1
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
2
|
Zhang Z, Chen Z. Exploring the causal links between inflammation-related genes and atherosclerosis through Mendelian randomization analysis. Medicine (Baltimore) 2025; 104:e42584. [PMID: 40489880 DOI: 10.1097/md.0000000000042584] [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] [Indexed: 06/11/2025] Open
Abstract
Atherosclerosis is characterized by chronic inflammation and plaque formation in arterial walls, accompanying by significant involvement of immune cells and mediators. Studies have demonstrated that factors, such as colony-stimulating factor-1, play critical roles in the development and exacerbation of these plaques, emphasizing the complexity of immune interactions in atherosclerosis. This study utilized GSE198600, GSE120521, GSE253903, and GSE224273 datasets to investigate the gene expression profiles and molecular mechanisms underlying atherosclerotic plaques. Differentially expressed genes were identified using DESeq2, followed by pathway analysis with clusterProfiler. Key hub genes were further explored through Cytoscape, and Mendelian randomization was performed using the TwoSampleMR package to assess genetic causality using ebi-a-GCST005840 and ebi-a-GCST006907 datasets. Finally, single-cell RNA sequencing data from GSE224273 and GSE253903 datasets were analyzed to characterize gene expression across various cell types in carotid atherosclerotic lesions. This study presented a comprehensive analysis of gene expression across various cell types in carotid atherosclerosis, highlighting the significant roles of key genes such as HMOX1, CD52, CSF1R, CD177, PTGDS, and TNC. HMOX1, primarily expressed in macrophages and dendritic cells, played a pivotal role in the immune response within atherosclerotic environments. CD52 and CSF1R, exhibiting high expression in immune cells, were implicated in modulating inflammatory processes crucial for plaque development and stability. CD177 and PTGDS emerged as top differentially expressed genes in symptomatic carotid plaques, linking them to critical biological processes, such as immune response and prostaglandin synthesis, which are essential for atherosclerosis progression. Similarly, TNC, a gene associated with extracellular matrix organization, showed significant upregulation in unstable plaque sections, underscoring its potential role in plaque destabilization and adverse cardiovascular events. These findings elucidated the complex cellular and molecular interactions in atherosclerosis, providing insights into potential therapeutic targets for preventing or treating atherosclerotic cardiovascular diseases. The differential expression of these genes across various cell types further emphasized the heterogeneity of the disease and the need for targeted approaches to effectively manage atherosclerosis.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Vascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | | |
Collapse
|
3
|
Jiang J, Li Y, Chen Y, Wu Q, Ding S. Ambient fine particulate matter induces cardiac fibrosis through triggering ferroptosis by heme degradation induced-iron overload. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118227. [PMID: 40280044 DOI: 10.1016/j.ecoenv.2025.118227] [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: 12/10/2024] [Revised: 04/14/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Previous studies have shown a significant correlation between exposure to ambient fine particulate matter (PM2.5) and cardiac fibrosis, yet the precise detrimental effects and underlying mechanisms of PM2.5 exposure on cardiac fibrosis remain incompletely understood. Cardiac remodeling, a process involving ferroptosis that can be initiated by iron overload, has been implicated in this phenomenon. In this study, we sought to explore the potential mechanism by which ferroptosis contributes to PM2.5-induced cardiac fibrosis. METHODS AND RESULTS Male C57BL/6 J mice were exposed to ambient PM2.5 by intratracheal instillation twice a week for 12 weeks to establish PM2.5-exposed murine models and cardiomyocytes were used to verify the role of ferroptosis in PM2.5-induced cardiac fibrosis. In this study, it was observed that exposure to PM2.5 resulted in cardiac fibrosis and a significant upregulation of cardiac fibrosis-related markers (TGF-β1, collagen-I and p-Smad3), heme oxygenase 1 (HO-1), and ACSL4 (a biomarker for ferroptosis). Additionally, PM2.5 exposure led to a decrease in heme content, iron overload, increased levels of the lipid peroxidation marker 4-HNE, and a reduction in the ratio of GSH/GSSG and GPX4 (a biomarker for ferroptosis) in murine hearts. Significantly, the use of ferrostatin-1 (an inhibitor of ferroptosis) mitigated PM2.5-induced cardiac fibrosis and decreased the levels of cardiac fibrosis-related markers (TGF-β1, collagen-I and p-Smad3) in murine hearts, indicating the essential role of ferroptosis in the development of cardiac fibrosis. In vitro experiments showed that PM2.5 upregulated the expression of HO-1 protein, promoted iron accumulation, increased 4-HNE levels, and triggered ferroptosis in cardiomyocytes. The inhibition of HO-1 (zinc protoporphyrin 9) and siRNA HO-1 effectively mitigated PM2.5-induced iron overload, ferroptosis, and heme accumulation in cardiomyocytes. Additionally, treatment with ferrostatin-1 markedly decreased the expression levels of cardiac fibrosis-related markers, such as TGF-β1 and p-Smad3. CONCLUSION Collectively, our study showed that the activation of ferroptosis/TGF-β1/Smad3 signaling pathway, initiated by heme degradation-induced iron overload in cardiomyocytes, serves as a mechanism in murine models of PM2.5-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Jinjin Jiang
- School of Public Health and Management, Jiangsu Medical College, Yancheng, Jiangsu Province 224005, PR China; Jiangsu Engineering Research Centers for Cardiovascular and Cerebrovascular Disease and Cancer Prevention and Control, Yancheng, Jiangsu Province 224005, PR China
| | - Yang Li
- School of Public Health and Management, Jiangsu Medical College, Yancheng, Jiangsu Province 224005, PR China; Jiangsu Engineering Research Centers for Cardiovascular and Cerebrovascular Disease and Cancer Prevention and Control, Yancheng, Jiangsu Province 224005, PR China
| | - Yuping Chen
- Jiangsu Engineering Research Centers for Cardiovascular and Cerebrovascular Disease and Cancer Prevention and Control, Yancheng, Jiangsu Province 224005, PR China; Department of Technology, Jiangsu Medical College, Yancheng, Jiangsu Province 224005, PR China
| | - Qin Wu
- Jiangsu Engineering Research Centers for Cardiovascular and Cerebrovascular Disease and Cancer Prevention and Control, Yancheng, Jiangsu Province 224005, PR China; Jiangsu Medical College, Yancheng, Jiangsu Province 224005, PR China.
| | - Shibin Ding
- School of Public Health and Management, Jiangsu Medical College, Yancheng, Jiangsu Province 224005, PR China; Jiangsu Engineering Research Centers for Cardiovascular and Cerebrovascular Disease and Cancer Prevention and Control, Yancheng, Jiangsu Province 224005, PR China.
| |
Collapse
|
4
|
Kadeerbieke B, Wu L, Zhang YM. The role of lncRNA H19/Hmox1 axis regulating ferroptosis in anthracycline-induced cardiotoxicity. Drug Chem Toxicol 2025:1-12. [PMID: 40432412 DOI: 10.1080/01480545.2025.2503946] [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: 11/11/2024] [Revised: 04/24/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025]
Abstract
This study investigates the molecular mechanisms underlying anthracyclines (ANT)-induced cardiotoxicity, with a specific focus on ferroptosis regulated by the long non-coding RNA (lncRNA) H19/heme oxygenase-1 (Hmox1) signaling axis. A retrospective analysis was performed on 50 breast cancer patients who developed ANT-associated cardiac dysfunction. Clinical assessments included measurements of left ventricular ejection fraction (LVEF) and serum markers, such as cardiac troponin I (cTnI), creatine kinase-MB (CK-MB), N-terminal pro-B-type natriuretic peptide (NT-proBNP), and serum iron levels. Serum analysis revealed a marked downregulation of lncRNA H19 and upregulation of Hmox1, both significantly correlated with impaired cardiac function and disrupted iron homeostasis. To further elucidate the mechanism, an Epirubicin (EPI)-induced injury model in HL-1 cardiomyocytes was established. EPI exposure led to suppression of lncRNA H19, upregulation of Hmox1, and induction of apoptosis and ferroptotic cell death. RNA-seq analysis identified potential downstream targets linking lncRNA H19 to iron metabolism via Hmox1 modulation. Functional assays demonstrated that overexpression of lncRNA H19 mitigated EPI-induced ferroptosis, while enforced expression of Hmox1 reversed these protective effects. Collectively, these findings identify the lncRNA H19/Hmox1 axis as a critical regulator of ferroptosis in ANT-induced cardiotoxicity and suggest it as a potential therapeutic target for mitigating cardiac injury in breast cancer patients undergoing anthracycline chemotherapy.
Collapse
Affiliation(s)
- Bayan Kadeerbieke
- Department of Oncology Cardiology, Xinjiang Medical University Cancer Hospital, Urumqi, China
| | - Li Wu
- Department of Oncology Cardiology, Xinjiang Medical University Cancer Hospital, Urumqi, China
| | - Yuan-Ming Zhang
- Department of Oncology Cardiology, Xinjiang Medical University Cancer Hospital, Urumqi, China
| |
Collapse
|
5
|
Bhatnagar A, Tripathi AD, Kumari S, Mishra A. Cadmium sulfide nanoparticles (CdSNPs) modulate key oncogenic pathways in PA1 ovarian cancer cells: Insights from transcriptomic analysis. Toxicol In Vitro 2025; 107:106079. [PMID: 40374020 DOI: 10.1016/j.tiv.2025.106079] [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: 09/09/2024] [Revised: 04/25/2025] [Accepted: 05/10/2025] [Indexed: 05/17/2025]
Abstract
Transcriptomics has become a useful tool for comparing the levels of gene expression in healthy and malignant cells, holding potential for the discovery of new cancer therapies. This study used RNA-sequencing and transcriptome analysis on the PA1 ovarian cancer cell line to examine the potential of Cadmium Sulfide Nanoparticles (CdSNPs) as a therapeutic agent. A total of 5.42 Gb of high-quality reads was estimated based on the findings of gene expression techniques, comprising 2.25 Gb of treated PA1 cells and 3.17 Gb of control cells. Of these, 1641 genes with padj<0.001 and log2 foldchange >2 were found to be significantly regulated DEGs (differentially expressed genes). Analysis of gene ontology (GO) assays demonstrates the molecular mechanism behind CdSNPs anticancer effects. GO:0006915, GO:0012501, GO:1903561, and GO:0070588 are a few significant highlights of elevated GO (enriched DEGs) that are involved in apoptotic pathways, extracellular vesicles, programmed cell death, and Ca++ signaling. KEGG analysis elucidated that up and downregulated DEGs were enriched in a few pathways: calcium signaling pathway, Apoptosis, and TNF signaling pathway. Important pathways like MAP kinase, JAK/STAT, cAMP, and folate biosynthesis, showed inhibitory effects on ovarian cancer cell proliferation. The results of this work provide insight into possible therapeutic approaches employing CdSNPs and encourage additional research using a variety of cell lines and in vivo models to improve ovarian cancer treatment.
Collapse
Affiliation(s)
- Aditi Bhatnagar
- School of Biochemical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Abhay Dev Tripathi
- School of Biochemical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Sonali Kumari
- School of Biochemical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Abha Mishra
- School of Biochemical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
6
|
Wilkinson T, Fletcher E, Ring A, Bradley C, Papoutsi E, Miserlis D, Smith RS, Bohannon WT, Pipinos II, Koutakis P. Deciphering the Role of Ferroptosis in the Pathogenesis of Peripheral Artery Disease Myopathy. BIOLOGY 2025; 14:537. [PMID: 40427725 PMCID: PMC12108827 DOI: 10.3390/biology14050537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
This study investigates ferroptosis in the context of peripheral artery disease (PAD), a vascular disease characterized by atherosclerosis of the lower extremities. Muscle atrophy and increased oxidative stress are hallmarks of PAD and correlate with worse clinical outcomes. Given ferroptosis' association with oxidative stress, we explored its role in PAD myopathy by examining gene and protein markers related to metabolic pathways implicated in ferroptosis using both human PAD patients and cultured myotubes. Intermittent claudication (IC) PAD patients, critical limb ischemia (CLI) PAD patients, and non-PAD controls were recruited for this study. Calf muscle biopsies were analyzed for gene expression using qPCR, and protein levels were determined by Western blotting. Cultured myotubes treated with the ferroptosis inducer erastin provided an in vitro comparison. Results demonstrated upregulation of ferroptosis markers such as lipid peroxidation and PTGS2 gene expression in the muscle of CLI PAD patients compared to controls. Increased expression of ferroptosis-related genes HMOX1, ACSL4, ELAVL1, and Beclin-1 was also observed. Protein analysis showed trends consistent with gene expression in some ferroptosis markers. The increase in ferroptosis markers in CLI PAD patients, particularly in iron metabolism and autophagy pathways, suggests ferroptosis contributes to PAD myopathy.
