcified vascular smooth muscle cells (VSMCs) and arteries. THBS1 knockdown exacerbated rat VSMC calcification induced by high phosphorus and calcium, as shown by Alizarin red staining and calcium content assays. Conversely, THBS1 overexpression attenuated VSMC calcification and abdominal aortic calcification in rats with CKD. Moreover, addition of recombinant THBS1 protein inhibited calcification of VSMCS and human arterial rings. Smooth muscle cell-specific knockout of THBS1 mice treated with vitamin D3 displayed aggravated aortic calcification. Mechanistically, the protein-protein interaction database STRING (http://string-db.org/) analysis and coimmunoprecipitation assays revealed THBS1 bound to integrin β3. Reduction of integrin β3 levels abrogated the protective effect of THBS1 on vascular calcification. RNA-seq analysis revealed that THBS1 overexpression modulated the nuclear factor-kappa B (NF-κB) signaling pathway. Of note, the inhibitory effect of THBS1 overexpression on the NF-κB signal was abolished by knockdown of integrin β3. In conclusion, THBS1 interacts with integrin β3 to inhibit vascular calcification through suppression of NF-κB signal, suggesting a promising therapeutic target for vascular calcification in CKD. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Thrombospondin 1/metabolism
- Thrombospondin 1/genetics
- Animals
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Vascular Calcification/prevention & control
- Humans
- NF-kappa B/metabolism
- Signal Transduction
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Integrin beta3/metabolism
- Integrin beta3/genetics
- Male
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Rats
- Mice
- Rats, Sprague-Dawley
- Disease Models, Animal
- Cells, Cultured
- Mice, Inbred C57BL
- Female
- Middle Aged
Collapse
Affiliation(s)
- Fang Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, PR China
| | - Li Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, PR China
| | - Yuan Gong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Mingxi Li
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, PR China
| | - Liyun Feng
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - An Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Yining Li
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Jing-Song Ou
- Division of Cardiac Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Lihe Lu
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, PR China
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| |
Collapse
|
13
|
Mavaddatiyan L, Naeini S, Khodabandeh S, Hosseini F, Skelton RP, Azizi V, Talkhabi M. Exploring the association between aging, ferroptosis, and common age-related diseases. Arch Gerontol Geriatr 2025; 135:105877. [PMID: 40339241 DOI: 10.1016/j.archger.2025.105877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
Aging is a natural biological process that is characterized by the progressive decline in physiological functions and an increased vulnerability to age-related diseases. The aging process is driven by different cell and molecular mechanisms. It has recently been shown that aging is associated with heightened vulnerability to ferroptosis (an intracellular iron-dependent form of programmed cell death). This susceptibility arises from various factors including oxidative stress, impaired antioxidant defences, and dysregulated iron homeostasis. The progressive decline in cellular antioxidant capacity and the accumulation of damaged components contribute to the increased susceptibility of aging cells to ferroptosis. Dysregulation of key regulators involved in ferroptosis, such as glutathione peroxidase 4 (GPX4), iron regulatory proteins, and lipid metabolism enzymes, further exacerbates this vulnerability. The decline in cellular defence mechanisms against ferroptosis during aging contributes to the accumulation of damaged cells and tissues, ultimately resulting in the manifestation of age-related diseases. Understanding the intricate relevance between aging and ferroptosis holds significant potential for developing strategies to counteract the detrimental effects of aging and age-related diseases. This will subsequently act to mitigate the negative consequences of aging and improving overall health in the elderly population. This review aims to clarify the relationship between aging and ferroptosis, and explores the underlying mechanisms and implications for age-related disorders, including neurodegenerative, cardiovascular, and neoplastic diseases. We also discuss the accumulating evidence suggesting that the imbalance of redox homeostasis and perturbations in iron metabolism contribute to the age-associated vulnerability to ferroptosis.
Collapse
Affiliation(s)
- Laleh Mavaddatiyan
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - SaghiHakimi Naeini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Sara Khodabandeh
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Hosseini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - RhysJ P Skelton
- Flinders Medical Centre, Department of Ophthalmology, Bedford Park, Australia
| | - Vahid Azizi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mahmood Talkhabi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
14
|
Chen K, Gu Y, Zhu X, Deng Q, Lei C. Neuronal ferroptosis in intracerebral hemorrhage shows gene regulation. Gene 2025; 946:149327. [PMID: 39952486 DOI: 10.1016/j.gene.2025.149327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Affiliation(s)
- Keyang Chen
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Yu Gu
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Xiaoyan Zhu
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Qionghua Deng
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Chunyan Lei
- From the First Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China.
| |
Collapse
|
15
|
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] [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
|
16
|
Massie PL, Garcia M, Decker A, Liu R, MazloumiBakhshayesh M, Kulkarni D, Justus MP, Gallardo J, Abrums A, Markle K, Pace C, Campen M, Clark RM. Essential and Non-Essential Metals and Metalloids and Their Role in Atherosclerosis. Cardiovasc Toxicol 2025:10.1007/s12012-025-09998-y. [PMID: 40251456 DOI: 10.1007/s12012-025-09998-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/10/2025] [Indexed: 04/20/2025]
Abstract
Peripheral arterial disease (PAD) is becoming more prevalent in the aging developed world and can have significant functional impacts on patients. There is a recent recognition that environmental toxicants such as circulating metals and metalloids may contribute to the pathogenesis of atherosclerotic disease, but the mechanisms are complex. While the broad toxic biologic effects of metals in human systems have been extensively reviewed, the role of non-essential exposure and essential metal aberrancy in PAD specifically is less frequently discussed. This review of the literature describes current scientific knowledge regarding the individual roles several major metals and metalloids play in atherogenesis and highlights areas where a dearth of data exist. The roles of lead (Pb), arsenic (As), cadmium (Cd), iron (Fe), copper (Cu), selenium (Se) are included. Contemporary outcomes of therapeutic trials aimed at chelation therapy of circulating metals to impact cardiovascular outcomes are also discussed. This review highlights the supported notion of differential metal presence within peripheral plaques themselves, although distinguishing their roles within these plaques requires further illumination.
Collapse
Affiliation(s)
- Pierce L Massie
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Marcus Garcia
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Aerlin Decker
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Rui Liu
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Milad MazloumiBakhshayesh
- Department of Biomedical Engineering, School of Engineering, University of New Mexico, Albuquerque, USA
| | - Deepali Kulkarni
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Matthew P Justus
- Department of Biomedical Engineering, School of Engineering, University of New Mexico, Albuquerque, USA
| | - Jorge Gallardo
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Avalon Abrums
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Kristin Markle
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Carolyn Pace
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA
| | - Matthew Campen
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Ross M Clark
- Department of Surgery, University of New Mexico School of Medicine, MSC10-5610, Albuquerque, NM, 87131, USA.
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, USA.
| |
Collapse
|
17
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
18
|
Jia L, Qu P, Zhao Y, Bai L, Ren H, Cheng A, Ma Z, Ding C, Deng Y, Kong L, Zhao Y, Rom O, Chen Y, Alam N, Cao W, Zhai S, Zheng Z, Hu Z, Wang L, Chen Y, Zhao S, Zhang J, Fan J, Chen YE, Liu E. Tripeptide DT-109 (Gly-Gly-Leu) attenuates atherosclerosis and vascular calcification in nonhuman primates. Signal Transduct Target Ther 2025; 10:122. [PMID: 40195303 PMCID: PMC11977015 DOI: 10.1038/s41392-025-02201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Advanced atherosclerotic lesions and vascular calcification substantially increase the risk of cardiovascular events. However, effective strategies for preventing or treating advanced atherosclerosis and calcification are currently lacking. This study investigated the efficacy of DT-109 (Gly-Gly-Leu) in attenuating atherosclerosis and calcification in nonhuman primates, exploring its broader therapeutic potential. In this study, twenty male cynomolgus monkeys were administered a cholesterol-rich diet ad libitum for 10 months. Then, the animals were treated either orally with DT-109 (150 mg/kg/day) or a vehicle (H2O) for 5 months while continuing on the same diet. Plasma lipid levels were measured monthly and at the end of the experiment, pathological examinations of the aortas and coronary arteries and RNA sequencing of the coronary arteries were performed. To explore possible molecular mechanisms, the effects of DT-109 on smooth muscle cells (SMCs) were examined in vitro. We found that DT-109 administration significantly suppressed atherosclerotic lesion formation in both the aorta and coronary arteries. Pathological examinations revealed that DT-109 treatment reduced lesional macrophage content and calcification. RNA sequencing analysis showed that DT-109 treatment significantly downregulated the pro-inflammatory factors NLRP3, AIM2, and CASP1, the oxidative stress factors NCF2 and NCF4, and the osteogenic factors RUNX2, COL1A1, MMP2, and MMP9, while simultaneously upregulating the expression of the SMCs contraction markers ACTA2, CNN1, and TAGLN. Furthermore, DT-109 inhibited SMC calcification and NLRP3 inflammasome activation in vitro. These results demonstrate that DT-109 effectively suppresses both atherosclerosis and calcification. These findings, in conjunction with insights from our previous studies, position DT-109 as a novel multifaceted therapeutic agent for cardiovascular diseases.