Collapse
Affiliation(s)
| | - Emma Fletcher
- Department of Public Health, University of West Florida, Pensacola, FL 32514, USA
| | - Andrew Ring
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | | | - Evlampia Papoutsi
- Department of Public Health, University of West Florida, Pensacola, FL 32514, USA
| | - Dimitrios Miserlis
- Department of Surgery and Perioperative Care, Dell Medical School, University of Texas, Austin, TX 78712, USA
| | - Robert S. Smith
- Department of Surgery, Baylor Scott and White Medical Center, Temple, TX 76508, USA
| | - William T. Bohannon
- Department of Surgery, Baylor Scott and White Medical Center, Temple, TX 76508, USA
| | - Iraklis I. Pipinos
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| | - Panagiotis Koutakis
- Department of Biology, Baylor University, Waco, TX 76798, USA
- Department of Public Health, University of West Florida, Pensacola, FL 32514, USA
- Department of Surgery, University of Nebraska at Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
7
|
Zhao L, Gao Q, Hu K, Lu S. Matrine Alleviates Atherosclerosis by Targeting REG1A and Activating the PI3K/AKT/mTOR Pathway to Inhibit Endothelial Cell Ferroptosis. Biochem Genet 2025:10.1007/s10528-025-11117-z. [PMID: 40281246 DOI: 10.1007/s10528-025-11117-z] [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: 12/26/2024] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Matrine, a natural alkaloid, has a wide range of pharmacological effects, such as antibacterial, anti-inflammatory, anti-oxidation, and anti-tumor. However, the molecular mechanism of matrine in the treatment of atherosclerosis (AS) is not fully understood. Human umbilical vein endothelial cells (HUVECs) were treated with 100 μg/mL ox-LDL to construct an AS cell model in vitro, and the cells were treated with matrine at different concentrations. Our results showed that matrine alleviated the decrease of HUVEC viability and the increase of ferroptosis induced by ox-LDL treatment. Subsequently, we found that matrine targeted regenerating family member 1 alpha (REG1A) and inhibited the expression level of REG1A in ox-LDL treated HUVECs. Overexpression of REG1A attenuated the improvement of matrine on activation of the PI3K/Akt/mTOR pathway and ferroptosis in ox-LDL treated HUVECs. In addition, both LY294002 (an inhibitor of the PI3K signaling) and Erastin (an inducer of ferroptosis) reversed the alleviation of matrine treatment on ferroptosis in ox-LDL treated HUVECs. The results in vivo showed that matrine treatment inhibited high-fat diet-induced aortic ferroptosis in ApoE-/- mice and alleviated arterial tissue lesions. In summary, matrine inhibits ferroptosis by targeting REG1A to activate PI3K/Akt/mTOR pathway, thereby alleviating aortic endothelial injury and lipid plaque formation in AS mice, suggesting that matrine has potential value for the treatment of AS.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Qing Gao
- Department of Nursing, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Kaifeng Hu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Shaoying Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
8
|
Yang H, Xiao G, Wang D, Xiong T, Wang J, Jing X, Xiong B, Xie J, Liu B, She Q. Inhibition of HMOX1 alleviates diabetic cardiomyopathy by targeting ferroptosis. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40235314 DOI: 10.3724/abbs.2024232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025] Open
Abstract
Diabetic cardiomyopathy (DCM) is an important complication of chronic diabetes mellitus. However, its pathologic process and pathogenesis have not been fully elucidated. This study aims to investigate the role of ferroptosis in DCM and clarify the effect of heme oxygenase-1 (HMOX1) on DCM by targeting ferroptosis. In vivo, an animal model of DCM is established by subjecting mice to a high-fat diet (HFD) combined with low-dose streptozotocin (STZ) injection. We induce an in vitro DCM model by exposing H9C2 cells to high glucose and palmitic acid. Transcriptome sequencing reveals that the differentially expressed genes (DEGs) are enriched primarily in fatty acid metabolism and mitochondrial fatty acid β-oxidation, which are closely related to ferroptosis. The experimental results show that the diabetic microenvironment induces ferroptosis both in vivo and in vitro. Western blot analysis reveals the decreased expressions of the antioxidant proteins GPX4, SLC7A11 and ferritin in the DCM group. However, qPCR demonstrates the elevated expressions of the ferroptosis markers PTGS2 and ACSL4. Biochemical indicators further support the occurrence of ferroptosis, with increased levels of malondialdehyde (MDA) and lactate dehydrogenase (LDH), along with decreased level of glutathione (GSH). In vitro, intervention with high glucose and palmitic acid in H9C2 cells results in ferroptosis, which is reversed by ferrostatin-1 (Fer-1). Results show the elevated expression of HMOX1 in DCM. Moreover, knockdown of HMOX1 ameliorates ferroptosis, thereby alleviating diabetic cardiomyopathy by reducing cardiac fibrosis and improving cardiac function. Our study elucidates the role of HMXO1 in DCM pathogenesis and provides a potential therapeutic strategy for clinical treatment.
Collapse
Affiliation(s)
- Huiping Yang
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Gongyi Xiao
- Department of Orthopedic Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- Department of Orthopedic Surgery, Chonggang General Hospital, Chongqing 400000, China
| | - Dinghui Wang
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Tianhua Xiong
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jing Wang
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiaodong Jing
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Bingquan Xiong
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Junmei Xie
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Bin Liu
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qiang She
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
9
|
Ma RX, Lin BH, Feng SX, Bu YT, Chen ZH, Huang YX, Li EL, Weng SJ, Yang L. Evaluation of proanthocyanidins in treating Type 2 diabetic osteoporosis via SIRT6/Nrf2/GPX4 pathways. FASEB J 2025; 39:e70487. [PMID: 40178920 DOI: 10.1096/fj.202403032r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/17/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025]
Abstract
This study investigates the therapeutic potential of proanthocyanidins (PAC) in addressing Type 2 diabetic osteoporosis (T2DOP) by activating the SIRT6/Nrf2/GPX4 signaling pathways. T2DOP is characterized by compromised bone structure and heightened oxidative stress, where ferroptosis plays a pivotal role. Utilizing a T2DOP mouse model and MC3T3-E1 cells under high glucose conditions, we evaluated the impact of PAC on bone health and iron homeostasis. Our results, obtained through micro-CT, histological staining, Western blot, and immunofluorescence analyses, revealed reductions in bone density and decreased GPX4 expression in T2DOP conditions, indicating ferroptosis and oxidative stress. However, PAC treatment improved trabecular bone structure, reduced bone marrow adipocytes, decreased oxidative stress, and enhanced expression of key osteogenic proteins. These findings highlight PAC's potential in mitigating T2DOP through the SIRT6/Nrf2/GPX4 pathways, offering promising therapeutic insights for managing diabetic osteoporosis.
Collapse
Affiliation(s)
- Run-Xun Ma
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Bing-Hao Lin
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Si-Xiang Feng
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yi-Tian Bu
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Zi-Hao Chen
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yi-Xun Huang
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - En-Li Li
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - She-Ji Weng
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Lei Yang
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| |
Collapse
|
10
|
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
|
11
|
Wang W, Gong J. Identification of Key Nucleotide Metabolism Genes in Diabetic Retinopathy Based on Bioinformatics Analysis and Experimental Verification. BIOLOGY 2025; 14:409. [PMID: 40282274 PMCID: PMC12024606 DOI: 10.3390/biology14040409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
A dysregulated nucleotide metabolism has been implicated in the pathogenesis of diabetic retinopathy (DR). RNA sequencing datasets, GSE102485, GSE60436, and GSE165784, were downloaded from the GEO database. The differentially expressed genes (DEGs) between the DR and controls overlapped with nucleotide metabolism-related genes (NM-RGs), resulting in the differentially expressed NM-RGs (DE-NMRGs). Next, the core genes were identified by the five algorithms of the CytoHubba plugin. Receiver Operating Characteristic (ROC) curves and gene expression analysis were utilized to confirm the biomarkers. Then, the correlations between biomarker expression and the immune-related module were analyzed. The miRNA and transcription factor (TF) predictions, biomarker-targeting drugs, and molecular docking were implemented separately. The interaction between each subcluster of DR was elucidated through single-cell RNA (scRNA) analysis. Moreover, RT-PCR was applied to verify the expression of the biomarkers. In GSE102485, 48 DE-NMRGs were identified via the intersection of 1359 DEGs and 882 NM-RGs. Using the CytoHubba plugin, HMOX1, TLR4, and ACE were selected as core genes. As per the GSVA result, the interferon alpha response, IL6_JAK_STAT3 signaling, and apoptosis were activated in the DR group. The TF prediction identified TLR4 and HMOX1 as potential target genes of USF2. In conclusion, ACE and HMOX1 were possible diagnostic biomarkers related to nucleotide metabolism in DR.
Collapse
Affiliation(s)
- Wei Wang
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China;
- Anhui Public Health Clinical Center, Hefei 230022, China
| | - Jianyang Gong
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China;
- Anhui Public Health Clinical Center, Hefei 230022, China
| |
Collapse
|
12
|
Muszka Z, Jenei V, Mácsik R, Mezhonova E, Diyab S, Csősz R, Bácsi A, Mázló A, Koncz G. Life-threatening risk factors contribute to the development of diseases with the highest mortality through the induction of regulated necrotic cell death. Cell Death Dis 2025; 16:273. [PMID: 40216765 PMCID: PMC11992264 DOI: 10.1038/s41419-025-07563-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025]
Abstract
Chronic diseases affecting the cardiovascular system, diabetes mellitus, neurodegenerative diseases, and various other organ-specific conditions, involve different underlying pathological processes. However, they share common risk factors that contribute to the development and progression of these diseases, including air pollution, hypertension, obesity, high cholesterol levels, smoking and alcoholism. In this review, we aim to explore the connection between four types of diseases with different etiologies and various risk factors. We highlight that the presence of risk factors induces regulated necrotic cell death, leading to the release of damage-associated molecular patterns (DAMPs), ultimately resulting in sterile inflammation. Therefore, DAMP-mediated inflammation may be the link explaining how risk factors can lead to the development and maintenance of chronic diseases. To explore these processes, we summarize the main cell death pathways activated by the most common life-threatening risk factors, the types of released DAMPs and how these events are associated with the pathophysiology of diseases with the highest mortality. Various risk factors, such as smoking, air pollution, alcoholism, hypertension, obesity, and high cholesterol levels induce regulated necrosis. Subsequently, the release of DAMPs leads to chronic inflammation, which increases the risk of many diseases, including those with the highest mortality rates.
Collapse
Affiliation(s)
- Zsuzsa Muszka
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Viktória Jenei
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
- Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Rebeka Mácsik
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Evgeniya Mezhonova
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Silina Diyab
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Réka Csősz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary
| | - Anett Mázló
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| | - Gábor Koncz
- Department of Immunology, Faculty of Medicine, University of Debrecen, Egyetem square 1, 4032, Debrecen, Hungary.
| |
Collapse
|
13
|
Zhang Y, Su F, Zhu E, Sun Y, Kuang H, Wang Q. A systematical review on traditional Chinese medicine treating chronic diseases via regulating ferroptosis from the perspective of experimental evidence and clinical application. CHINESE HERBAL MEDICINES 2025; 17:246-260. [PMID: 40256717 PMCID: PMC12009076 DOI: 10.1016/j.chmed.2025.01.003] [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: 06/21/2024] [Revised: 09/12/2024] [Accepted: 01/20/2025] [Indexed: 04/22/2025] Open
Abstract
Ferroptosis is a unique regulated form of cell death that is distinct from apoptosis, necrosis, and other well-characterized regulated cell death types, and plays an important role in the occurrence and development of chronic metabolic diseases, including diabetes, hypertension, hyperlipidemia, and non-alcoholic steatohepatitis. Recently, increasing evidence has supported traditional Chinese medicine (TCM) as a new hot spot for the treatment of chronic metabolic diseases by mediating ferroptosis. Unfortunately, few systematic reviews have described the importance of TCM in treating chronic metabolic diseases through the ferroptosis pathway. In the current review, the mechanism of ferroptosis and the roles of ferroptosis in chronic metabolic diseases are summarized. Additionally, this review illustrates that the regulation of ferroptosis by TCM could be an effective approach for treating chronic metabolic diseases based on experimental evidence and clinical application. In summary, this work will improve the understanding of ferroptosis and the ability of TCM to regulate ferroptosis in chronic metabolic diseases, thereby promoting the development and application of natural TCM.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Fazhi Su
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Enlin Zhu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Qiuhong Wang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 511400, China
| |
Collapse
|
14
|
Shen Y, Zhang C, Jiang X, Li X, Chen B, Jiang W. Capsiate attenuates atherosclerosis by activating Nrf2/GPX4 pathway and reshaping the intestinal microbiota in ApoE -/- mice. Microbiol Spectr 2025; 13:e0315524. [PMID: 40029381 PMCID: PMC11960139 DOI: 10.1128/spectrum.03155-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025] Open
Abstract
Atherosclerosis (AS) is the basis of cardiovascular diseases (CVDs) and remains the major contributor to death worldwide. Capsiate is derived from sweet pepper fruit and exhibits numerous pharmacological activities. The objective of this study was to elucidate the protective role of capsiate in atherosclerosis by examining its effect and the underlying regulatory pathways. Here, we showed that capsiate treatment alleviates atherosclerosis in atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice. We found that capsiate effectively reduced the plaque area and body weight compared to the Model group. Capsiate inhibited inflammatory response by downregulating phosphoinositide 3-kinase/protein kinase B/nuclear factor-κB pathway. Additionally, further investigation indicated that capsiate could regulate lipid levels in mice via reducing the expressions of 3-hydroxy-3-methylglutaryl coenzyme A reductase and low-density lipoprotein receptor, and increasing the expression of recombinant cytochrome P450 7A1. Furthermore, capsiate effectively activated transient receptor potential vanilloid subfamily member 1 in ApoE-/- mice fed a high-fat diet. The microbial sequencing demonstrated capsiate administration significantly regulated the gut microbiota disturbance and increased some beneficial bacterial (Lachnospiraceae NK4A136 group) levels in ApoE-/- mice. Human umbilical vein endothelial cells (HUVECs) were exposed to oxidized low-density lipoprotein (ox-LDL) to stimulate atherosclerotic endothelial damage in vitro. Our study revealed that capsiate inhibited ox-LDL-induced HUVECs injury and inflammation. We further investigated the effects of capsiate on ferroptosis in vivo and in vitro; it was found that capsiate exhibited anti-ferroptosis through regulating nuclear factor erythroid 2-related factor 2/glutathione peroxidase 4 pathway. Interestingly, ML385 reversed the anti-ferroptosis effect of capsiate in HUVECs. Taken together, our findings suggest a promising use of small-molecule drug capsiate for the treatment of AS and related CVDs. IMPORTANCE Capsiate has been found to inhibit fat accumulation, promote energy metabolism, and exhibit anti-inflammatory and antioxidative properties. However, there has still been no study on the ferroptosis and gut microbiota of capsiate in atherosclerosis (AS) mouse models. Our study is the first to report on the reshaping of the structure of the gut microbiota by capsiate in AS, and to explore the potential mechanism underlying the improvement of AS. In this study, we demonstrated that capsiate could effectively alleviate high-fat diet-induced AS in apolipoprotein E-deficient mice by inhibiting inflammatory response, improving serum lipid profiles, activating transient receptor potential vanilloid subfamily member 1 pathway, and suppressing ferroptosis. Moreover, the study reported the potential of gut microbiota as mediators of capsiate therapy for AS in animal models. Therefore, these findings may provide robust experimental support for the clinical use of capsiate for AS treatment.
Collapse
Affiliation(s)
- Yongbin Shen
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuanqi Zhang
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue Jiang
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xianwei Li
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Chen
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weiliang Jiang
- Department of Vascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
15
|
Wu M, Zhang S, Wu X, Zhou Y, Zhou M, Du A, Zhang Y, Wei T, Wang B, Wang S, Jiang C, Hu S, Xiao J, Wu Y. Acute hyperglycemia impedes spinal cord injury recovery via triggering excessive ferroptosis of endothelial cells. Int J Biol Macromol 2025; 301:140453. [PMID: 39884601 DOI: 10.1016/j.ijbiomac.2025.140453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Spinal cord injury (SCI) is a serious central nervous system injury that often causes sensory and motor dysfunction in patients. Diabetes seriously destroys the blood spinal cord barrier (BSCB) and aggravates SCI. Ferroptosis is a new type of programmed cell death. The role of ferroptosis in diabetes-medicated BSCB destruction has not been clearly elucidated. Here, we built a type 1 diabetes (T1D) combined with SCI rat model and confirmed that hyperglycemia exacerbates SCI-mediated BSCB destruction. Pathological mechanism demonstrated that except for apoptosis, the excessive ferroptosis is another caused factor for endothelial cells (ECs) loss under hyperglycemic condition. More importantly, ferrostatin-1(a ferroptosis inhibitor) treatment significantly inhibited the ferroptosis of ECs, and promoted the BSCB repair in T1D combined with SCI rat. The mechanism study further revealed that hyperglycemia not only induces the elevated reactive oxygen species (ROS) via activating RAGE, but also suppresses the xCT expression in system Xc- in ECs, which decreases GPX4 expression and induces ferroptosis. Additionally, hyperglycemia also accelerated the transfer of iron ions from serum to spinal cord after SCI. In summary, our results suggest that the excessive ferroptosis of ECs is essential for the severe BSCB destruction in T1D combined with spinal cord injury rat.
Collapse
Affiliation(s)
- Man Wu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China; The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China; National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Susu Zhang
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Xuejuan Wu
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Yongxiu Zhou
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Mei Zhou
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China
| | - Anyu Du
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Yanren Zhang
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China
| | - Tao Wei
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China
| | - Beini Wang
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Shuangshuang Wang
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Chang Jiang
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China
| | - Siwang Hu
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China.
| | - Jian Xiao
- The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China.
| | - Yanqing Wu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Wenzhou University, Wenzhou 325035, China; The Orthopaedic Center, The First People's Hospital of Wenling, Affiliated Wenling Hospital and School of Pharmaceutical Science, Wenzhou Medical University, Taizhou 317500, China.
| |
Collapse
|
16
|
Wang T, Chen X, Li C. SRC involves in lysosomal function and regulates ferroptosis in polycystic ovary syndrome. J Ovarian Res 2025; 18:42. [PMID: 40045372 PMCID: PMC11881414 DOI: 10.1186/s13048-025-01637-y] [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: 12/22/2024] [Accepted: 02/23/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The pathogenesis of polycystic ovary syndrome (PCOS) is still unknown, so finding the molecular mechanisms of pathogenesis is crucial in PCOS. METHODS The GSE34526 dataset from the Gene Expression Omnibus (GEO) database was used to screen biomarkers in this study. KEGG enrichment analysis of GSE34526 was performed using Gene Set Enrichment Analysis (GSEA). The differentially expressed genes(DEGs) were screened and analyzed for lysosome-related genes. Subsequently, further KEGG and GO analyses were performed, and it was found that it was enriched in the ferroptosis pathway, and then the ferroptosis-related differential genes were obtained. The genes at the core position were obtained by the Protein-Protein Interaction(PPI) network. We then focused our attention on SRC and verified the differential expression of SRC in ovarian tissues of hyperandrogenemic, hyperlipemic and control groups, as well as the differences in conception rate and litter rate of each group by rat test. RESULTS GSEA analysis of the gene dataset GSE34526 revealed that LYSOSOME was significantly enriched in the PCOS group. There were 188 lysosome-related differentially expressed genes(LRDEGs) in granulosa cells from patients with PCOS, and 41 ferroptosis-related differentially expressed genes(FRDEGs). It was found that six of these genes, SRC, NCF2, SLC2A8, FTL, SLC2A6, SLC3A2, were present in all three datasets. SRC was the top ranked gene in the PPI network of FRDEGs.As verified by the rat model, the expression of SRC in the ovarian tissues of the hyperandrogenemic group was significantly higher than that of the control group (P=0.004) and the hyperlipemic group (P=0.002). CONCLUSION SRC, as an important gene involved in lysosomal function and regulating ferroptosis, is expected to be a potential target for PCOS.
Collapse
Affiliation(s)
- Tianmei Wang
- Department of Gynecology, First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400016, P.R. China
| | - Xin Chen
- Department of Gynecology and Obstetrics, The 958th Army Hospital of the Chinese People'S Liberation Army, Jiangbei District, Chongqing, 400000, P.R. China
| | - Cong Li
- Department of Gynecology, First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400016, P.R. China.
| |
Collapse
|
17
|
Zhou T, Zhang Q, Yu W, Cui Y, Yan J, Ni T, Fu X, Li J. Exploration of ferroptosis-related biomarkers with prognostic capability in RIF based on WGCNA. J Assist Reprod Genet 2025; 42:949-959. [PMID: 39715942 PMCID: PMC11950601 DOI: 10.1007/s10815-024-03370-9] [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: 09/10/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
PURPOSE To explore the association of ferroptosis with repeated implantation failure (RIF) and prognostic capability of ferroptosis-related genes. METHODS Data in GSE106602 from the GEO database were used for gene co-expression network construction to confirm ferroptosis-related genes compared to gene sets that were downloaded from FerrDB. Then these genes were analyzed for functional enrichment and validated using endometrium samples from our center. ImplantScore and ROC curve were constructed for prognostic correlation analysis. RESULTS We observed that ferroptosis probably participated in RIF according to bioinformatics analysis on a gene set which exhibited a strong association with RIF from WGCNA. Fifty-four ferroptosis-related genes in the gene set were subsequently verified, and the PPI network was established for underlying interactions among them. There were 23 hub genes with differential expression in RIF and six of them (PML, LCN2, PRKAA1, BACH1, SLC7A11, and CAMKK2) showed significant correlation with implantation outcomes using samples collected from our center. Therefore, we combined the six genes and constructed an ImplantScore whose AUC reached 0.891, higher than the AUC of each single gene, respectively. ImplantScore of six genes with down-regulated expression in the group with failed implantation were much lower than that with successful outcome. CONCLUSION Our results demonstrated the potential prognostic functions of ferroptosis-related biomarkers in RIF, which will provide novel perspectives for further research and clinical applications.
Collapse
Affiliation(s)
- Tingting Zhou
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qian Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Wenhao Yu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Yuqian Cui
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Junhao Yan
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Tianxiang Ni
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Xiaohua Fu
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Junwei Li
- Department of Anatomy, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
18
|
Zhou L, Li L, Yang J, Mansuer M, Deng X, Wang Y, Ren H, Cui D, Jiang Y, Gao L. TNFAIP3 affects ferroptosis after traumatic brain injury by affecting the deubiquitination and ubiquitination pathways of the HMOX1 protein and ACSL3. Free Radic Biol Med 2025; 228:221-239. [PMID: 39743027 DOI: 10.1016/j.freeradbiomed.2024.12.048] [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: 02/06/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
The occurrence and progression of traumatic brain injury involve a complex process. The pathophysiological mechanisms triggered by neuronal damage include various forms of programmed cell death, including ferroptosis. We observed upregulation of TNFAIP3 in mice after traumatic brain injury. Overexpression of TNFAIP3 inhibits HT-22 proliferation and cell viability through ferroptosis. Mechanistically, TNFAIP3 interacts with the HMOX1 protein and promotes its stability through the deubiquitination pathway. Additionally, TNFAIP3 can enhance lipoperoxidation, mitochondrial damage, and neuronal cell death by promoting ACSL3 degradation via NEDD4-mediated ubiquitination. Mice injected with AAV-shTNFAIP3 exhibited reduced neuronal degeneration and improved motor and cognitive function following cortical impact injury. In conclusion, our findings demonstrate that TNFAIP3 deficiency inhibits neuronal cell ferroptosis and ameliorates cognitive impairment caused by traumatic brain injury and demonstrate its potential applicability in the treatment of traumatic brain injury.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Lei Li
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jinghao Yang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Maierdan Mansuer
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xianyu Deng
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yida Wang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hui Ren
- Department of Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200435, China
| | - Daming Cui
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yang Jiang
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Liang Gao
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
19
|
Wang Z, Wu C, Yin D, Dou K. Ferroptosis: mechanism and role in diabetes-related cardiovascular diseases. Cardiovasc Diabetol 2025; 24:60. [PMID: 39920799 PMCID: PMC11806630 DOI: 10.1186/s12933-025-02614-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
Cardiovascular diseases represent the principal cause of death and comorbidity among people with diabetes. Ferroptosis, an iron-dependent non-apoptotic regulated cellular death characterized by lipid peroxidation, is involved in the pathogenesis of diabetic cardiovascular diseases. The susceptibility to ferroptosis in diabetic hearts is possibly related to myocardial iron accumulation, abnormal lipid metabolism and excess oxidative stress under hyperglycemia conditions. Accumulating evidence suggests ferroptosis can be the therapeutic target for diabetic cardiovascular diseases. This review summarizes ferroptosis-related mechanisms in the pathogenesis of diabetic cardiovascular diseases and novel therapeutic choices targeting ferroptosis-related pathways. Further study on ferroptosis-mediated cardiac injury can enhance our understanding of the pathophysiology of diabetic cardiovascular diseases and provide more potential therapeutic choices.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Wu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Yin
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Kefei Dou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Cardiometabolic Medicine Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
20
|
Liu Q, Han Z, Li T, Meng J, Zhu C, Wang J, Wang J, Zhang Z, Wu H. Microglial HO-1 aggravates neuronal ferroptosis via regulating iron metabolism and inflammation in the early stage after intracerebral hemorrhage. Int Immunopharmacol 2025; 147:113942. [PMID: 39740507 DOI: 10.1016/j.intimp.2024.113942] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
Heme oxygenase 1 (HO-1), an enzyme involved in heme catabolism, has been shown upregulated in microglia cells and plays a critical roles in neurological damages after intracerebral hemorrhage (ICH). However, the mechanisms by which HO-1 mediates the neuronal damages are still obscure. Here, our findings demonstrate that HO-1 over-expression exacerbates the pro-inflammatory response of microglia and induces neuronal ferroptosis through promoting intracellular iron deposition in the ICH model both in vitro and in vivo. Furthermore, in the co-cultured ICH model in vitro, we verify that HO-1 over-expression disrupts the balance of iron metabolism in microglia, which increases the iron efflux to the extracellular space and promotes iron ion uptake in neurons, leading to lipid peroxidation injury and further contributing to neuronal ferroptosis. Moreover, the specific ferroptosis inhibitor Ferrostatin-1 (Fer-1) treatment could mitigate the damages in the co-cultured HT22 cells that caused by HO-1 over-expression in microglia, and improve the neurological function in the ICH model in mice. By shedding light on the mechanisms of aggravating neuronal ferroptosis due to HO-1 overexpression in the early stages after ICH, our study provides insights into the potential therapy of targeting HO-1 to treat ICH.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ziyi Han
- College of Medical Laboratory Technology, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Tao Li
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jincheng Meng
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Chenwei Zhu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jian Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Zhen Zhang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - He Wu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
21
|
Dong H, Zhu L, Sun J, Chen Q, Liu P, Zhang W, Zeng H, Lin R, Yu Z, Lu J. IFNβ drives ferroptosis through elevating TRIM22 and promotes the cytotoxicity of RSL3. Front Immunol 2025; 16:1535554. [PMID: 39975542 PMCID: PMC11836015 DOI: 10.3389/fimmu.2025.1535554] [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: 11/27/2024] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
Background Cyclic GMP-AMP synthase (cGAS)-stimulator-of-interferon genes (STING) pathway is a cytosolic DNA sensor system. The production of this pathway, interferon-β (IFNβ), could suppress the growth of tumor cells, yet it is unclear whether ferroptosis is involved in IFNβ-induced cell death. Methods The effects of IFNβ on ferroptosis were analyzed in HT1080, 4T1, HCT116 and 786-O cells. HT1080 and 4T1 cells treated with IFNβ were subjected to RNA-Seq analysis. STAT1, STAT3, TRIM21, and TRIM22 were silenced by siRNAs to examine their effects on IFNβ-induced ferroptosis. The cGAS-STING signaling pathway-activated mice were used to evaluate the effects of IFNβ on ferroptosis in vivo. HT1080 cells, three-dimensional (3D) spheroids, and the xenograft mouse models were treated with IFNβ, RSL3, or IFNβ combination with RSL3 to analyze whether IFNβ enhances RSL3-induced ferroptosis. Results Here, we found that IFNβ could promote intracellular Fe2+ and lipid peroxidation levels, and decrease GSH levels in tumor cells. RNA sequencing data revealed that IFNβ induced a transcriptomic disturbance in ferroptosis-related genes. Knockdown of tripartite motif-containing 22 (TRIM22) suppressed the levels of intracellular Fe2+ and lipid ROS. It also reduced heme oxygenase (HMOX1) protein levels and increased ferroptosis suppressor protein 1 (FSP1) levels in HT1080 cells treated with IFNβ. Furthermore, our results illustrated that IFNβ enhanced the RAS-selective lethal 3 (RSL3)-induced ferroptosis and the inhibitory effect of RSL3 on GPX4. Meanwhile, compared to the groups treated with either IFNβ or RSL3 alone, the combination treatment of IFNβ and RSL3 significantly inhibited the growth of HT1080 three-dimensional (3D) spheroids and tumor in a mouse xenograft model. Conclusions Our work reveals a role for IFNβ in promoting ferroptosis and provides evidence that IFNβ could be used with RSL3 to increase cytotoxic effects in tumor cells.