Collapse
Affiliation(s)
- Linying Jia
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Pengxiang Qu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yang Zhao
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Liang Bai
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Honghao Ren
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Ao Cheng
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Zeyao Ma
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Cheng Ding
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yongjie Deng
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Lingxuan Kong
- Department of Biostatistics, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Ying Zhao
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Oren Rom
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
- Department of Pathology and Translational Pathobiology, Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Yajie Chen
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, 529000, China
| | - Naqash Alam
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Wenbin Cao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Sixue Zhai
- Department of Imaging, the Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, 710038, China
| | - Zuowen Zheng
- Spring Biological Technology Development Co., Ltd, Fangchenggang, Guangxi, 538000, China
| | - Zhi Hu
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Lu Wang
- Department of Biostatistics, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Yabing Chen
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University; Research Department, Portland Veterans Affairs Medical Center, Portland, OR, 97239, USA
| | - Sihai Zhao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Jianglin Fan
- Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, 529000, China.
| | - Y Eugene Chen
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, Shaanxi, 710061, China.
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
19
|
Xiao W, Wang F, Wang X, Wang N. A mitochondrion-targeted poly(N-isopropylacrylamide-coacrylic acid) nanohydrogel with a fluorescent bioprobe for ferrous ion imaging in vitro and in vivo. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 330:125725. [PMID: 39809013 DOI: 10.1016/j.saa.2025.125725] [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: 08/20/2024] [Revised: 12/15/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
An imbalance in iron homeostasis contributes to mitochondrial dysfunction, which is closely linked to the pathogenesis of various diseases. Herein, we developed a nanosensor for detecting mitochondrial ferrous ions in vitro and in vivo. A poly(N-isopropylacrylamine)-coacrylic acid nanohydrogel was synthesized, and ferrous ions were detected using the fluorescent probe FeRhonox-1 embedded within it. (3-Carboxypropyl)-triphenylphosphonium bromide was chemically conjugated to the hydrogel matrix to enable mitochondrial targeting. The developed nanosensor showed a narrow particle size distribution, high sensitivity and selectivity for ferrous ions, and low cytotoxicity, enabling the nanosensor to sense and image ferrous ions in mitochondria with high spatial resolution. Changes in ferrous ion concentrations in human umbilical vein endothelial cells were measured and imaged after lipopolysaccharide (LPS) or iron dextran treatment. Moreover, the nanosensor was successfully used for ferrous ion imaging in live mice. The in vivo results showed that LPS injection induced the accumulation of mitochondrial ferrous ions. The proposed nanosensor could serve as a powerful tool for monitoring ferrous ions in mitochondria, providing strong support for studying disorders of iron metabolism.
Collapse
Affiliation(s)
- Wenlong Xiao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310007, China; Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, China
| | - Fang Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, China
| | - Xuchen Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, China
| | - Nani Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310007, China; Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang 310007, China.
| |
Collapse
|
20
|
Wang L, ChenLiu Z, Wang D, Tang D. Cross-talks of GSH, mitochondria, RNA m6A modification, NRF2, and p53 between ferroptosis and cuproptosis in HCC: A review. Int J Biol Macromol 2025; 302:140523. [PMID: 39894098 DOI: 10.1016/j.ijbiomac.2025.140523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with high morbidity and mortality, as well as poor prognosis. Therefore, it is imperative to explore alternative therapeutic targets for HCC treatment. Ferroptosis and cuproptosis have recently been identified as metal-dependent cell death mechanisms that play significant roles in HCC treatment. This study identified potential cross-talk between ferroptosis and cuproptosis, including the common hub glutathione, common site of occurrence, mitochondria, shared epigenetic modification mode, RNA N6 methyladenosine modification, mutual inhibitor, nuclear factor erythroid 2-related factor 2, and dual regulator, p53. These findings provide a theoretical foundation for the joint induction of HCC cell death and effective inhibition of HCC progression. However, some immune cells are susceptible to ferroptosis or cuproptosis, which may impair or enhance anti-cancer immune function. We propose strategies to target specific targets molecules such as tripartite motif containing 25, ferroptosis suppressor protein 1, and peroxisome proliferator-activated receptor gamma or exploit the unique acidic environment surrounding cancer cells to precisely induce ferroptosis in cancer cells. This approach aims to advance the development of precision medicine for HCC treatment.
Collapse
Affiliation(s)
- Leihan Wang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Zhenni ChenLiu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China.
| |
Collapse
|
21
|
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
|
22
|
Pei X, Cui F, Chen Y, Yang Z, Xie Z, Wen Y. miR-214-3p Promotes ox-LDL-Induced Macrophages Ferroptosis and Inflammation via GPX4. J Inflamm Res 2025; 18:3937-3950. [PMID: 40125091 PMCID: PMC11927573 DOI: 10.2147/jir.s507076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose Atherosclerosis (AS) is a chronic inflammatory disease caused by the dysregulation of lipid metabolism. It has been established that oxidized low-density lipoprotein (ox-LDL)-induced macrophage inflammation and ferroptosis play important roles in AS. However, the mechanism by which ox-LDL induces inflammation in macrophages requires further investigation. Materials and Methods A foam cell model derived from ox-LDL-induced macrophages was constructed to study its mechanism of action. The levels of interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α were evaluated using an Enzyme-Linked Immunosorbent Assay (ELISA). Oil Red O staining was used to detect intracellular lipid accumulation levels. Lactate dehydrogenase (LDH), malondialdehyde (MDA), reactive oxygen species (ROS), and Fe2+ levels were assessed. Dual-luciferase and RNA-binding protein immunoprecipitation (RIP) experiments validated the binding relationship between microRNA (miR)-214-3p and glutathione peroxidase 4 (GPX4). Results The levels of IL-6, IL-1β, and TNF-α were significantly increased in ox-LDL-induced macrophages, accompanied by increased lipid accumulation, indicating the promotion of foam cell formation. Additionally, ox-LDL increased LDH, MDA, ROS, and Fe2+. The expression of miR-214-3p positively correlated with ox-LDL concentration in macrophages. Treatment with an miR-214-3p inhibitor reduces lipid accumulation, inflammatory responses, and ferroptosis in macrophages. Dual-luciferase and RIP experiments confirmed that miR-214-3p regulates GPX4 transcription. Silenced GPX4 reversed the inflammatory effects of the miR-214-3p inhibitor on ox-LDL-induced macrophages. Low GPX4 expression also increased lipid accumulation and ferroptosis in macrophages. Conclusion miR-214-3p promotes macrophage ferroptosis and inflammation induced by ox-LDL. This mechanism may be associated with miR-214-3p-induced GPX4 expression, which underscores the therapeutic significance of targeting macrophage inflammation and ferroptosis in addressing AS.
Collapse
Affiliation(s)
- Xueliang Pei
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Facai Cui
- Clinical Laboratory, Henan Provincial People’s Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Yu Chen
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Zhiyuan Yang
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Zhouliang Xie
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| | - Yongjin Wen
- Department of Cardiovascular Surgery, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, People’s Republic of China
| |
Collapse
|
23
|
Xiong Z, Hu X, Wang R, Li C, Cheng H, Zhao W, Shen Y, Wang L, Li W, Zhu X, Ba Y. Jingtian granule alleviates adenine-induced renal fibrosis in mice through SIRT3-Mediated deacetylation of P53. Front Pharmacol 2025; 16:1526414. [PMID: 40144655 PMCID: PMC11936886 DOI: 10.3389/fphar.2025.1526414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/11/2025] [Indexed: 03/28/2025] Open
Abstract
Background Renal fibrosis is a hallmark and the final outcome of chronic kidney disease (CKD). Jingtian Granule (JT), a traditional formula used in the clinical treatment of CKD for many years. However, the mechanism of action of JT against renal interstitial fibrosis remain unknown. Objective This study aimed to explore the potential effects and mechanisms of JT on adenine - diet - induced CKD in mice. Methods Renal interstitial fibrosis was induced in mice by adenine - diet and treated with JT. Renal function was assessed by measuring blood urea nitrogen and serum creatinine levels. Masson's staining and type I collagen expression were used to evaluate renal collagen deposition. RNA sequencing was used to analyze the expression levels of mRNA in mouse kidney samples after JT treatment. The levels of glutathione (GSH) and malondialdehyde (MDA) were measured to assess lipid peroxidation in the kidneys. Iron metabolism levels were detected by Prussian blue staining and measurement of iron content. The protein levels of SIRT3, P53, glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11) were detected by Western blot. Subsequently, under the premise of SIRT3 knockout, renal function, fibrosis level, iron metabolism level, and lipid peroxidation level were detected, and mitochondrial damage was observed by transmission electron microscope (TEM). In addition, human proximal tubule epithelial cells (HK - 2) were treated with Erastin to induce ferroptosis, followed by exposure to JT. The levels of reactive oxygen species (ROS) were detected. Results JT significantly reduced collagen deposition in the kidneys. RNA sequencing identified 20 mRNAs that were differentially expressed in response to JT treatment. Bioinformatics analysis revealed that SIRT3 was a key mRNA regulated by JT. JT activated SIRT3 in fibrotic kidneys to inhibit the acetylation of P53. Under the premise of SIRT3 knockout, JT did not show significant therapeutic effects in inhibiting ferroptosis and fibrosis. In vitro experiments also showed that JT promoted the downregulation of ROS. Conclusion SIRT3 is the key ferroptosis - related mRNA regulated by JT. The ability of JT to modulate the SIRT3/P53 signaling pathway may be a viable approach for the treatment of renal interstitial fibrosis.
Collapse
Affiliation(s)
- Zhili Xiong
- Hubei University of Chinese Medicine, Wuhan, China
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Sizhen Laboratory, Wuhan, China
- Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, China
| | - Xinyu Hu
- Hubei University of Chinese Medicine, Wuhan, China
| | - Rui Wang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Sizhen Laboratory, Wuhan, China
- Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, China
| | - Chengyin Li
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Sizhen Laboratory, Wuhan, China
- Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, China
| | - Huanbo Cheng
- Hubei University of Chinese Medicine, Wuhan, China
| | - Wei Zhao
- Hubei University of Chinese Medicine, Wuhan, China
| | - Yinfeng Shen
- Hubei University of Chinese Medicine, Wuhan, China
| | - Linqun Wang
- Hubei University of Chinese Medicine, Wuhan, China
| | - Weinan Li
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Sizhen Laboratory, Wuhan, China
- Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, China
| | - Xiaoyun Zhu
- Hubei University of Chinese Medicine, Wuhan, China
| | - Yuanming Ba
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Sizhen Laboratory, Wuhan, China
- Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
24
|
Lin G, Liu Q, Xie C, Ding K, Mo G, Zeng L, Zhang F, Liu R, Lu L, Hong W, Mao Y, Su H, Li S. Upregulated FSP1 by GPD1/1L mediated lipid droplet accumulation enhances ferroptosis resistance and peritoneal metastasis in gastric cancer. Cell Commun Signal 2025; 23:132. [PMID: 40075460 PMCID: PMC11899195 DOI: 10.1186/s12964-025-02126-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
To successfully metastasize, cancer cells must evade detachment induced cell death, known as anoikis. Unraveling the mechanisms that gastric cancer (GC) circumvent anoikis and achieve peritoneal metastasis especially during unanchored growth, could significantly improve patient outcomes. Our study reveals that GC cells exhibit increased lipid peroxidation, MDA production, and cell death during suspension culture, which can be mitigated by the intervention with liproxstatin-1 and ferrostatin-1. We discovered that oleic acid (OA) or adipocytes stimulate lipid accumulation in GC cells, thereby inhibiting lipid peroxidation and cell death. Lipid mass spectrometry confirmed an upregulation of triglyceride synthesis, indicating that the accumulation of lipid droplet may confer resistance to ferroptosis during suspension growth. In vitro assays demonstrated that OA not only induces lipid droplet accumulation but also upregulates the expression of ferroptosis suppressor protein 1 (FSP1), a process that can be abrogated by the double knockout of GPD1/1L genes. Additionally, we have demonstrated that a decrease in the ubiquitination of FSP1 in GC cells upon lipid droplet accumulation, as well as silencing or pharmacological targeting FSP1, promotes ferroptosis and disrupts the peritoneal metastatic potential of GC cells. Collectively, our findings highlight the potential of FSP1 as a promising therapeutic target for metastatic gastric cancer.
Collapse
Affiliation(s)
- Guoliang Lin
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Qingnan Liu
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Chengjie Xie
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Ke Ding
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Guanghua Mo
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Lu Zeng
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Fan Zhang
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - RuiXuan Liu
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Lei Lu
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yuling Mao
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Haibo Su
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China.
| | - Shuai Li
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511436, PR China.
| |
Collapse
|
25
|
Wang Y, Su H, Lin X, Dai C, Cheng Q, Deng Z, Yang Y, Pu X. 1,25-(OH) 2D 3 improves SD rats high-altitude pulmonary edema by inhibiting ferroptosis and ferritinophagy in alveolar epithelial cells. J Steroid Biochem Mol Biol 2025; 247:106663. [PMID: 39681240 DOI: 10.1016/j.jsbmb.2024.106663] [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: 08/18/2024] [Revised: 11/21/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND This study investigates the protective effects and potential mechanisms of 1,25-(OH)2D3 against high-altitude pulmonary edema (HAPE). METHODS Hypoxia-induced rats were administered 1,25-(OH)2D3 for 24, 48, and 72 hours, and we observed lung tissue injury and pulmonary edema. Immunohistochemistry (IHC) and Western blot analyses were employed to analyze the expression of markers associated with ferroptosis and ferritinophagy in rat lungs. Metabolomics analysis was conducted to investigate changes in serum lipid metabolites. We validated the mechanism of action of 1,25-(OH)2D3 in type II alveolar epithelial cells induced by hypoxia. RESULTS Our results demonstrated that hypoxic exposure significantly altered sodium-water transport in the lungs, leading to edema formation. The degree of pulmonary edema was most pronounced at 48 hours of hypoxi. Treatment with 1,25-(OH)2D3 improved lung function and reduced the degree of pulmonary edema in hypoxic rats. Hypoxia-induced increases in 4-HNE and MDA levels in the lungs, along with iron accumulation, were observed. Hypoxia also resulted in elevated levels of NCOA4, LC3Ⅱ, and FTH1 proteins in the lungs. Furthermore, treatment with 1,25-(OH)2D3 significantly inhibited ferroptosis and ferritinophagy in the lungs after hypoxia. The levels of lipid metabolites, such as L-Aspartic acid and L-Fucose, were significantly elevated in the serum of hypoxic rats. After 1,25-(OH)2D3 treatment, these levels exhibited a significant reduction. CONCLUSION In hypoxic type II alveolar epithelial cells, 1,25-(OH)2D3 improved hypoxia-induced sodium-water transport, ferroptosis, and ferritinophagy, which were reversed by the autophagy agonist Rapamycin.By modulating ferroptosis and ferritinophagy, 1,25-(OH)2D3 mitigated the deleterious effects of hypoxia on pulmonary function.
Collapse
Affiliation(s)
- Yaxuan Wang
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Hong Su
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Xue Lin
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China.
| | - Chongyang Dai
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Qian Cheng
- Qinghai University, Xining, Qinghai Province 810016, China.
| | | | - Yangyang Yang
- Qinghai University, Xining, Qinghai Province 810016, China.
| | - Xiaoyan Pu
- Qinghai University, Xining, Qinghai Province 810016, China.
| |
Collapse
|
26
|
Huang Y, Ru Q, Ruan H, Zhang J, Wang Y, Wang C, Chen C, Yu D, Luo J, Yang M. Changyanning tablet alleviates Crohn's disease by inhibiting GPX4-mediated ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119415. [PMID: 39870334 DOI: 10.1016/j.jep.2025.119415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Changyanning tablets (CYN) are a marketed traditional Chinese medicine composed of Diijincao (Euphorbia humifusa Willd.), Jinmaoercao (Hedyotis chrysotricha (Palib.) Merr.), Zhangshugen (root of Cinnamomum camphora (L.) J.Presl), Xiangru (Elsholtzia ciliate (Thunb.) Hyl.), and Fengxiangshuye (leaf of Liquidambar formosana Hance). They possess the functions of clearing heat, removing dampness, and regulating qi. CYN is used for the treatment of diarrhea and dysentery caused by damp heat in the large intestine, with symptoms such as diarrhea, or stools with pus and blood, tenesmus, abdominal pain and distension, acute and chronic gastroenteritis, diarrhea, bacterial diarrhea, and indigestion in children. AIM OF THE STUDY This study aims to explore the intervention effects of CYN on Crohn's disease (CD) and its potential mechanisms. MATERIALS AND METHODS The therapeutic effect and potential mechanism of CYN on CD were investigated based on the 2,4,6-Trinitrobenzenesulfonic acid solution (TNBS)-induced rat model. In vivo and in vitro experiments confirmed that CYN can alleviate CD by inhibiting GPX4-mediated ferroptosis. siRNA was used to knock down GPX4 for reverse validation. Finally, active components of CYN inhibiting ferroptosis were identified using UPLC-MS and the RSL3-induced HCoEpiC ferroptosis cell model. RESULTS CYN significantly improved ferroptosis-related indicators (GSH, MDA, GPX4, and SLC7A11) in the colons of TNBS-induced CD rats. Screening with three ferroptosis inducers (RSL3, FINO2, and erastin) revealed that CYN was most effective against RSL3 (a ferroptosis inducer targeting GPX4)-induced apoptosis. Subsequently, the resistance effect of CYN on RSL3-induced ferroptosis was confirmed in vitro. Further in vivo experiments showed that CYN alleviated local CD-like intestinal injury induced by RSL3 enema. siRNA knockdown of GPX4 in HCoEpiC cells further validated GPX4 as major target of CYN in inhibiting ferroptosis. Finally, UPLC-MS and in vitro experiments identified rutin, rosmarinic acid, and kaempferol-3-O-sophoroside as key active components of CYN for inhibiting ferroptosis. CONCLUSIONS CYN alleviates CD by inhibiting GPX4-mediated ferroptosis, highlighting its clinical potential for treating CD and enhancing the understanding of the pathogenic and therapeutic mechanisms associated with CD.