Collapse
Affiliation(s)
- Huiyue Dong
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Ling Zhu
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jingjing Sun
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Clinical Laboratory, Wuhan Children’s Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyan Chen
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Pengyang Liu
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wei Zhang
- Fuzong Teaching Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Huajing Zeng
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Rong Lin
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Zongyang Yu
- Fuzong Teaching Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Pulmonary and Critical Care Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Fujian Provincial Key Laboratory of Transplant Biology, Dongfang Hospital (the 900th Hospital of Joint Logistic Support Force), Xiamen University, Fuzhou, China
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
- Fuzong Teaching Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Organ transplant institute, Dongfang Hospital, Xiamen University, Fuzhou, China
| |
Collapse
|
22
|
Li Q, Yu X, Yu R, Shi X, Lu Y. Therapeutic Potential of Inhibiting Hmox1 in Sepsis-Induced Lung Injury: A Molecular Mechanism Study. J Biochem Mol Toxicol 2025; 39:e70134. [PMID: 39959930 DOI: 10.1002/jbt.70134] [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: 10/23/2024] [Revised: 12/11/2024] [Accepted: 12/29/2024] [Indexed: 05/09/2025]
Abstract
Sepsis induces severe multiorgan dysfunction, with the lungs being particularly susceptible to damage. This study reveals that Hmox1 inhibitors effectively activate the FSP1/CoQ10/NADPH pathway, significantly enhancing autophagic activity while suppressing ferroptosis in alveolar epithelial cells, thereby alleviating lung injury in septic mice. To identify key gene modules and regulatory factors associated with sepsis-induced lung injury, we analyzed public transcriptomic data, including bulk RNA-seq datasets (GSE236391 and GSE263867) and a single-cell RNA-seq (scRNA-seq) data set (GSE207651). In vitro experiments were conducted using an LPS-induced alveolar epithelial cell injury model to evaluate the effects of Hmox1 inhibitors on cell viability, autophagy markers (LC3-II/LC3-I and p62), ROS levels, and intracellular iron content. Transmission electron microscopy was used to observe mitochondrial structural changes. In vivo, a cecal ligation and puncture (CLP)-induced sepsis mouse model was established to assess the therapeutic effects of Hmox1 inhibitors. This included evaluating survival rates, lung histopathological scores, lung wet-to-dry weight ratios, myeloperoxidase (MPO) activity, inflammatory cytokine levels, and changes in autophagy and ferroptosis markers. The results demonstrated that Hmox1 inhibitors effectively mitigate lung injury by modulating the autophagy-ferroptosis pathway, highlighting their potential as a therapeutic strategy for sepsis-induced lung damage.
Collapse
Affiliation(s)
- Qingying Li
- Department of Critical Care Medicine, Xinyang Central Hospital, Xinyang, China
| | - Xu Yu
- Department of Critical Care Medicine, Xinyang Central Hospital, Xinyang, China
| | - Renjie Yu
- Department of Critical Care Medicine, Xinyang Central Hospital, Xinyang, China
| | - Xinge Shi
- Department of Critical Care Medicine, Xinyang Central Hospital, Xinyang, China
| | - Yibin Lu
- Department of Critical Care Medicine, Xinyang Central Hospital, Xinyang, China
| |
Collapse
|
23
|
Wang S, Zhuo D, Lin J, Zhang C. Key Genes and Biological Pathways in Pulmonary Arterial Hypertension Related to Endoplasmic Reticulum Stress Identified by Bioinformatics. J Cardiovasc Pharmacol 2025; 85:108-119. [PMID: 39907642 DOI: 10.1097/fjc.0000000000001651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/30/2024] [Indexed: 02/06/2025]
Abstract
ABSTRACT Pulmonary arterial hypertension (PAH) is a cardiopulmonary vascular condition with an unclear pathogenesis. Targeting endoplasmic reticulum (ER) stress has been suggested as a novel treatment approach for PAH, but the mechanisms involving ER stress-related genes in PAH are not well understood. Microarray data for PAH and ER stress-related genes were analyzed. Differential and Venn analyses identified 17 differentially expressed ER stress-related genes in PAH. Candidate drugs targeting these genes were predicted using the CMap database. A protein-protein interaction (PPI) network was constructed, and hub genes (LCN2, IGF1, VCAM1, EDN1, HMOX1, TLR4) with complex interplays were identified using the STRING database and Cytoscape plugins. The clinical diagnostic performance of the hub genes was evaluated using ROC curves. The GeneMANIA Web site was utilized to predict enriched pathways associated with the hub genes and their functionally similar genes. MiRNAs and transcription factors targeting the hub genes were predicted using the Networkanalyst Web site. The immune levels in control samples and PAH samples were assessed using various algorithms. Nine drug candidates were found to potentially target the identified ER stress-related genes. The hub genes and their correlated genes were significantly enriched in immune-related pathways. The PAH group showed increased immune cell infiltration, indicating a heightened immune response. This study sheds light on the role of ER stress-associated hub genes in PAH and proposes potential drugs targeting these genes. These findings provide valuable insights into PAH mechanisms and support the exploration of ER stress as a therapeutic target.
Collapse
Affiliation(s)
| | - Debin Zhuo
- Respiratory and Critical Care Medicine, The Affiliated Hospital of Putian University, Putian City, China
| | - Juan Lin
- Respiratory and Critical Care Medicine, The Affiliated Hospital of Putian University, Putian City, China
| | - Chunxia Zhang
- Respiratory and Critical Care Medicine, The Affiliated Hospital of Putian University, Putian City, China
| |
Collapse
|
24
|
Wang E, Li S, Li Y, Zhou T. HMOX1 as a potential drug target for upper and lower airway diseases: insights from multi-omics analysis. Respir Res 2025; 26:41. [PMID: 39871287 PMCID: PMC11773792 DOI: 10.1186/s12931-025-03124-w] [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: 09/30/2024] [Accepted: 01/20/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Oxidative stress is key in inflammatory airway diseases. Heme oxygenase 1 (HMOX1) regulates oxidative stress, but its role in airway diseases needs exploration. METHODS Differentially expressed genes (DEGs) between healthy nasal mucosa and chronic rhinosinusitis with nasal polyps (CRSwNP) were identified from Gene Expression Omnibus (GEO). Candidate genes were further screened using Gene Set Enrichment Analysis (GSEA) and Random Forest (RF) algorithms. Causal inference between candidate genes and upper and lower airway diseases (CRSwNP, allergic rhinitis (AR), and asthma (AS)) was conducted using bidirectional two-sample Mendelian randomization (TwoSampleMR) analysis. Single-cell RNA sequencing (scRNA-seq) data were used to determine the cellular localization and intercellular interactions of candidate genes. Molecular docking was used to identify potential therapeutic agents. RESULTS HMOX1 expression was significantly elevated in CRSwNP. TwoSampleMR analysis indicated a negative causal relationship between HMOX1 exposure and the occurrence of upper and lower airway diseases (CRSwNP [(odds ratio (OR)/95% confidence interval (CI): 0.945/(0.893-0.999), P = 0.044], AR [OR/95% CI: 0.997/(0.994-0.999), P = 0.007], and AS [OR/95% CI: 0.935/(0.895-0.977), P = 0.003]). scRNA-seq data revealed HMOX1 localization in M2 macrophages. Molecular docking identified 15 antioxidants, including Acetylcysteine and Quercetin, that can upregulate HMOX1 expression. CONCLUSION HMOX1 may have a protective role in the pathogenesis of upper and lower airway diseases (CRSwNP, AR, and AS) by modulating oxidative stress. Antioxidants that increase HMOX1 expression could offer new therapeutic avenues for these diseases. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
- Enhao Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Shazhou Li
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Yang Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.
| | - Tao Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| |
Collapse
|
25
|
Lu Y, Zhu F, Zhou X, Li Y, Rong G, Liu N, Hong J, Cheng Y. A Supramolecular Deferoxamine-Crisaborole Nanoparticle Targets Ferroptosis, Inflammation, and Oxidative Stress in the Treatment of Retinal Ischemia/Reperfusion Injury. NANO LETTERS 2025; 25:1058-1066. [PMID: 39670541 DOI: 10.1021/acs.nanolett.4c05012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Retinal ischemia-reperfusion (IR) injury is a major cause of vision loss worldwide, with ferroptosis, oxidative stress, and inflammation playing key roles in its pathogenesis. Currently, treatments targeting multiple aspects of this condition are limited. This study introduces a supramolecular nanoparticle combining the phosphodiesterase 4 (PDE4) inhibitor crisaborole and the ferroptosis inhibitor deferoxamine to address these pathological processes. Crisaborole forms a dynamic bond with deferoxamine via benzoxaborole-catechol chemistry, creating an amphiphilic molecule that assembles into nanoparticles. Treatment with these nanoparticles enhances glutathione peroxidase 4 (GPX4) levels, downregulates ferroptosis-related genes [Acyl-CoA synthetase long chain family member 4 (Acsl4), heme oxygenase 1 (Hmox1)], reduces inflammatory markers (interleukin-1 beta, interleukin-6, tumor necrosis factor alpha), and decreases reactive oxygen species. Electroretinogram and histochemical analysis confirm the nanoparticles' superior protective effects compared to control treatments. This study proposes a novel nanoparticle approach for retinal IR injury by simultaneously targeting multiple pathogenic pathways.