Collapse
Affiliation(s)
- Ying Huang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Qing Ru
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Haonan Ruan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Jing Zhang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Yunyun Wang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Chuang Wang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Changyong Chen
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Defa Yu
- Jiangxi Kangenbei Traditional Chinese Medicine Co., Ltd., Shangrao, 334400, China.
| | - Jiaoyang Luo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Meihua Yang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
27
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [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: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
28
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
29
|
Yang B, Yang K, Chen Y, Li Q, Chen J, Li S, Wu Y. Exposure of A2E to blue light promotes ferroptosis in the retinal pigment epithelium. Cell Mol Biol Lett 2025; 30:22. [PMID: 39984833 PMCID: PMC11846388 DOI: 10.1186/s11658-025-00700-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/06/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Age-dependent accumulation of lipofuscin in the retinal pigment epithelium (RPE) is closely related to the etiology of autosomal recessive Stargardt's disease (STGD1) and dry age-related macular degeneration (AMD). N-retinylidene-N-retinylethanolamine (A2E) is a leading component of RPE lipofuscin that is highly susceptible to blue light. Ferroptosis is an iron-dependent form of non-apoptotic cell death characterized by the accumulation of lipid peroxides to a lethal level, which plays an important role in retinal diseases. However, it remains unknown whether A2E functions as a physiological trigger for eliciting blue light-induced ferroptosis of RPE cells. METHODS A2E-loaded RPE cells and Abca4-/-Rdh8-/- mice were exposed to blue light, respectively. Western blotting, immunofluorescence staining, reactive oxygen species (ROS) staining, intracellular iron staining, lipid peroxidation staining, fundus imaging, optical coherence tomography (OCT), hematoxylin-eosin (HE) staining, and electroretinography (ERG) were utilized to elucidate the role of blue light in A2E induced ferroptosis in the RPE and its potential mechanisms. RESULTS Exposure of A2E to blue light promoted ferroptotic cell death in RPE cells by elevating ferrous ion (Fe2+) levels and inhibiting the solute carrier family 7 membrane 11 (SLC7A11)-glutathione (GSH)-glutathione peroxidase 4 (GPX4) axis. GPX4 inactivation and ROS generated by Fe2+ overload and GSH depletion precipitated lipid peroxidation and subsequent ferroptosis in A2E-containing RPE cells upon exposure to blue light. In addition to GSH supplement, repressing either Fe2+ by deferiprone (DFP) or lipid peroxidation with ferrostatin-1 (Fer-1) significantly protected RPE cells against ferroptosis caused by blue light illumination of A2E. Abca4-/-Rdh8-/- mice featured by an accelerated deposition of A2E in the RPE is an animal model for STGD1 and dry AMD. It was observed that ferroptosis was indeed present in the RPE of Abca4-/-Rdh8-/- mice following exposure to blue light. Notably, alleviating ferroptosis by intraperitoneally injected Fer-1 effectively rescued retinal function and ameliorated RPE/photoreceptor degeneration in blue light-exposed Abca4-/-Rdh8-/- mice. CONCLUSIONS Our results suggest the importance of blue light in A2E-mediated ferroptosis in the RPE, and deeply broaden the understanding of mechanisms underlying RPE atrophy arising from lipofuscin accumulation in STGD1 and dry AMD.
Collapse
Affiliation(s)
- Bo Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Kunhuan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yuling Chen
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Qingjian Li
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, Guangdong, China
| | - Shiying Li
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China.
| | - Yalin Wu
- Department of Ophthalmology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, Fujian, China.
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, Guangdong, China.
| |
Collapse
|
30
|
Huang S, Sun J, Shen C, He G. Dietary and nutritional interventions for human diseases: their modulatory effects on ferroptosis. Food Funct 2025; 16:1186-1204. [PMID: 39866046 DOI: 10.1039/d4fo05606j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
A balanced diet is essential for maintaining human health. Increasing evidence suggests that dietary and nutritional interventions contribute to disease management and are associated with reduced healthcare costs and economic burden. Ferroptosis, a novel type of regulated cell death (RCD) driven by lipid peroxidation, has been shown to be involved in various pathological conditions, including diabetes, ischemia/reperfusion (I/R) injury, inflammation-related diseases, and cancer. Therefore, specifically targeting the uncontrolled ferroptosis process may offer new therapeutic opportunities. Of note, certain interventions, such as small-molecule compounds, natural products, herbal medicines, and non-pharmacological approaches, have been reported to prevent and treat multiple human diseases by reversing the dysregulation of ferroptosis. In this review, we present the key molecular mechanisms that regulate ferroptosis. Importantly, interventions targeting ferroptosis are summarized from the perspective of dietary patterns, food and nutrients. By understanding these advances, innovative ideas can be provided for individualized dietary interventions and treatment strategies.
Collapse
Affiliation(s)
- Shiqiong Huang
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha 410000, China.
| | - Ji Sun
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha 410000, China.
| | - Chaozan Shen
- Department of Clinical Pharmacy, The Second People's Hospital of Huaihua, Huaihua 418000, China.
| | - Gefei He
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha 410000, China.
| |
Collapse
|
31
|
Xiong X, Li W, Yu C, Qiu M, Zhang Z, Hu C, Zhu S, Yang L, Pen H, Song X, Chen J, Xia B, Han S, Yang C. SMURF1-Induced Ubiquitination of FTH1 Disrupts Iron Homeostasis and Suppresses Myogenesis. Int J Mol Sci 2025; 26:1390. [PMID: 39941157 PMCID: PMC11818545 DOI: 10.3390/ijms26031390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Ferritin heavy chain 1 (FTH1) is pivotal in the storage, release, and utilization of iron, plays a crucial role in the ferroptosis pathway, and exerts significant impacts on various diseases. Iron influences skeletal muscle development and health by promoting cell growth, ensuring energy metabolism and ATP synthesis, maintaining oxygen supply, and facilitating protein synthesis. However, the precise molecular mechanisms underlying iron's regulation of skeletal muscle growth and development remain elusive. In this study, we demonstrated that the conditional knockout (cKO) of FTH1 in skeletal muscle results in muscle atrophy and impaired exercise endurance. In vitro studies using FTH1 cKO myoblasts revealed notable decreases in GSH concentrations, elevated levels of lipid peroxidation, and the substantial accumulation of Fe2+, collectively implying the induction of ferroptosis. Mechanistically, E3 ubiquitin-protein ligase SMURF1 (SMURF1) acts as an E3 ubiquitin ligase for FTH1, thereby facilitating the ubiquitination and subsequent degradation of FTH1. Consequently, this activation of the ferroptosis pathway by SMURF1 impedes myoblast differentiation into myotubes. This study identifies FTH1 as a novel regulator of muscle cell differentiation and skeletal muscle development, implicating its potential significance in maintaining skeletal muscle health through the regulation of iron homeostasis.
Collapse
Affiliation(s)
- Xia Xiong
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Wen Li
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Chunlin Yu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Mohan Qiu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Zengrong Zhang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Chenming Hu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Shiliang Zhu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Li Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Han Pen
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Xiaoyan Song
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Jialei Chen
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Bo Xia
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| | - Shunshun Han
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; (X.X.); (W.L.); (C.Y.); (M.Q.); (Z.Z.); (C.H.); (S.Z.); (L.Y.); (H.P.); (X.S.); (J.C.); (B.X.)
| |
Collapse
|
32
|
Wang W, Chen J, Zhan L, Zou H, Wang L, Guo M, Gao H, Xu J, Wu W. Iron and ferroptosis in kidney disease: molecular and metabolic mechanisms. Front Immunol 2025; 16:1531577. [PMID: 39975561 PMCID: PMC11835690 DOI: 10.3389/fimmu.2025.1531577] [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: 11/20/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Maintaining iron homeostasis is necessary for kidney functioning. There is more and more research indicating that kidney disease is often caused by iron imbalance. Over the past decade, ferroptosis' role in mediating the development and progression of renal disorders, such as acute kidney injury (renal ischemia-reperfusion injury, drug-induced acute kidney injury, severe acute pancreatitis induced acute kidney injury and sepsis-associated acute kidney injury), chronic kidney disease (diabetic nephropathy, renal fibrosis, autosomal dominant polycystic kidney disease) and renal cell carcinoma, has come into focus. Thus, knowing kidney iron metabolism and ferroptosis regulation may enhance disease therapy. In this review, we discuss the metabolic and molecular mechanisms of iron signaling and ferroptosis in kidney disease. We also explore the possible targets of ferroptosis in the therapy of renal illness, as well as their existing limitations and future strategies.
Collapse
Affiliation(s)
- Wenjie Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jingdi Chen
- Department of orthopedics, The Airborne Military Hospital, Wuhan, Hubei, China
| | - Liying Zhan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Handong Zou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lu Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mengmeng Guo
- The First Clinical College of Wuhan University, Wuhan, Hubei, China
| | - Hang Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
33
|
Zuo R, Cao B, Kong L, Wang F, Li S, Shan H, Guan J, Kang Q. MiR-370-3p regulate TLR4/SLC7A11/GPX4 to alleviate the progression of glucocorticoids-induced osteonecrosis of the femoral head by promoting osteogenesis and suppressing ferroptosis. J Orthop Translat 2025. [DOI: 10.1016/j.jot.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2025] Open
|
34
|
Xu J, Chen S, Liu D, Zhang Q, Luo T, Zhu J, Zhou L, Lin Y, Pan H, Chen Y, Zhao Q, Wang T, Andrea S, Nashan B, Stefan TG, Cai C, Cui J, He X, Guo Z. Suppression of Hepatocyte Ferroptosis via USP19-Mediated Deubiquitination of SLC7A11 in Ischemia-Free Liver Transplantation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406200. [PMID: 39574305 PMCID: PMC11809379 DOI: 10.1002/advs.202406200] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/17/2024] [Indexed: 02/11/2025]
Abstract
Ischemia-free liver transplantation (IFLT) is developed as a novel clinical approach to avoid ischemia-reperfusion injury (IRI). This study aims to identify the most distinguished programmed cell death pathway in grafts undergoing IFLT versus conventional liver transplantation (CLT) and to explore the underlying mechanism. Ferroptosis is the most distinct programmed cell death form between IFLT and CLT grafts. Among various cell death inhibitors, the ferroptosis inhibitor (Ferrostain-1) is the most effective one to prevent hepatocytes from damage induced by oxygen deprivation/reoxygenation (OGD/R). Hepatocyte ferroptosis is significantly alleviated in IFLT versus CLT grafts in both human beings and pigs. Ubiquitination enzyme screening identifies augmented amounts of ubiquitin-specific protease 19 (USP19) in IFLT versus CLT grafts. The upregulation of USP19 in the grafts is correlated with reduced pathological Suzuki's score, lower post-transplant peak liver enzyme level, and less early allograft dysfunction in liver transplant recipients. USP19 overexpression mitigates post-transplant liver injury in mice. Mechanistically, USP19 inhibits the degradation of solute carrier family 7 member 11 (SLC7A11) by removing its K63-linked ubiquitin chains. Notably, USP19 overexpression reduces ferroptosis and IRI in a SLC7A11-dependent manner in mice. Collectively, USP19-mediated suppression of hepatocyte ferroptosis via deubiquitinating SLC7A11 is a key mechanism by which IFLT abrogates graft IRI.
Collapse
Affiliation(s)
- Jinghong Xu
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of AnesthesiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Shirui Chen
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Di Liu
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Qi Zhang
- Department of Thyroid and Breast SurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230601China
| | - Tao Luo
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Jiaxing Zhu
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Liang Zhou
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Yuan Lin
- Department of PathologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Hongyu Pan
- Department of PathologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Yichao Chen
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Qiang Zhao
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Tielong Wang
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Schlegel Andrea
- Transplantation CenterDigestive Disease and Surgery Institute and Department of ImmunologyLerner Research Institute, Cleveland ClinicClevelandOhio44113USA
| | - Björn Nashan
- Organ Transplant CenterThe First Affiliated Hospital of the University of Science and Technology of ChinaHefeiAnhui230001China
| | - Tullius G. Stefan
- Division of Transplant SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Changjie Cai
- Department of Critical CareThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Jun Cui
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xiaoshun He
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Zhiyong Guo
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
- NHC Key Laboratory of Assisted CirculationSun Yat‐sen UniversityGuangzhouGuangdong510080China
| |
Collapse
|
35
|
Bedolla N, Liu L, Liu X, Xie Q, Ren Y. Ursolic acid enhances radiosensitivity in esophageal squamous cell carcinoma by modulating p53/SLC7A11/GPX4 pathway-mediated ferroptosis. Toxicon 2025; 255:108233. [PMID: 39788329 DOI: 10.1016/j.toxicon.2025.108233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
BACKGROUND Radiotherapy is essential for the management of esophageal squamous cell carcinoma (ESCC). However, ESCC cells are highly susceptible to developing resistance to radiotherapy, leading to poor prognosis. Ursolic acid (UA) is a herbal monomer, has multiple medicinal benefits like anti-tumor. The impact of UA on the sensitivity of ESCC cells to radiotherapy is currently unclear. METHODS The impact of UA and ionizing radiation (IR) on the viability of TE-1 and KYSE30 cells was assessed by the MTT assay. EdU staining, flow cytometry, clone formation, Wound healing and Transwell assay detected the biological properties of ESCC cells. FerroOrange, DCFH-DA, and kits to detect the influences of UA and/or IR treatment on cellular ferroptosis. The levels of p53/solute carrier family 7a member 11 (SLC7A11)/glutathione peroxidase 4 (GPX4) pathway proteins were detected by Western blot. Additionally, a subcutaneous graft tumor model was constructed in nude mice. RESULTS 10 μM UA reduced the viability and induced death of ESCC cells. UA enhanced the impacts of IR by suppressing cell proliferation, migration and invasion, inducing cell death, and causing cell cycle arrest. Ferroptosis inhibitor impaired the inhibitory impacts of UA and IR on the biological properties of ESCC cells. The combination of UA and IR led to ferroptosis through the modulation of the p53/SLC7A11/GPX4 pathway, and UA enhanced the responsiveness of ESCC cells to IR both in vitro and in vivo. CONCLUSION UA inhibits the malignant biological behavior of ESCC by modulating ferroptosis through the p53/SLC7A11/GPX4 pathway, and enhances the sensitivity of ESCC cells to IR.
Collapse
Affiliation(s)
- Nuran Bedolla
- College of Biological Sciences and Technology, YiLi Normal University, China
| | - Linyu Liu
- College of Biological Sciences and Technology, YiLi Normal University, China
| | - Xueting Liu
- College of Biological Sciences and Technology, YiLi Normal University, China
| | - Qiuxian Xie
- College of Biological Sciences and Technology, YiLi Normal University, China
| | - Yanli Ren
- College of Biological Sciences and Technology, YiLi Normal University, China.
| |
Collapse
|
36
|
Wang K, Wang L, Wu C, Chen H, Cai D, Lu L, Liu X, Jiao Z. Lycopene Ameliorates Polycystic Ovary Syndrome in Rats by Inhibiting Ovarian Ferroptosis Through Activation of the AMPK/Nrf2 Pathway. J Biochem Mol Toxicol 2025; 39:e70158. [PMID: 39871526 DOI: 10.1002/jbt.70158] [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: 10/14/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 01/29/2025]
Abstract
Lycopene (LYC) is an extremely powerful antioxidant with the potential to treat a range of diseases and to inhibit ferroptosis. This research aims to elucidate how LYC impacts polycystic ovarian syndrome (PCOS) and the action mechanisms. A PCOS rat model was constructed by injecting DHEA. Different doses of LYC were injected intraperitoneally in PCOS rats, the estrous cycle was recorded. The histopathological damage of ovary in PCOS rats was observed by HE staining, testosterone (T), estradiol (E2), luteinizing hormone (LH) and follicle stimulating hormone (FSH) levels were examined by ELISA kits. Transmission electron microscopy, prussian blue staining, biochemical kits to determine ferroptosis. Immunohistochemistry and Western blot to assess the levels of ferroptosis-related and AMPK/Nrf2 pathway-related proteins to explore whether LYC affects ferroptosis in PCOS through this pathway. PCOS rats had significantly higher body weights, ovarian weights and ovarian indices, and disorganized estrous cycles, which were dose-dependently ameliorated by LYC. In addition, LYC significantly ameliorated the histopathological damage of ovary in PCOS rats and restored the normal secretion of T, E2, LH, and FSH. LYC attenuates iron deposition in PCOS ovarian tissues, reduces iron and ROS levels, and inhibits ferroptosis. Notably, LYC activated the AMPK/Nrf2 pathway, and AMPK inhibitor intervention attenuated the therapeutic effect of LYC in PCOS rats, suggesting that LYC acts through the AMPK/Nrf2 pathway. LYC attenuates estrous cycle disruption, ameliorates pathological impairments, and inhibits ferroptosis in PCOS rats by modulating the AMPK/Nrf2 pathway.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Gynecology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Lin Wang
- Department of Gynecology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Chengyong Wu
- Department of Gynecology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Hongxiang Chen
- Department of Gynecology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Donghui Cai
- Department of Gynecology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Linglan Lu
- Department of Gynecology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xuli Liu
- Department of Gynecology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Zhen Jiao
- Department of Gynecology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
37
|
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
|
38
|
Wang W, Chen J, Lai S, Zeng R, Fang M, Wan L, Li Y. METTL14 promotes ferroptosis in smooth muscle cells during thoracic aortic aneurysm by stabilizing the m 6A modification of ACSL4. Am J Physiol Cell Physiol 2025; 328:C387-C399. [PMID: 39672203 DOI: 10.1152/ajpcell.00577.2024] [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: 08/09/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/15/2024]
Abstract
Thoracic aortic aneurysm (TAA) is a vascular disease associated with high mortality rates. Ferroptosis has been shown to mediate the transformation of vascular smooth muscle cells (VSMCs). However, the regulatory mechanisms by which ferroptosis influences TAA remain unclear. In this study, we induced TAA mouse models using angiotensin II (Ang II) and evaluated the impact of ferroptosis on the pathological changes observed in TAA mice, employing liproxstatin-1 as a treatment. In addition, we assessed the regulatory effect of METTL14 on the ferroptosis of VSMCs after treating them with a ferroptosis activator (imidazole ketone erastin, IKE). RNA binding protein immunoprecipitation (RIP) and RNA pull-down assays were conducted to investigate the interaction between acyl-CoA synthase long-chain family member 4 (ACSL4) mRNA and the proteins METTL14 or IGF2BP2. The results indicated that the level of ferroptosis was elevated in the thoracic aorta of TAA mice, and METTL14 was upregulated in TAA models and IKE-induced VSMCs. Knockdown of METTL14 was found to inhibit the progression of TAA by reducing the ferroptosis of VSMCs. Furthermore, IGF2BP2 recognized METTL14-modified ACSL4 mRNA and regulated its stability, thereby mediating the ferroptosis of VSMCs. Collectively, the effects of METTL14 on VSMC ferroptosis present therapeutic potential for the treatment of TAA.NEW & NOTEWORTHY Our study confirmed that METTL14 can induce ferroptosis in vascular smooth muscle cells during the progression of thoracic aortic aneurysm by mediating the m6A modification of ACSL4 mRNA.