Collapse
Affiliation(s)
- Yiteng Lu
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, China
| | - Fang Zhu
- Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xujiao Zhou
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, China
| | - Yuhan Li
- Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Guangyu Rong
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, China
- Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Nan Liu
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, China
- Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiaxu Hong
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, China
- NHC Key laboratory of Myopia and Related Eye Diseases, Shanghai, 200031, China
- Shanghai Engineering Research Center of Synthetic Immunology, Shanghai, 200032, China
- Department of Ophthalmology, Children's Hospital of Fudan University, National Pediatric Medical Center of China, Shanghai, 201102, China
| | - Yiyun Cheng
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200031, China
- Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| |
Collapse
|
26
|
Jia Y, Li R, Li Y, Kachler K, Meng X, Gießl A, Qin Y, Zhang F, Liu N, Andreev D, Schett G, Bozec A. Melanoma bone metastasis-induced osteocyte ferroptosis via the HIF1α-HMOX1 axis. Bone Res 2025; 13:9. [PMID: 39814705 PMCID: PMC11735842 DOI: 10.1038/s41413-024-00384-y] [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: 05/22/2024] [Revised: 09/11/2024] [Accepted: 10/15/2024] [Indexed: 01/30/2025] Open
Abstract
Osteocytes are the main cells in mineralized bone tissue. Elevated osteocyte apoptosis has been observed in lytic bone lesions of patients with multiple myeloma. However, their precise contribution to bone metastasis remains unclear. Here, we investigated the pathogenic mechanisms driving melanoma-induced osteocyte death. Both in vivo models and in vitro assays were combined with untargeted RNA sequencing approaches to explore the pathways governing melanoma-induced osteocyte death. We could show that ferroptosis is the primary mechanism behind osteocyte death in the context of melanoma bone metastasis. HMOX1 was identified as a crucial regulatory factor in this process, directly involved in inducing ferroptosis and affecting osteocyte viability. We uncover a non-canonical pathway that involves excessive autophagy-mediated ferritin degradation, highlighting the complex relationship between autophagy and ferroptosis in melanoma-induced osteocyte death. In addition, HIF1α pathway was shown as an upstream regulator, providing a potential target for modulating HMOX1 expression and influencing autophagy-dependent ferroptosis. In conclusion, our study provides insight into the pathogenic mechanisms of osteocyte death induced by melanoma bone metastasis, with a specific focus on ferroptosis and its regulation. This would enhance our comprehension of melanoma-induced osteocyte death.
Collapse
Affiliation(s)
- Yewei Jia
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Rui Li
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixuan Li
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katerina Kachler
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xianyi Meng
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andreas Gießl
- Department of Opthalmology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Yi Qin
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Fulin Zhang
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ning Liu
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Darja Andreev
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Technische Universität Dresden (TUD), Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
27
|
Yang H, Ye F, Chen L, Yang L, Kang J. EGCG attenuated spinal cord injury by inhibiting ferroptosis via activation of HMOX1 expression and suppression of HIF-1 signaling pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:799-807. [PMID: 40343292 PMCID: PMC12057747 DOI: 10.22038/ijbms.2025.82651.17864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/18/2025] [Indexed: 05/11/2025]
Abstract
Objectives Epigallocatechin gallate (EGCG) exhibits various biological effects, including antiviral, anti-inflammatory, cardioprotective, and lipid-regulating properties. This study aims to investigate the therapeutic effects and mechanisms of EGCG in spinal cord injury (SCI). Materials and Methods The bioinformatic databases were used to screen therapeutic target genes for drugs against SCI. Component-Target-Disease networks were constructed with Cytoscape software, and inter-target interactions were analyzed using the String database. Additionally, KEGG pathway enrichment analyses were conducted on the identified target genes. SCI was evaluated by detecting inflammation-related factors, H&E staining, and immunohistochemistry. Furthermore, ROS and JC1 staining were performed on HT22 cells subjected to various treatments. Molecular mechanisms were investigated using western blot and qRT-PCR analyses. Results Forty-four overlapping genes were identified as potential targets, with HMOX1, GPX-4, and HIF-1A emerging as central hub genes. Key pathways associated with these targets included Ferroptosis and HIF-1 signaling. In vivo studies demonstrated that EGCG effectively promotes motor function recovery and reduces the expression of proteins and genes such as IL-1β, IL-6, HIF-1α, and 4HNE. In vitro experiments showed that EGCG decreases ROS and intracellular lipid ROS levels in HT22 cells while increasing GPX-4 and HMOX1 expression to inhibit ferroptosis and HIF-1 signaling pathways. Conclusion Our findings reveal a significant new mechanism by which EGCG can reduce SCI through the inhibition of ferroptosis, facilitated by the activation of HMOX1 expression and the down-regulation of the HIF-1 signaling pathway. This suggests its potential as a therapeutic option for this condition.
Collapse
Affiliation(s)
| | | | | | | | - Jianping Kang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, P.R. China
| |
Collapse
|
28
|
Wu J, Luo D, Tou L, Xu H, Jiang C, Wu D, Que H, Zheng J. NEK2 affects the ferroptosis sensitivity of gastric cancer cells by regulating the expression of HMOX1 through Keap1/Nrf2. Mol Cell Biochem 2025; 480:425-437. [PMID: 38503948 PMCID: PMC11695390 DOI: 10.1007/s11010-024-04960-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/05/2024] [Indexed: 03/21/2024]
Abstract
NEK2 is a serine/threonine protein kinase that is involved in regulating the progression of various tumors. Our previous studies have found that NEK2 is highly expressed in gastric cancer and suggests that patients have a worse prognosis. However, its role and mechanism in gastric cancer are only poorly studied. In this study, we established a model of ferroptosis induced by RSL3 or Erastin in AGS cells in vitro, and konckdown NEK2, HOMX1, Nrf2 by siRNA. The assay kit was used to analyzed cell viability, MDA levels, GSH and GSSG content, and FeRhoNox™-1 fluorescent probe, BODIPY™ 581/591 C11 lipid oxidation probe, CM-H2DCFDA fluorescent probe were used to detected intracellular Fe2+, lipid peroxidation, and ROS levels, respectively. Calcein-AM/PI staining was used to detect the ratio of live and dead cells, qRT-PCR and Western blot were used to identify the mRNA and protein levels of genes in cells, immunofluorescence staining was used to analyze the localization of Nrf2 in cells, RNA-seq was used to analyze changes in mRNA expression profile, and combined with the FerrDb database, ferroptosis-related molecules were screened to elucidate the impact of NEK2 on the sensitivity of gastric cancer cells to ferroptosis. We found that inhibition of NEK2 could enhance the sensitivity of gastric cancer cells to RSL3 and Erastin-induced ferroptosis, which was reflected in the combination of inhibition of NEK2 and ferroptosis induction compared with ferroptosis induction alone: cell viability and GSH level were further decreased, while the proportion of dead cells, Fe2+ level, ROS level, lipid oxidation level, MDA level, GSSG level and GSSG/GSH ratio were further increased. Mechanism studies have found that inhibiting NEK2 could promote the expression of HMOX1, a gene related to ferroptosis, and enhance the sensitivity of gastric cancer cells to ferroptosis by increasing HMOX1. Further mechanism studies have found that inhibiting NEK2 could promote the ubiquitination and proteasome degradation of Keap1, increase the level of Nrf2 in the nucleus, and thus promote the expression of HMOX1. This study confirmed that NEK2 can regulate HMOX1 expression through Keap1/Nrf2 signal, and then affect the sensitivity of gastric cancer cells to ferroptosis, enriching the role and mechanism of NEK2 in gastric cancer.
Collapse
Affiliation(s)
- Jianyong Wu
- Gastroenterology Department, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Desheng Luo
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Laizhen Tou
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Hongtao Xu
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Chuan Jiang
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Dan Wu
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Haifeng Que
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Jingjing Zheng
- Gastrointestinal Surgery, Lishui Municipal Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China.
| |
Collapse
|
29
|
Guo W, Yang H, He W. Paeonol alleviates ox-LDL-induced endothelial cell injury by targeting the heme oxygenase-1/phosphoinositide 3-kinase/protein kinase B pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:591-600. [PMID: 39037459 DOI: 10.1007/s00210-024-03307-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Atherosclerosis is a leading cause of vascular disease worldwide. Paeonol has been reported to have therapeutical potential in atherosclerosis. The aim of this study is to explore the effect of paeonol on oxidized low-density lipoprotein (ox-LDL)-induced endothelial cells injury and the underlying mechanism. Human umbilical vein endothelial cells (HUVECs) were treated with ox-LDL (100 μg/ml) to mimic atherosclerosis in vitro. The cell viability, proliferation, and apoptosis were assessed by cell counting kit-8 (CCK8), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry, respectively. The angiogenesis was detected by tube formation assay. The levels of inflammatory factor were measured by enzyme-linked immunosorbent assay (ELISA). In addition, the levels of Fe2+, reactive oxygen species (ROS), and glutathione (GSH) were detected to assess ferroptosis. The western blot was used to detect the protein expression. Ox-LDL inhibited cell viability, proliferation, and angiogenesis, but induced apoptosis and inflammation in HUVECs, and paeonol (75 μM) relieves ox-LDL-induced HUVEC injury. Also, paeonol inhibited ox-LDL-induced ferroptosis of HUVECs. Interestingly, heme oxygenase-1 (HMOX1) knockdown alleviated ox-LDL-induced HUVECs injury and ferroptosis. Paeonol affected ox-LDL-induced HUVECs via regulating HMOX1. In addition, paeonol regulated PI3K/AKT pathway via HMOX1, and the inhibitor of phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) pathway reversed the effects of HMOX1 knockdown on ox-LDL-induced HUVECs. Paeonol alleviated ox-LDL-induced HUVEC injury by regulating the PI3K/AKT pathway via targeting HMOX1.
Collapse
Affiliation(s)
- Weichong Guo
- Department of Cardiovascular Medicine, Nanyang First People's Hospital, Nanyang, 473000, China
| | - Han Yang
- Department of Geriatrics, Nanshi Hospital of Nanyang, Nanyang, 473065, China
| | - Wenguang He
- Department of Thyroid and Breast Surgery, The Fourth Affiliated Hospital, Guangzhou Medical University, No. 1, Guangming East Road, Zengjiang Street, Zengcheng District, Guangzhou, 511300, China.
| |
Collapse
|
30
|
Wang H, Xu X, Ouyang Y, Fei X, He C, Yang X, Ren Y, Zhou Y, Chen S, Hu Y, Liu J, Ge Z, Wu WKK, Lu N, Xie C, Wu X, Zhu Y, Li N. The Protective Role of DDIT4 in Helicobacter pylori-induced Gastric Metaplasia Through Metabolic Regulation of Ferroptosis. Cell Mol Gastroenterol Hepatol 2024; 19:101448. [PMID: 39943905 PMCID: PMC11937681 DOI: 10.1016/j.jcmgh.2024.101448] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 03/17/2025]
Abstract
BACKGROUND & AIMS Helicobacter pylori (H pylori) infection is a significant factor leading to gastric atrophy, metaplasia and cancer development. Here, we investigated the role of the stress response gene DDIT4 in the pathogenesis of H pylori infection. METHODS Cell lines, transgenic mice, and human tissue samples were implemented. Proteomics were performed on Ddit4+/+ and Ddit4-/- mice infected with H pylori strain PMSS1. C57BL/6 mice were administered with tamoxifen to induce gastric metaplasia. Stomach tissues were analyzed for histopathologic features, reactive oxygen species, Fe2+, lipid peroxidation, expression of DDIT4, and ferroptosis-related proteins. RESULTS DDIT4 expression was upregulated at 6 hours but significantly decreased at 24 hours in response to H pylori infection in gastric epithelial cells. Gastric DDIT4 were downregulated in INS-GAS mice at 4 months post H pylori infection. Notably, H pylori infection led to more severe gastric metaplasia lesion in Ddit4-knockout mice. The proteomic profiling revealed an increase in ferroptosis in the gastric tissues of infected Ddit4-deficient mice, compared with infected wild-type mice. Mechanistically, knockout of DDIT4 promoted H pylori-induced ferroptosis through the accumulation of lipid peroxides and ROS levels, and alterations in proteins such as GPX4, ALOX15, and HMOX1. Overexpression of DDIT4 counteracted H pylori-induced stem cell marker CD44V9 through modulation of ferroptosis. Similarly, in another mouse model of gastric metaplasia treated with tamoxifen, as well as in human GIM tissues, we observed the loss of DDIT4 and induction of ferroptosis. CONCLUSIONS Our results indicate that DDIT4 serves as a protective factor against H pylori-induced gastric metaplasia by metabolic resistance to ferroptosis.