Collapse
MESH Headings
- Animals
- Ferroptosis/physiology
- Ferroptosis/drug effects
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Coenzyme A Ligases/metabolism
- Coenzyme A Ligases/genetics
- Mice
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Methyltransferases/metabolism
- Methyltransferases/genetics
- Male
- Mice, Inbred C57BL
- Disease Models, Animal
- Humans
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Angiotensin II
- Aorta, Thoracic/pathology
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Jiayi Chen
- The First Clinical Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Songqing Lai
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Ruiyuan Zeng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Ming Fang
- Department of Emergency, Gaoxin Branch of The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Li Wan
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Yiying Li
- Department of Prenatal Diagnostic Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
39
|
Qi H, Gao Y, Zhang Z, Zhang X, Tian D, Jiang Y, Zhang L, Zeng N, Yang R. HouShiHeiSan attenuates sarcopenia in middle cerebral artery occlusion (MCAO) rats. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118917. [PMID: 39423947 DOI: 10.1016/j.jep.2024.118917] [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: 07/21/2024] [Revised: 09/15/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Physical therapy is the main clinical treatment for limb symptoms after ischemic stroke, and there is a lack of reliable drug intervention programs. HouShiHeiSan (HS)comes from "Synopsis of the Golden Chamber", where it is recorded: "seauelae of wind stroke and heaviness of limbs", indicating this formulae is a promising opion for clinical practice. AIM OF THE STUDY The aim of this study is to explore the therapeutic effect of HS on sarcopenia after ischemic stroke (ISS) by using the middle cerebral artery occlusion (MCAO) rats. MATERIALS AND METHODS After 7 days of adaptive feeding Sprague-Dawley (SD) rats were randomly divided into sham and MCAO surgery groups. After MCAO operation, the agreement of the models was evaluated with a laser speckle instrument, and then, treatment groups were administered HS and related solvent. During the 7 days treatment period, the Zea-Longa score was used to assess the neural function, the treadmill for exercise capacity and traction instrument for grip strength. Besides, the physiological electrical signal system was used to record muscular electrical signals, while the muscle thickness was measured by ultrasound. After data acquisition on the 7th day after MCAO operation, the soleus muscle was dissected, and the indexes of length, weight of whole muscle tissue and cross-sectional area of muscular cells by H&E were recorded. Subsequently, mechanistic indicators were examined. MuRF1 and MAFbx expression was detected by immunohistochemistry (IHC). Furthermore, the expression level of more related indicators of muscular differentiation and cellular proterin balance, including mTOR, p-mTOR, AKT, p-AKT, p70s6k, p-p70s6, FOXO1, p-FOXO1, MyoD1, Myostatin, MuRF1 and MAFbx, were tested via Western blot. RESULTS HS improved motor performance and promoted muscle regeneration in MCAO rats. In terms of motor ability, HS mixed with alcohol significantly improved the neurological function damage, reduce the weight loss, increase the running distance per unit time and increase the grip strength. The postoperative muscle electrical signal intensity increased, and muscle thickness, weight, and length were maintained. The HS with alcohol group significantly maintained the cross-sectional size of muscle cells and reduced the number of MyoD1 and myostatin-positive cells in the muscle tissue. It simultaneously promoted the expression of p-mTOR, p-AKT, p-p70s6k, and MyoD1 to promote the synthesis of muscle proteins and inhibited the expression of p-FOXO1, myostatin, MAFbx, and MuRF1 to reduce muscle protein degradation. CONCLUSION HS can enhance muscle protein synthesis and decrease protein breakdown by activating the AKT/mTOR/FOXO1 pathway, thereby preserving muscle health and enhancing motor performance following stroke in rats.
Collapse
Affiliation(s)
- Hu Qi
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuanlin Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zeyang Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiongwei Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dan Tian
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yanning Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lihong Zhang
- Department of Otorhinolaryngology, Chengdu Xinjin District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, China.
| | - Nan Zeng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Ruocong Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
40
|
An W, Luo J, Zhang C, Xiao Q. Integrative Bioinformatics Analysis to Identify Key Ferroptosis-Related Genes and Immune Infiltration in Aortic Aneurysm and Dissection: Implication of PTGS2. J Inflamm Res 2025; 18:1377-1394. [PMID: 39897521 PMCID: PMC11787787 DOI: 10.2147/jir.s488651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/22/2025] [Indexed: 02/04/2025] Open
Abstract
Background Aortic aneurysm and dissection (AAD) represent a highly lethal cardiovascular condition. Ferroptosis has recently been implicated in AAD development and progression. However, ferroptosis-related genes (FRGs) have not been systematically identified and verified in AAD. Methods and Results Seven human AAD datasets downloaded from Gene Expression Omnibus were analyzed, and 113 potential AAD-related FRGs were identified. Function enrichment analyses revealed that the FRGs were mainly associated with responses to chemical stress and cytokine signaling in the immune system. Protein-protein interaction network analyses identified 8 hub FRGs including EZH2, EGFR, HIF1A, IL6, PTGS2, MAPK1, IL1B and SRC. All these FRGs were significantly increased in patients with aortic aneurysm. Additionally, immune cell infiltration analyses revealed these FRGs were strongly correlated with the higher CD4+ Tem and macrophages fraction in AAD patients. Particularly, increased expression of PTGS2 in AAD patients was further validated using our newly collected clinical aortic specimens. Importantly, we found that PTGS2 knockdown could reduce the expression of MMP9 and MMP2 but increase GPX4 expression in macrophages. Conversely, while PTGS2 overexpression upregulated MMP9 and MMP2 expression but downregulated GPX4 expression, the regulatory effects of PTGS2 on these genes were largely blunted by ferroptosis inhibitors. Functionally, administration of celecoxib, a PTGS2-specific inhibitor, into mice significantly reduced β-aminopropionitrile-induced AAD development and progression. Conclusion Through an integrative bioinformatics analysis, we have identified multiple key AAD-related FRGs including PTGS2. Functional studies also suggest a functional role of PTGS2 in ferroptosis and AAD development, offering novel insights into pathogenesis of human AAD.
Collapse
Affiliation(s)
- Weiwei An
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People’s Republic of China
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Jun Luo
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Cheng Zhang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Qingzhong Xiao
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| |
Collapse
|
41
|
Wang D, Zhang C, Guo H, Cui T, Pu W, Huang B, Zhu J, Dai X. Co-exposure to Environmentally Relevant Levels of Molybdenum and Cadmium Induces Oxidative Stress and Ferroptosis in the Ovary of Ducks. Biol Trace Elem Res 2025; 203:374-383. [PMID: 38467966 DOI: 10.1007/s12011-024-04144-1] [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] [Received: 02/04/2024] [Accepted: 03/04/2024] [Indexed: 03/13/2024]
Abstract
Excessive doses of molybdenum (Mo) and cadmium (Cd) have toxic effects on animals. Nevertheless, the reproductive toxicity elicited by Mo and Cd co-exposure remains obscure. To evaluate the co-induce toxic impacts of Mo and Cd on ovaries, 8-day-old 40 healthy ducks were stochastically distributed to four groups and were raised a basal diet supplemented with Cd (4 mg/kg Cd) and/or Mo (100 mg/kg Mo). In the 16th week, ovary tissues were gathered. The data revealed that Mo and/or Cd decreased GSH content, CAT, T-SOD, and GSH-Px activities and increased MDA and H2O2 levels. Moreover, there was a significant decrease in nuclear Nrf2 protein level and its related downstream factors, while cytoplasmic Nrf2 protein level showed a substantial increase. Additionally, a marked elevation was observed in ferrous ion content and TFRC, GCLC, SLC7A11, ACSL4, and PTGS2 expression levels, while FTH1, FTL1, FPN1, and GPX4 expression levels were conversely reduced. These indicators exhibited more marked changes in the joint exposure group. In brief, our results announced that Mo and/or Cd resulted in oxidative stress and ferroptosis in duck ovaries. Synchronously, the Cd and Mo mixture intensified the impacts.