Collapse
Affiliation(s)
- Huan Wang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Postdoctoral Innovation Practice Base, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xinbo Xu
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yaobin Ouyang
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiao Fei
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Cong He
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xianhe Yang
- Department of Science and Technology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuping Ren
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanan Zhou
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Sihai Chen
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yi Hu
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jianping Liu
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhongming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Chuan Xie
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xidong Wu
- Department of Drug Safety Evaluation, Jiangxi Testing Center of Medical Instruments, Nanchang, China.
| | - Yin Zhu
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Nianshuang Li
- Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Institute of Digestive Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
31
|
Yang Z, Zhang T, Zhu X, Zhang X. Ferroptosis-Related Transcriptional Level Changes and the Role of CIRBP in Glioblastoma Cells Ferroptosis. Biomedicines 2024; 13:41. [PMID: 39857625 PMCID: PMC11761263 DOI: 10.3390/biomedicines13010041] [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: 11/19/2024] [Revised: 12/24/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVE We aimed to elucidate the roles of ferroptosis-associated differentially expressed genes (DEGs) in glioblastoma and provide a comprehensive resource for researchers in the field of glioblastoma cell ferroptosis. METHODS We used RNA sequencing to identify the DEGs associated with erastin-induced ferroptosis in glioblastoma cells. We further unraveled the biological functions and clinical implications of cold-inducible RNA-binding protein (CIRBP) in the context of glioblastoma by using a multifaceted approach, encompassing gene expression profiling, survival analysis, and functional assays to elucidate its role in glioblastoma cell mortality and its potential influence on patient prognosis. RESULTS We identified and validated the gene encoding CIRBP, the expression of which is altered during glioblastoma ferroptosis. Our findings highlight the relationship between CIRBP expression and ferroptosis in glioblastoma cells. We demonstrated that CIRBP modulates key aspects of cell death, thereby altering the sensitivity of glioblastoma cells to erastin-induced ferroptosis. A prognostic model, constructed based on CIRBP expression levels, revealed an association between lower CIRBP levels and poorer prognosis in glioma patients; this finding was corroborated by our comprehensive in vitro and in vivo assays that highlighted the impact of modulating CIRBP expression on glioblastoma cell viability and ferroptotic response. CONCLUSION Our research unravels the complex molecular dynamics of ferroptosis in glioblastoma and underscores CIRBP as a potential biomarker and therapeutic target. This improved understanding of the role of CIRBP in ferroptosis paves the way for more precise and efficacious treatments for glioblastoma, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Zijiang Yang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
| | - Ting Zhang
- Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, Jiangyin 214400, China;
| | - Xuanlin Zhu
- School of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai 200433, China;
| | - Xiaobiao Zhang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Digital Medical Research Center, Fudan University, Shanghai 200032, China
| |
Collapse
|
32
|
Zeng Y, Fu S, Xia Y, Meng G, Xu X. Itchy E3 Ubiquitin Ligase-Mediated Ubiquitination of Ferritin Light Chain Contributes to Endothelial Ferroptosis in Atherosclerosis. Int J Mol Sci 2024; 25:13524. [PMID: 39769287 PMCID: PMC11677933 DOI: 10.3390/ijms252413524] [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: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
This research seeks to investigate the function and fundamental mechanisms of Itchy E3 ubiquitin ligase (ITCH), a HECT (homologous to E6AP carboxyl terminus)-type E3 ubiquitin ligase, in endothelial ferroptosis, particularly in the context of atherosclerosis, which has been underexplored. The levels of ITCH protein in the aortas of mice with atherosclerosis were analyzed. Constructs for ITCH RNA interference were generated and introduced into human aortic endothelial cells (HAECs). The findings indicated that ITCH protein expression was elevated in atherosclerotic mice and HAECs exposed to oxidized low-density lipoprotein (ox-LDL). ITCH downregulation significantly mitigated ox-LDL-induced endothelial injury and dysfunction. Reducing ITCH expression inhibited ox-LDL-induced endothelial ferroptosis. This study also revealed that ITCH mediates ox-LDL-induced ubiquitin-dependent degradation of ferritin light chain (FTL) in HAECs. The protective impact of ITCH knockdown against ox-LDL-induced ferroptosis and endothelial injury was reversed by FTL siRNA. Additionally, in vivo experiments showed that inhibiting ITCH reduced atherosclerosis progression and reversed ferroptosis in the aorta, with an associated increase in FTL protein expression in the aortas of mice. This study demonstrates that ITCH interacts with and regulates the stability of the FTL protein via the ubiquitin-proteasome system, contributing to ox-LDL-induced ferroptosis and endothelial cell dysfunction. Targeting components of the ITCH-FTL pathway holds potential as a therapeutic strategy against atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | - Xiaole Xu
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong 226001, China; (Y.Z.); (S.F.); (Y.X.); (G.M.)
| |
Collapse
|
33
|
Zhou X, Wang H, Yan B, Nie X, Chen Q, Yang X, Lei M, Guo X, Ouyang C, Ren Z. Ferroptosis in Cardiovascular Diseases and Ferroptosis-Related Intervention Approaches. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07642-5. [PMID: 39641901 DOI: 10.1007/s10557-024-07642-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Cardiovascular diseases (CVDs) are major public health problems that threaten the lives and health of individuals. The article has reviewed recent progresses about ferroptosis and ferroptosis-related intervention approaches for the treatment of CVDs and provided more references and strategies for targeting ferroptosis to prevent and treat CVDs. METHODS A comprehensive review was conducted using the literature researches. RESULTS AND DISCUSSION Many ferroptosis-targeted compounds and ferroptosis-related genes may be prospective targets for treating CVDs and our review provides a solid foundation for further studies about the detailed pathological mechanisms of CVDs. CONCLUSION There are challenges and limitations about the translation of ferroptosis-targeted potential therapies from experimental research to clinical practice. It warrants further exploration to pursure safer and more effective ferroptosis-targeted thereapeutic approaches for CVDs.
Collapse
Affiliation(s)
- Xianpeng Zhou
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Hao Wang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Biao Yan
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xinwen Nie
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Qingjie Chen
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiaosong Yang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Min Lei
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Xiying Guo
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Changhan Ouyang
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China
| | - Zhanhong Ren
- Hubei University of Science and Technology, Xianning, 437100, Hubei, People's Republic of China.
| |
Collapse
|
34
|
Hao J, Lu M, Zhao X, Li C, Ge C, Zhang J, Tu L, Zhang Q. Exploring the underlying mechanism by transcriptome sequencing in rats with high-voltage electrical burns and the role of iron metabolism. Burns 2024; 50:107226. [PMID: 39368958 DOI: 10.1016/j.burns.2024.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/24/2024] [Accepted: 07/28/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Clinically, the condition of skeletal muscle injury is the key to the process of high voltage electrical burn (HVEB) wound repair. The aim of this study was to identify the potential mechanisms and intervention targets of skeletal muscle injury after HVEB. METHODS A skeletal muscle injury model in SD rats with HVEB was made. Pathological examination and transcriptome sequencing of injured skeletal muscles were performed, and the expression levels of key proteins and genes in related signaling pathways were verified. RESULTS Skeletal muscle injury was progressively aggravated within 48 h, then the injury was gradually repaired with scar formation occurring within 1 week. The mechanism of skeletal muscle injury is complex and varied, and ferroptosis is one of the mechanisms. The ferrous iron content in the injured skeletal muscle tissue of model rats increased significantly at 24 h after injury. After 24 h, damage to injured skeletal muscle tissue could be alleviated by increasing iron storage and blocking lysosomal phagocytosis of autophagy. CONCLUSIONS Skeletal muscle injury caused by HVEB is characterized by adjacent endangered tissue progression after injury. Ferroptosis is involved in the mechanism of HVEB, and iron metabolism-related proteins may be potential targets for preventing progressive skeletal muscle injury.
Collapse
Affiliation(s)
- Jiawen Hao
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Mengyuan Lu
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Xuegang Zhao
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Congying Li
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Chenyang Ge
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Jing Zhang
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Lihong Tu
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China
| | - Qingfu Zhang
- Burn and Wound Repair Center, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province 050035, China.
| |
Collapse
|
35
|
Li Z, Gan H, Li S, Xue Y, Luo K, Huang K, Zhang Y, Wang Y, Jiang L, Zhang H. Bioinformatics Identification and Validation of Ferroptosis-Related Key Genes and Therapeutic Compounds in Septic Lung Injury. J Inflamm Res 2024; 17:9215-9230. [PMID: 39600675 PMCID: PMC11589777 DOI: 10.2147/jir.s476522] [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: 05/02/2024] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Background Septic lung injury (SLI) is a severe condition with high mortality, and ferroptosis, a form of programmed cell death, is implicated in its pathogenesis. However, the explicit mechanisms underlying this condition remain unclear. This study aimed to elucidate and validate key ferroptosis-related genes involved in the pathogenesis of SLI through bioinformatics analysis and experimental validation. Methods Microarray data related to SLI from the GSE130936 dataset were downloaded from the Gene Expression Omnibus (GEO) database. These data were then intersected with the FerrDb database to obtain ferroptosis-related differentially expressed genes (DEGs). Protein-protein interaction (PPI) networks and functional enrichment analysis were employed to identify key ferroptosis-related DEGs. The Connectivity Map (c-MAP) tool was used to search for potential compounds or drugs that may inhibit ferroptosis-related DEGs. The transcriptional levels of the key genes and potential therapeutic compounds were verified in an LPS-induced mouse model of lung injury. The expression of these key genes was further verified using the GSE60088 and GSE137342 datasets. Results 38 ferroptosis-related DEGs were identified between the septic and control mice. PPI network analysis revealed four modules, the most significant of which included eight ferroptosis-related DEGs. Functional enrichment analysis showed that these genes were enriched in the HIF-1 signaling pathway, including IL-6 (Interleukin-6), TIMP1 (Tissue Inhibitor of Metalloproteinase 1), HIF-1α (Hypoxia-Inducible Factor-1α), and HMOX1 (Heme Oxygenase-1). Phloretin, a natural compound, was identified as a potential inhibitor of these genes. Treatment with phloretin significantly reduced the expression of these genes (p < 0.05), mitigated lung injury, improved inflammatory profiles by approximately 50%, and ferroptosis profiles by nearly 30% in the SLI models. Conclusion This study elucidates the significant role of ferroptosis in SLI and identifies phloretin as a potential therapeutic agent. However, further research, particularly involving human clinical trials, is necessary to validate these findings for clinical use.
Collapse
Affiliation(s)
- Zhile Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Han Gan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Siyuan Li
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yuchen Xue
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kai Luo
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Kai Huang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yunqian Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Hui Zhang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
36
|
Chen Z, Wang W, Hou J, Gao C, Song M, Zhao Z, Guan R, Chen J, Wu H, Abdul Razak SR, Han T, Zhang J, Wang L, Ahmad NH, Li X. NEDD4L contributes to ferroptosis and cell growth inhibition in esophageal squamous cell carcinoma by facilitating xCT ubiquitination. Cell Death Discov 2024; 10:473. [PMID: 39557844 PMCID: PMC11574128 DOI: 10.1038/s41420-024-02243-5] [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: 02/21/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
The oncogene xCT plays an indispensable role in tumor growth by protecting cancer cells from oxidative stress and ferroptosis. Emerging evidence indicated xCT function is tightly controlled by posttranslational modifications, especially ubiquitination. However, it still remains unclear what specific regulatory mechanism of xCT by ubiquitin ligases in human cancers. Here, we reported that NEDD4L, an E3 ubiquitin ligases, inhibited esophageal squamous cell carcinoma (ESCC) tumor growth and facilitated ferroptosis by ubiquitination of xCT. NEDD4L expression was declined in ESCC and was associated with tumor invasion, lymph node metastasis and distant metastasis. Silencing NEDD4L triggered ESCC tumor growth. Meanwhile, knock down of NEDD4L prevented the accumulation of ROS, elevated the level of GSH, reduced the content of MDA in ESCC cells, thereby inhibiting ferroptosis. Mechanistically, NEDD4L directly bound to the ∆CT domain of xCT through its WW and HECT domain. More importantly, NEDD4L promoted xCT degradation by facilitating its polyubiquitination in ESCC cells. Collectively, these findings suggest that NEDD4L is crucial in governing the stability of xCT and mediating ferroptosis in ESCC.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Weilong Wang
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Jinghan Hou
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Can Gao
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Meili Song
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Zijun Zhao
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Ruirui Guan
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Jingsheng Chen
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Huicheng Wu
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Siti Razila Abdul Razak
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
| | - Tao Han
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Junbo Zhang
- Department of Surgery, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Lidong Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, Henan Province, PR China
| | - Nor Hazwani Ahmad
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia.
| | - Xiumin Li
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China.
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| |
Collapse
|
37
|
Wang R, Zhang X, Ye H, Yang X, Zhao Y, Wu L, Liu H, Wen Y, Wang J, Wang Y, Yu M, Ma C, Wang L. Fibroblast growth factor 21 improves diabetic cardiomyopathy by inhibiting ferroptosis via ferritin pathway. Cardiovasc Diabetol 2024; 23:394. [PMID: 39488694 PMCID: PMC11531115 DOI: 10.1186/s12933-024-02469-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/12/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a serious complication in patients with type 2 diabetes mellitus, and its mechanisms are complex and poorly understood. Despite growing evidence suggesting that ferroptosis plays a significant role in cardiovascular disease, it has been less extensively studied in DCM. Fibroblast growth factor 21 (FGF21), whose mechanism of action is closely related to ferroptosis, is widely utilized in studies focused on the prevention and treatment of glucolipid metabolism-related diseases and cardiovascular diseases. OBJECTIVE To confirm the significant role of ferroptosis in DCM and to investigate whether FGF21 improves DCM by inhibiting ferroptosis and elucidating its specific molecular mechanisms. METHODS The animal DCM models were established through high-fat feeding combined with streptozotocin injection in C57BL/6J mice or by db/db mice, and the diabetic cardiomyocyte injury model was created using high glucose and high fat (HG/HF) culture of primary cardiomyocytes. Intervention modeling of FGF21 were performed by injecting adeno-associated virus 9-FGF21 in mice and transfecting FGF21 siRNA or overexpression plasmid in primary cardiomyocytes. RESULTS The findings indicated that ferroptosis was exacerbated and played a significant role in DCM. The overexpression of FGF21 inhibited ferroptosis and improved cardiac injury and function, whereas the knockdown of FGF21 aggravated ferroptosis and cardiac injury and function in DCM. Furthermore, we discovered that FGF21 inhibited ferroptosis in DCM by directly acting on ferritin and prolonging its half-life. Specifically, FGF21 binded to the heavy and light chains of ferritin, thereby reducing its excessive degradation in the proteasome and lysosomal-autophagy pathways in DCM. Additionally, activating transcription factor 4 (ATF4) served as the upstream regulator of FGF21 in DCM. CONCLUSIONS The ATF4-FGF21-ferritin axis mediates the protective effects in DCM through the ferroptosis pathway and represents a potential therapeutic target for DCM.
Collapse
Affiliation(s)
- Ruxin Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaofang Zhang
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Haowen Ye
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Xian Yang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Yongting Zhao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Liangyan Wu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Han Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Yun Wen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Jiaxin Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Ying Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Meixin Yu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Caixia Ma
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Lihong Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China.
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
Wang R, Xu J, Wei S, Liu X. Increased Lipocalin 2 detected by RNA sequencing regulates apoptosis and ferroptosis in COPD. BMC Pulm Med 2024; 24:535. [PMID: 39462322 PMCID: PMC11515215 DOI: 10.1186/s12890-024-03357-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a complex respiratory condition influenced by environmental and genetic factors. Using next-generation sequencing, we aimed to identify dysregulated genes and potential therapeutic targets for COPD. METHODS Peripheral blood leukocyte RNA profiles from COPD patients and healthy controls were analyzed using next-generation sequencing. Key genes involved in COPD pathogenesis were identified through protein-protein interaction network analysis. In vitro, bronchial epithelial cells treated with cigarette smoke extract (CSE) were used to study the effects on gene expression, cell viability, apoptosis, and ferroptosis. Additionally, Lipocalin 2 (LCN2) inhibition experiments were conducted to elucidate its role in COPD-related cellular processes. RESULTS Analysis of RNA profiles revealed consistent downregulation of 17 genes and upregulation of 21 genes across all COPD groups. Among these, Cathelicidin Antimicrobial Peptide(CAMP), Defensin Alpha 4(DEFA4), Neutrophil Elastase(ELANE), LCN2 and Lactotransferrin(LTF) were identified as potentially important players in COPD pathogenesis. Particularly, LCN2 exhibited a close association with COPD and was found to be involved in cellular processes. In vitro experiments demonstrated that CSE treatment significantly increased LCN2 expression in bronchial epithelial cells in a concentration-dependent manner. Moreover, CSE-induced apoptosis and ferroptosis were observed, along with alterations in cell viability, Glutathione content, Fe2 + accumulation, ROS: Reactive Oxygen Species and Malondialdehyde levels, Lactate Dehydrogenase(LDH) release and Glutathione Peroxidase 4(GPX4) expression. Inhibition of LCN2 expression partially reversed these effects, indicating the pivotal role of LCN2 in COPD-related cellular processes. CONCLUSION Our study identified six candidate genes: CAMP, DEFA4, ELANE, LCN2, and LTF were upregulated, HSPA1B was downregulated. Notably, LCN2 emerges as a significant biomarker in COPD pathogenesis, exerting its effects by promoting apoptosis and ferroptosis in bronchial epithelial cells.
Collapse
Affiliation(s)
- Ruiying Wang
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030032, China.
| | - Jianying Xu
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030032, China
| | - Shuang Wei
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030032, China
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiansheng Liu
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030032, China.
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
39
|
Guo Z, Zhang W, Gao H, Li Y, Li X, Yang X, Fan L. High expression levels of haem oxygenase-1 promote ferroptosis in macrophage-derived foam cells and exacerbate plaque instability. Redox Biol 2024; 76:103345. [PMID: 39255694 PMCID: PMC11414708 DOI: 10.1016/j.redox.2024.103345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
Plaque rupture with consequent thrombosis is the leading cause of acute cardiovascular events, during which macrophage death is a hallmark. Ferroptosis is a pivotal intermediate link between early and advanced atherosclerosis. Existing evidence indicates the involvement of macrophage ferroptosis in plaque vulnerability; however, the exact mechanism remains elusive. The aim of this study was to explore key ferroptosis-related genes (FRGs) involved in plaque progression and the underlying molecular mechanisms involved. The expression landscape of FRGs was obtained from atherosclerosis-related GEO datasets. Molecular mechanism studies of ferroptosis were performed using bone marrow-derived macrophages (BMDMs) and macrophage-derived foam cells (MDFCs). Bioinformatics analysis and immunohistochemistry revealed that macrophage haem oxygenase-1 (HMOX1) is the key FRG involved in plaque destabilization. Hypoxic conditions induced a significant increase in Hmox1 expression in MDFCs but not in macrophages. In addition, the beneficial or deleterious effects of Hmox1 were dependent on the degree of Hmox1 induction. Hmox1 overexpression drove inflammatory responses and ferroptotic oxidative stress in MDFCs and aggravated the plaque burden in atherosclerotic model mice. Further mechanistic investigations demonstrated that hypoxia-mediated degradation of egl-9 family hypoxia-inducible factor 3 (Egln3) stabilized Hif1a, which subsequently promoted Hmox1 transcription. Our findings suggest that high Hmox1 expression under hypoxia is deleterious to MDFC viability and plaque stability, providing a reference for the management of acute cardiovascular events.
Collapse
Affiliation(s)
- Zhenyu Guo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wan Zhang
- Department of Vascular Surgery, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Hongxia Gao
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Yang Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xu Li
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Xiaohu Yang
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| | - Longhua Fan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China.
| |
Collapse
|
40
|
Sanz-Alcázar A, Portillo-Carrasquer M, Delaspre F, Pazos-Gil M, Tamarit J, Ros J, Cabiscol E. Deciphering the ferroptosis pathways in dorsal root ganglia of Friedreich ataxia models. The role of LKB1/AMPK, KEAP1, and GSK3β in the impairment of the NRF2 response. Redox Biol 2024; 76:103339. [PMID: 39243573 PMCID: PMC11408871 DOI: 10.1016/j.redox.2024.103339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024] Open
Abstract
Friedreich ataxia (FA) is a rare neurodegenerative disease caused by decreased levels of the mitochondrial protein frataxin. Frataxin has been related in iron homeostasis, energy metabolism, and oxidative stress. Ferroptosis has recently been shown to be involved in FA cellular degeneration; however, its role in dorsal root ganglion (DRG) sensory neurons, the cells that are affected the most and the earliest, is mostly unknown. In this study, we used primary cultures of frataxin-deficient DRG neurons as well as DRG from the FXNI151F mouse model to study ferroptosis and its regulatory pathways. A lack of frataxin induced upregulation of transferrin receptor 1 and decreased ferritin and mitochondrial iron accumulation, a source of oxidative stress. However, there was impaired activation of NRF2, a key transcription factor involved in the antioxidant response pathway. Decreased total and nuclear NRF2 explains the downregulation of both SLC7A11 (a member of the system Xc, which transports cystine required for glutathione synthesis) and glutathione peroxidase 4, responsible for increased lipid peroxidation, the main markers of ferroptosis. Such dysregulation could be due to the increase in KEAP1 and the activation of GSK3β, which promote cytosolic localization and degradation of NRF2. Moreover, there was a deficiency in the LKB1/AMPK pathway, which would also impair NRF2 activity. AMPK acts as a positive regulator of NRF2 and it is activated by the upstream kinase LKB1. The levels of LKB1 were reduced when frataxin decreased, in agreement with reduced pAMPK (Thr172), the active form of AMPK. SIRT1, a known activator of LKB1, was also reduced when frataxin decreased. MT-6378, an AMPK activator, restored NRF2 levels, increased GPX4 levels and reduced lipid peroxidation. In conclusion, this study demonstrated that frataxin deficiency in DRG neurons disrupts iron homeostasis and the intricate regulation of molecular pathways affecting NRF2 activation and the cellular response to oxidative stress, leading to ferroptosis.
Collapse
Affiliation(s)
- Arabela Sanz-Alcázar
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, Catalonia, Spain.
| | | | - Fabien Delaspre
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, Catalonia, Spain.
| | - Maria Pazos-Gil
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, Catalonia, Spain.
| | - Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, Catalonia, Spain.
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, Catalonia, Spain.
| | - Elisa Cabiscol
- Departament de Ciències Mèdiques Bàsiques, IRBLleida, Universitat de Lleida, Catalonia, Spain.
| |
Collapse
|
41
|
Jin B, Zhang Z, Zhang Y, Yang M, Wang C, Xu J, Zhu Y, Mi Y, Jiang J, Sun Z. Ferroptosis and myocardial ischemia-reperfusion: mechanistic insights and new therapeutic perspectives. Front Pharmacol 2024; 15:1482986. [PMID: 39411064 PMCID: PMC11473306 DOI: 10.3389/fphar.2024.1482986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a significant factor in the development of cardiac dysfunction following a myocardial infarction. Ferroptosis, a type of regulated cell death driven by iron and marked by lipid peroxidation, has garnered growing interest for its crucial involvement in the pathogenesis of MIRI.This review comprehensively examines the mechanisms of ferroptosis, focusing on its regulation through iron metabolism, lipid peroxidation, VDAC signaling, and antioxidant system dysregulation. We also compare ferroptosis with other forms of cell death to highlight its distinct characteristics. Furthermore, the involvement of ferroptosis in MIRI is examined with a focus on recent discoveries concerning ROS generation, mitochondrial impairment, autophagic processes, ER stress, and non-coding RNA regulation. Lastly, emerging therapeutic strategies that inhibit ferroptosis to mitigate MIRI are reviewed, providing new insights into potential clinical applications.
Collapse
Affiliation(s)
- Binwei Jin
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhiming Zhang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yang Zhang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Minjun Yang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Cheng Wang
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Cardiology, Taizhou hospital of Zhejiang Province, Shaoxing University, Linhai, China
| | - Jiayi Xu
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Cardiology, Taizhou hospital of Zhejiang Province, Shaoxing University, Linhai, China
| | - Yu Zhu
- Medical Research Center, Taizhou Hospital of Zhejiang Province, Linhai, China
| | - Yafei Mi
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Cardiology, Taizhou hospital of Zhejiang Province, Shaoxing University, Linhai, China
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhenzhu Sun
- Department of Cardiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| |
Collapse
|
42
|
Gawargi FI, Mishra PK. MMP9 drives ferroptosis by regulating GPX4 and iron signaling. iScience 2024; 27:110622. [PMID: 39252956 PMCID: PMC11382059 DOI: 10.1016/j.isci.2024.110622] [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: 12/12/2023] [Revised: 02/08/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
Ferroptosis, defined by the suppression of glutathione peroxidase-4 (GPX4) and iron overload, is a distinctive form of regulated cell death. Our in-depth research identifies matrix metalloproteinase-9 (MMP9) as a critical modulator of ferroptosis through its influence on GPX4 and iron homeostasis. Employing an innovative MMP9 construct without collagenase activity, we reveal that active MMP9 interacts with GPX4 and glutathione reductase, reducing GPX4 expression and activity. Furthermore, MMP9 suppresses key transcription factors (SP1, CREB1, NRF2, FOXO3, and ATF4), alongside GPX1 and ferroptosis suppressor protein-1 (FSP1), thereby disrupting the cellular redox balance. MMP9 regulates iron metabolism by modulating iron import, storage, and export via a network of protein interactions. LC-MS/MS has identified 83 proteins that interact with MMP9 at subcellular levels, implicating them in ferroptosis regulation. Integrated pathway analysis (IPA) highlights MMP9's extensive influence on ferroptosis pathways, underscoring its potential as a therapeutic target in conditions with altered redox homeostasis and iron metabolism.