Collapse
Affiliation(s)
- Dianyun Wang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Huiling Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Ting Cui
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Wenjing Pu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Bingyan Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Jiamei Zhu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Xueyan Dai
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China.
| |
Collapse
|
42
|
Li H, Wang X, Liang X, Meng M, Zhang H, Li Z, Lin Y, Li J, Ma C. Verapamil inhibits ferroptosis in septic acute lung injury by blocking L-type calcium channels. Biochem Biophys Res Commun 2025; 744:151202. [PMID: 39708394 DOI: 10.1016/j.bbrc.2024.151202] [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: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), result from pulmonary edema and alveolar-capillary barrier disruption due to inflammation, often triggered by conditions like sepsis. Sepsis-induced ALI (SALI) involves extensive damage to vascular endothelium and alveolar epithelium, leading to respiratory failure. Our study explores ferroptosis, an iron-dependent cell death pathway, and calcium dysregulation in SALI. Elevated cytosolic calcium early in ferroptosis exacerbates lipid peroxidation and cellular damage. We investigated verapamil, a calcium channel blocker, and found it reduces calcium influx, alleviates iron overload, and decreases oxidative stress, protecting against ferroptosis-induced apoptosis in lung cells. These insights suggest targeting ferroptosis pathways, including calcium and iron homeostasis, may offer new therapeutic strategies for SALI, potentially improving outcomes in ALI/ARDS.
Collapse
Affiliation(s)
- Hongru Li
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Xuan Wang
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China; Diagnostic Center of Infections, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China.
| | - Xiangyang Liang
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China; School and Hospital of Stomatology, Hebei Medical University, 383 Zhongshan East Road, Shijiazhuang, Hebei, PR China.
| | - Meiqi Meng
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Haixia Zhang
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Zixin Li
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Yushan Lin
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| | - Jihong Li
- Diagnostic Center of Infections, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China.
| | - Cuiqing Ma
- Immunology Department of Hebei Medical University, Shijiazhuang, PR China.
| |
Collapse
|
43
|
Wang Z, Gui Z, Zhang L, Wang Z. Advances in the mechanisms of vascular calcification in chronic kidney disease. J Cell Physiol 2025; 240:e31464. [PMID: 39392232 DOI: 10.1002/jcp.31464] [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: 05/15/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Vascular calcification (VC) is common in patients with advanced chronic kidney disease (CKD).A series of factors, such as calcium and phosphorus metabolism disorders, uremic toxin accumulation, inflammation and oxidative stress and cellular senescence, cause osteoblast-like differentiation of vascular smooth muscle cells, secretion of extracellular vesicles, and imbalance of calcium regulatory factors, which together promote the development of VC in CKD. Recent advances in epigenetics have provided better tools for the investigation of VC etiology and new approaches for finding more accurate biomarkers. These advances have not only deepened our understanding of the pathophysiological mechanisms of VC in CKD, but also provided valuable clues for the optimization of clinical predictors and the exploration of potential therapeutic targets. The aim of this article is to provide a comprehensive overview of the pathogenesis of CKD VC, especially the new advances made in recent years, including the various key factors mentioned above. Through the comprehensive analysis, we expect to provide a solid theoretical foundation and research direction for future studies targeting the specific mechanisms of CKD VC, the establishment of clinical predictive indicators and the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Zebin Gui
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Lirong Zhang
- Department of Radiology, Affliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| |
Collapse
|
44
|
Pan M, Zhang L, Chang S, Jiang X, Shen J, Feng X, Xu F, Zha X, Chen X, Fan X. Poly-l-arginine promotes ferroptosis in asthmatic airway epithelial cells by modulating PBX1/GABARAPL1 axis. Int J Biol Macromol 2025; 286:138478. [PMID: 39645127 DOI: 10.1016/j.ijbiomac.2024.138478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Eosinophils play a featured role among inflammatory cells participating in the onset and development of asthma. Activated eosinophils release several cytotoxic granular proteins, such as major basic protein (MBP), posing a significant threat to airway epithelium. Ferroptosis, a novel form of cell death, is gaining recognition for its involvement in asthma pathogenesis, though the specific mechanisms remain largely unknown. Herein, we revealed that poly-l-arginine (PLA), an MBP mimic, induced ferroptosis in airway epithelium by downregulating γ-aminobutyric acid receptor-associated protein-like 1 (GABARAPL1). Reduced GABARAPL1 expression was further confirmed in ovalbumin (OVA)-induced asthma mice and PLA-treated human airway organoids (hAOs). Mechanistically, PLA activated mechanistic target of rapamycin complex 1 (mTORC1) signaling, inhibiting pre-B-cell leukemia transcription factor 1 (PBX1), which in turn leads to transcriptional downregulation of GABARAPL1. Furthermore, MBP extracted from eosinophils, similar to PLA, induced ferroptosis in airway epithelial cells, as well as modulating mTORC1/PBX1/GABARAPL1 pathway. Finally, Ferrostatin-1 treatment or GABARAPL1 overexpression alleviated ferroptosis and airway inflammation in asthmatic mice. Overall, our findings highlight the cell communication between eosinophils and airway epithelial cells. MBP modulates the mTORC1/PBX1/GABARAPL1 axis, thereby serving as a significant contributor to ferroptosis in airway epithelium and airway inflammation. This suggests that suppressing ferroptosis in airway epithelium or targeting eosinophils and MBP could lead to novel therapeutic strategies for asthma management.
Collapse
Affiliation(s)
- Min Pan
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Ling Zhang
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Shuang Chang
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Xueqin Jiang
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, China
| | - Jiapan Shen
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Xiaoxia Feng
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China
| | - Fangzhou Xu
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Xu Chen
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China.
| | - Xiaoyun Fan
- Department of Geriatric Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Geriatric Institute, Hefei, China; Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, China.
| |
Collapse
|
45
|
Jin R, Dai Y, Wang Z, Hu Q, Zhang C, Gao H, Yan Q. Unraveling Ferroptosis: A New Frontier in Combating Renal Fibrosis and CKD Progression. BIOLOGY 2024; 14:12. [PMID: 39857243 PMCID: PMC11763183 DOI: 10.3390/biology14010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025]
Abstract
Chronic kidney disease (CKD) is a global health concern caused by conditions such as hypertension, diabetes, hyperlipidemia, and chronic nephritis, leading to structural and functional kidney injury. Kidney fibrosis is a common outcome of CKD progression, with abnormal fatty acid oxidation (FAO) disrupting renal energy homeostasis and leading to functional impairments. This results in maladaptive repair mechanisms and the secretion of profibrotic factors, and exacerbates renal fibrosis. Understanding the molecular mechanisms of renal fibrosis is crucial for delaying CKD progression. Ferroptosis is a type of discovered an iron-dependent lipid peroxidation-regulated cell death. Notably, Ferroptosis contributes to tissue and organ fibrosis, which is correlated with the degree of renal fibrosis. This study aims to clarify the complex mechanisms of ferroptosis in renal parenchymal cells and explore how ferroptosis intervention may help alleviate renal fibrosis, particularly by addressing the gap in CKD mechanisms related to abnormal lipid metabolism under the ferroptosis context. The goal is to provide a new theoretical basis for clinically delaying CKD progression.
Collapse
Affiliation(s)
- Rui Jin
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Dai
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng Wang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qinyang Hu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongyu Gao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Yan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Provincial Clinical Medical Research Center for Nephropathy, Enshi 445000, China
| |
Collapse
|
46
|
Liu Q, Mao T, Liu F, Chen B, Liu Z, Pathak JL, Li J. Apigenin alleviates Sjögren's syndrome-induced salivary gland epithelial cell ferroptosis via ERα signaling-mediated regulation of the ATF3/SLC7A1l axis. Int Immunopharmacol 2024; 143:113409. [PMID: 39426238 DOI: 10.1016/j.intimp.2024.113409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND In Sjögren's syndrome (SS)-an autoimmune disease characterized by dry mouth and eyes-salivary gland epithelial cells (SGECs) undergo ferroptosis, which disrupts their integrity and impairs saliva secretion. Apigenin, a phytoestrogen, is known to activate estrogen signalling and alleviate xerostomia in ovariectomized mice; however, its effect on SGEC survival and function in SS remains unclear. We hypothesized that apigenin alleviates SS symptoms and progression by inhibiting ferroptosis in SGECs and aimed to elucidate the underlying mechanism. METHODS Apigenin (50 mg/kg) was orally gavaged to non-obese diabetic (NOD)/LtJ female mice (SS model); changes in SS functional indicators were analyzed using mRNA sequencing and bioinformatic analyses of submandibular glands. Interferon-gamma (IFN-γ)-stimulated SGECs were used to model SS in vitro; SGEC activity and aquaporin 5 (AQP5) expression were analyzed. Immunohistochemical staining, transmission electron microscopy, RT-qPCR, western blotting and other methods were used to verify the mechanisms. RESULTS Apigenin significantly increased salivary secretion and AQP5 expression while inhibiting ferroptosis and immune infiltration in NOD mouse submandibular glands. The oxidative stress gene ATF3 was upregulated and GPX4 was downregulated in NOD mice compared to that in control group (ICR mice); however, apigenin reversed this effect. IFN-γ treatment downregulated AQP5, SLC7A11, and GPX4 expression while promoting ATF3 expression and ferroptosis, which was mitigated by apigenin. ATF3 knockdown increased SLC7A11 and GPX4 expression, inhibiting SS and ferroptosis. Furthermore, apigenin inhibited ferroptosis in SGECs through ESR1 binding to ATF3. CONCLUSION Apigenin alleviates SS by regulating SGEC ferroptosis via the ERα-regulated ATF3/SLC7A11 axis, highlighting its therapeutic potential in SS.
Collapse
Affiliation(s)
- Qianwen Liu
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou 510140, China
| | - Tianjiao Mao
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou 510140, China
| | - Fangqi Liu
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou 510140, China
| | - Bo Chen
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou 510140, China
| | - Zhuoyuan Liu
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou 510140, China
| | - Janak L Pathak
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou 510140, China.
| | - Jiang Li
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou 510140, China.
| |
Collapse
|
47
|
Dong Q, Liu F, Zhu J, Li M, Chen A, Feng L, Lan Z, Ye Y, Lu L, Liang Q, Yan J. 4-Octyl itaconate inhibits vascular calcification partially via modulation of HMOX-1 signaling. Eur J Pharmacol 2024; 985:177122. [PMID: 39532225 DOI: 10.1016/j.ejphar.2024.177122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Vascular calcification frequently occurs in patients with chronic conditions such as chronic kidney disease (CKD), diabetes, and hypertension and represents a significant cause of cardiovascular events. Thus, identifying effective therapeutic targets to inhibit the progression of vascular calcification is essential. 4-Octyl itaconate (4-OI), a derivative of itaconate, exhibits anti-inflammatory and antioxidant activity, both of which play an essential role in the progression of vascular calcification. However, the role and molecular mechanisms of 4-OI in vascular calcification have not yet been elucidated. In this study, we investigated the effects of exogenous 4-OI on vascular calcification using vascular smooth muscle cells (VSMCs), arterial rings, and mice. Alizarin red staining and western blot revealed that 4-OI inhibited calcification and osteogenic differentiation of human VSMCs. Similarly, 4-OI inhibited calcification of rat and human arterial rings and VitD3-overloaded mouse aortas. Mechanistically, RNA sequencing analysis revealed that 4-OI treatment is most likely to affect heme oxygenase 1 (HMOX-1) mRNA expression. The study demonstrated that 4-OI treatment increased HMOX-1 mRNA and protein levels, but suppressed inflammation and oxidative stress in VSMCs under osteogenic conditions. Moreover, HMOX-1 knockdown by siRNA or treatment with the HMOX-1 inhibitor ZnPP9 significantly reversed the suppression effect on calcification of VSMCs and aortas of VitD3-overloaded mice by 4-OI. Furthermore, HMOX-1 knockdown by siRNA markedly abrogated the inhibitory effect of 4-OI on inflammation in VSMCs. These findings suggest that 4-OI alleviates vascular calcification and inhibits oxidative stress and inflammation through modulation of HMOX-1, indicating its potential as a therapeutic target for vascular calcification.
Collapse
MESH Headings
- Vascular Calcification/drug therapy
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Animals
- Humans
- Heme Oxygenase-1/metabolism
- Succinates/pharmacology
- Signal Transduction/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice
- Male
- Rats
- Osteogenesis/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Mice, Inbred C57BL
- Rats, Sprague-Dawley
- Cells, Cultured
Collapse
Affiliation(s)
- Qianqian Dong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Fang Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Jiahui Zhu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Mingxi Li
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, China
| | - An Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Liyun Feng
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China
| | - Lihe Lu
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, China.
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, China.
| |
Collapse
|
48
|
Huang N, Liu C, Liu Z, Lei H. Disulfidptosis-related gene in acute myocardial infarction and immune microenvironment analysis: A bioinformatics analysis and validation. PLoS One 2024; 19:e0314935. [PMID: 39666769 PMCID: PMC11637291 DOI: 10.1371/journal.pone.0314935] [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: 06/07/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024] Open
Abstract
Disulfidptosis is a newly discovered method of cell death. However, no studies have fully elucidated the role of disulfidptosis-related genes (DSRGs) in acute myocardial infarction (AMI). The potential role of DSRGs in AMI was analyzed through a comprehensive bioinformatics approach. Finally, hub genes were verified in vitro by qPCR. Sixteen DE-DSRGs were in the AMI. Thereafter, seven hub genes were determined by machine learning algorithms, which had potential diagnostic value in AMI. The risk model showed a robust diagnostic value (area under curve, AUC = 0.940). Prognostic analysis revealed the potential prognostic value of INF2 and CD2AP. Immune landscape analysis showed that hub genes were closely related to the immune microenvironment. By predictive analysis, we obtained four miRNAs, thirteen small molecule drugs, and five TFs closely related to hub genes. Experimental verification revealed that Slc3a2 and Inf2 were significantly up-regulated and Dstn was significantly down-regulated in the hypoxic model. Our study demonstrated that DSRGs are disorderedly expressed in AMI and identified seven hub genes through machine learning. In addition, a diagnostic model was constructed based on hub genes, providing a new perspective for the early diagnosis of AMI.
Collapse
Affiliation(s)
- Nan Huang
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, Hunan Province, PR China
| | - Chan Liu
- Clinical Pharmacy, Liuyang People’s Hospital, Liuyang, Hunan Province, China
| | - Zheng Liu
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, Hunan Province, PR China
| | - Haibo Lei
- Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, Hunan Province, PR China
| |
Collapse
|
49
|
Du Z, Shi Y, Tan J. Advances in integrating single-cell sequencing data to unravel the mechanism of ferroptosis in cancer. Brief Funct Genomics 2024; 23:713-725. [PMID: 38874174 DOI: 10.1093/bfgp/elae025] [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: 03/18/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
Ferroptosis, a commonly observed type of programmed cell death caused by abnormal metabolic and biochemical mechanisms, is frequently triggered by cellular stress. The occurrence of ferroptosis is predominantly linked to pathophysiological conditions due to the substantial impact of various metabolic pathways, including fatty acid metabolism and iron regulation, on cellular reactions to lipid peroxidation and ferroptosis. This mode of cell death serves as a fundamental factor in the development of numerous diseases, thereby presenting a range of therapeutic targets. Single-cell sequencing technology provides insights into the cellular and molecular characteristics of individual cells, as opposed to bulk sequencing, which provides data in a more generalized manner. Single-cell sequencing has found extensive application in the field of cancer research. This paper reviews the progress made in ferroptosis-associated cancer research using single-cell sequencing, including ferroptosis-associated pathways, immune checkpoints, biomarkers, and the identification of cell clusters associated with ferroptosis in tumors. In general, the utilization of single-cell sequencing technology has the potential to contribute significantly to the investigation of the mechanistic regulatory pathways linked to ferroptosis. Moreover, it can shed light on the intricate connection between ferroptosis and cancer. This technology holds great promise in advancing tumor-wide diagnosis, targeted therapy, and prognosis prediction.
Collapse
Affiliation(s)
- Zhaolan Du
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Yi Shi
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Jianjun Tan
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
50
|
Wang Y, Zhao X, Chen B, Chen S, Liang Y, Chen D, Li X. Methylophiopogonanone A Inhibits Ferroptosis in H9c2 Cells: An Experimental and Molecular Simulation Study. Molecules 2024; 29:5764. [PMID: 39683922 DOI: 10.3390/molecules29235764] [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: 10/04/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
In this study, homoisoflavone methylophiopogonanone A (MOA) was investigated for its inhibitory effect on ferroptosis of H9c2 cells using a set of cellular assays, such as BODIPY-probed and H2DCFDA-probed flow cytometry analyses, cell counting kit-8 analysis (CCK-8), and lactate dehydrogenase (LDH) release analysis. All these cellular assays adopted Fer-1 as the positive control. Subsequently, MOA and Fer-1 were subjected to two antioxidant assays, i.e., 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide radical (PTIO•)-scavenging and 2,2'-azinobis(3-ethylbenzo-thiazoline-6-sulfonic acid radical (ABTS•+)-scavenging. Finally, MOA, along with Fer-1, were systematically analyzed for molecular docking and dynamics simulations using a set of software tools. The experimental results revealed that MOA could inhibit ferroptosis of H9c2 cells but did not effectively scavenge PTIO• and ABTS•+ free radicals. Two molecular simulation methods or algorithms suggested that MOA possessed similar binding affinity and binding free energy (∆Gbind) to Fer-1. Visual analyses indicated various hydrophobic interactions between MOA and one of the seven enzymes, including superoxide dismutase (SOD), dihydroorotate dehydrogenase (DHODH), ferroportin1 (FPN), ferroptosis suppressor protein 1 (FSP1), glutathione peroxidase 4 (GPX4), nicotinamide adenine dinucleotide phosphate (NADPH), and solute carrier family 7 member 11 (SLC7A11). Based on these experimental and molecular simulation results, it is concluded that MOA, a homoisoflavonoid with meta-di-OHs, can inhibit ferroptosis in H9c2 cells. Its inhibitory effect is mainly attributed to the regulation of enzymes rather than direct free radical scavenging. The regulation of enzymes primarily depends on hydrophobic interactions rather than H-bond formation. During the process, flexibility around position 9 allows MOA to adjust to the enzyme binding site. All these findings provide foundational information for developing MOA and its derivatives as potential drugs for myocardial diseases.
Collapse
Affiliation(s)
- Yanqing Wang
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xi Zhao
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ban Chen
- School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, China
| | - Shaoman Chen
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yongbai Liang
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Dongfeng Chen
- Department of Anatomy, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xican Li
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|