Collapse
Affiliation(s)
- Flobater I Gawargi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
43
|
Yan X, Liu Q, Wu S, Fan X, Teng Y, Wang N, Zhang J. Tert-butyl hydroperoxide induces trabecular meshwork cells injury through ferroptotic cell death. J Cell Commun Signal 2024; 18:e12050. [PMID: 39524143 PMCID: PMC11544637 DOI: 10.1002/ccs3.12050] [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: 05/13/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 11/16/2024] Open
Abstract
Trabecular meshwork (TM) tissue has a crucial role in regulating aqueous humor circulation in the eye, thus maintaining normal intraocular pressure (IOP). TM dysfunction causes IOP elevation, which leads to glaucoma. To investigate biological changes in TM tissue in patients with glaucoma, we analyzed the mRNA expression microarray dataset, GSE27276. Gene ontology analysis indicated that redox microenvironment imbalance is among the main changes of TM tissue in patients with glaucoma. Subsequently, we induced oxidative stress in TM cells using the tert-butyl hydroperoxide (tBHP) treatment, to generate in vivo and in vitro models, and conducted mRNA sequencing to identify genes with critical roles in maintaining the redox microenvironment balance. We found that the tBHP caused TM dysfunction in vivo, characterized by aqueous humor circulation resistance, IOP elevation, and TM cell death. Further, Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that ferroptosis signaling was enriched in tBHP-treated TM cells. Consistently, in vitro analyses showed that levels of reactive oxygen species, ferric ion, and malondialdehyde were increased after the tBHP treatment, indicating TM cell ferroptosis. Furthermore, inhibiting ferroptosis alleviated tBHP-induced TM cell injury. This study provides new insights suggesting that inhibition of ferroptosis has potential as a treatment for glaucoma.
Collapse
Affiliation(s)
- Xuejing Yan
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical UniversityBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijingChina
- Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Qian Liu
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical UniversityBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijingChina
- Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Shen Wu
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical UniversityBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijingChina
- Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Xiaowei Fan
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical UniversityBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijingChina
| | - Yufei Teng
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical UniversityBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijingChina
| | - Ningli Wang
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical UniversityBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijingChina
- Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Jingxue Zhang
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical UniversityBeijing Ophthalmology & Visual Sciences Key LaboratoryBeijingChina
- Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| |
Collapse
|
44
|
Zhang M, Li J, Hu W. The complex interplay between ferroptosis and atherosclerosis. Biomed Pharmacother 2024; 178:117183. [PMID: 39079265 DOI: 10.1016/j.biopha.2024.117183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024] Open
Abstract
Atherosclerosis, characterized by the accumulation of plaque within the arterial walls, is an intricate cardiovascular disease that often results in severe health issues. Recent studies have emphasized the importance of ferroptosis, a controlled type of cell death dependent on iron, as a critical factor in this disease state. Ferroptosis, distinguished by its reliance on iron and the accumulation of lipid hydroperoxides, offers a unique insight into the pathology of atherosclerotic lesions. This summary encapsulates the current knowledge of the intricate role ferroptosis plays in the onset and progression of atherosclerosis. It explores the molecular processes through which lipid peroxidation and iron metabolism contribute to the development of atheromatous plaques and evaluates the possibility of utilizing ferroptosis as a novel treatment approach for atherosclerosis. By illuminating the intricate relationship between ferroptosis-related processes and atherosclerosis, this review paves the way for future clinical applications and personalized medicine approaches aimed at alleviating the effects of atherosclerosis.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Vascular Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiangping Li
- Department of Oncological Radiotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Hu
- Department of Vascular Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
45
|
Fan J, Zhu T, Tian X, Liu S, Zhang SL. Exploration of ferroptosis and necroptosis-related genes and potential molecular mechanisms in psoriasis and atherosclerosis. Front Immunol 2024; 15:1372303. [PMID: 39072329 PMCID: PMC11272566 DOI: 10.3389/fimmu.2024.1372303] [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: 01/17/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024] Open
Abstract
Objective Ferroptosis and necroptosis are two recently identified forms of non-apoptotic cell death. Their dysregulation plays a critical role in the development and progression of Psoriasis (PsD) and Atherosclerosis (AS). This study explores shared Ferroptosis and necroptosis-related genes and elucidates their molecular mechanisms in PsD and AS through the analysis of public databases. Methods Data sets for PsD (GSE30999) and AS (GSE28829) were retrieved from the GEO database. Differential gene expression (DEG) and weighted gene co-expression network analysis (WGCNA) were performed. Machine learning algorithms identified candidate biomarkers, whose diagnostic values were assessed using Receiver Operating Characteristic (ROC) curve analysis. Additionally, the expression levels of these biomarkers in cell models of AS and PsD were quantitatively measured using Western Blot (WB) and real-time quantitative PCR (RT-qPCR). Furthermore, CIBERSORT evaluated immune cell infiltration in PsD and AS tissues, highlighting the correlation between characteristic genes and immune cells. Predictive analysis for candidate drugs targeting characteristic genes was conducted using the DGIdb database, and an lncRNA-miRNA-mRNA network related to these genes was constructed. Results We identified 44 differentially expressed ferroptosis-related genes (DE-FRGs) and 30 differentially expressed necroptosis-related genes (DE-NRGs). GO and KEGG enrichment analyses revealed significant enrichment of these genes in immune-related and inflammatory pathways, especially in NOD-like receptor and TNF signaling pathways. Two ferroptosis-related genes (NAMPT, ZFP36) and eight necroptosis-related genes (C7, CARD6, CASP1, CTSD, HMOX1, NOD2, PYCARD, TNFRSF21) showed high sensitivity and specificity in ROC curve analysis. These findings were corroborated in external validation datasets and cell models. Immune infiltration analysis revealed increased levels of T cells gamma delta, Macrophages M0, and Macrophages M2 in PsD and AS samples. Additionally, we identified 43 drugs targeting 5 characteristic genes. Notably, the XIST-miR-93-5p-ZFP36/HMOX1 and NEAT1-miR-93-5p-ZFP36/HMOX1 pathways have been identified as promising RNA regulatory pathways in AS and PsD. Conclusion The two ferroptosis-related genes (NAMPT, ZFP36) and eight necroptosis-related genes (C7, CARD6, CASP1, CTSD, HMOX1, NOD2, PYCARD, TNFRSF21) are potential key biomarkers for PsD and AS. These genes significantly influence the pathogenesis of PsD and AS by modulating macrophage activity, participating in immune regulation, and mediating inflammatory responses.
Collapse
Affiliation(s)
- Jilin Fan
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tingting Zhu
- Department of Neurosurgery Ward 5, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiaoling Tian
- Department of Neurosurgery Ward 5, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Sijia Liu
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Shi-Liang Zhang
- Cardiovascular Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
46
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Decoding ferroptosis: Revealing the hidden assassin behind cardiovascular diseases. Biomed Pharmacother 2024; 176:116761. [PMID: 38788596 DOI: 10.1016/j.biopha.2024.116761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
The discovery of regulatory cell death processes has driven innovation in cardiovascular disease (CVD) therapeutic strategies. Over the past decade, ferroptosis, an iron-dependent form of regulated cell death driven by excessive lipid peroxidation, has been shown to drive the development of multiple CVDs. This review provides insights into the evolution of the concept of ferroptosis, the similarities and differences with traditional modes of programmed cell death (e.g., apoptosis, autophagy, and necrosis), as well as the core regulatory mechanisms of ferroptosis (including cystine/glutamate transporter blockade, imbalance of iron metabolism, and lipid peroxidation). In addition, it provides not only a detailed review of the role of ferroptosis and its therapeutic potential in widely studied CVDs such as coronary atherosclerotic heart disease, myocardial infarction, myocardial ischemia/reperfusion injury, heart failure, cardiomyopathy, and aortic aneurysm but also an overview of the phenomenon and therapeutic perspectives of ferroptosis in lesser-addressed CVDs such as cardiac valvulopathy, pulmonary hypertension, and sickle cell disease. This article aims to integrate this knowledge to provide a comprehensive view of ferroptosis in a wide range of CVDs and to drive innovation and progress in therapeutic strategies in this field.
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, 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, 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, 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, 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, China.
| |
Collapse
|
47
|
Ji W, Xie X, Bai G, He Y, Li L, Zhang L, Qiang D. Metabolomic approaches to dissect dysregulated metabolism in the progression of pre-diabetes to T2DM. Mol Omics 2024; 20:333-347. [PMID: 38686662 DOI: 10.1039/d3mo00130j] [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: 05/02/2024]
Abstract
Many individuals with pre-diabetes eventually develop diabetes. Therefore, profiling of prediabetic metabolic disorders may be an effective targeted preventive measure. We aimed to elucidate the metabolic mechanism of progression of pre-diabetes to type 2 diabetes mellitus (T2DM) from a metabolic perspective. Four sets of plasma samples (20 subjects per group) collected according to fasting blood glucose (FBG) concentration were subjected to metabolomic analysis. An integrative approach of metabolome and WGCNA was employed to explore candidate metabolites. Compared with the healthy group (FBG < 5.6 mmol L-1), 113 metabolites were differentially expressed in the early stage of pre-diabetes (5.6 mmol L-1 ⩽ FBG < 6.1 mmol L-1), 237 in the late stage of pre-diabetes (6.1 mmol L-1 ⩽ FBG < 7.0 mmol L-1), and 245 in the T2DM group (FBG ⩾ 7.0 mmol L-1). A total of 27 differentially expressed metabolites (DEMs) were shared in all comparisons. Among them, L-norleucine was downregulated, whereas ethionamide, oxidized glutathione, 5-methylcytosine, and alpha-D-glucopyranoside beta-D-fructofuranosyl were increased with the rising levels of FBG. Surprisingly, 15 (11 lyso-phosphatidylcholines, L-norleucine, oxidized glutathione, arachidonic acid, and 5-oxoproline) of the 27 DEMs were ferroptosis-associated metabolites. WGCNA clustered all metabolites into 8 modules and the pathway enrichment analysis of DEMs showed a significant annotation to the insulin resistance-related pathway. Integrated analysis of DEMs, ROC and WGCNA modules determined 12 potential biomarkers for pre-diabetes and T2DM, including L-norleucine, 8 of which were L-arginine or its metabolites. L-Norleucine and L-arginine could serve as biomarkers for pre-diabetes. The inventory of metabolites provided by our plasma metabolome offers insights into T2DM physiology metabolism.
Collapse
Affiliation(s)
- Wenrui Ji
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Xiaomin Xie
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Guirong Bai
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Yanting He
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Ling Li
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Li Zhang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| | - Dan Qiang
- Department of Endocrinology, The First People's Hospital of Yinchuan, Yinchuan, People's Republic of China.
| |
Collapse
|
48
|
Zhen J, Sheng X, Chen T, Yu H. Histone acetyltransferase Kat2a regulates ferroptosis via enhancing Tfrc and Hmox1 expression in diabetic cardiomyopathy. Cell Death Dis 2024; 15:406. [PMID: 38858351 PMCID: PMC11164963 DOI: 10.1038/s41419-024-06771-x] [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: 11/21/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Diabetic cardiomyopathy (DCM) is a prevalent myocardial microvascular complication of the myocardium with a complex pathogenesis. Investigating the pathogenesis of DCM can significantly contribute to enhancing its prevention and treatment strategies. Our study revealed an upregulation of lysine acetyltransferase 2 A (Kat2a) expression in DCM, accompanied by a decrease in N6-methyladenosine (m6A) modified Kat2a mRNA levels. Our study revealed an upregulation of lysine acetyltransferase 2 A (Kat2a) expression in DCM, accompanied by a decrease in N6-methyladenosine (m6A) modified Kat2a mRNA levels. Functionally, inhibition of Kat2a effectively ameliorated high glucose-induced cardiomyocyte injury both in vitro and in vivo by suppressing ferroptosis. Mechanistically, Demethylase alkB homolog 5 (Alkbh5) was found to reduce m6A methylation levels on Kat2a mRNA, leading to its upregulation. YTH domain family 2 (Ythdf2) played a crucial role as an m6A reader protein mediating the degradation of Kat2a mRNA. Furthermore, Kat2a promoted ferroptosis by increasing Tfrc and Hmox1 expression via enhancing the enrichment of H3K27ac and H3K9ac on their promoter regions. In conclusion, our findings unveil a novel role for the Kat2a-ferroptosis axis in DCM pathogenesis, providing valuable insights for potential clinical interventions.
Collapse
Affiliation(s)
- Juan Zhen
- Department of Cadre Ward, the First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Xia Sheng
- Department of Cadre Ward, the First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Tianlong Chen
- Department of Cardiology, the First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Haitao Yu
- Department of Cardiology, the First Hospital of Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
49
|
Wu D, Zhu P, Shi Z, Li C, Wu C, Sun W, Ran J. Autologous Platelet-Rich Gel Accelerates Diabetic Wound Healing Through Inhibition of Ferritinophagy. INT J LOW EXTR WOUND 2024:15347346241258528. [PMID: 38839257 DOI: 10.1177/15347346241258528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Aims: The objective was to examine the efficacy of autologous platelet-rich gel (APG) in treating diabetic wound and investigate the association between APG and ferritinophagy. Methods: A total of 32 patients with diabetic foot (DF) and Wagner grade 1 to 2 were included. Within the APG group, individuals with DF received weekly APG treatment. In the non-APG group, DF patients received daily dressing changes. Flow cytometry quantified the proportion of endothelial progenitor cells (EPCs) in peripheral blood on days 0 and 10. The diabetic rat model was induced using Streptozotocin. Two circular skin wounds were created on the backs of rats. The normal glucose group received daily dressing changes on the wound. In the diabetic group, the left wound underwent daily dressing changes, whereas the right wound was treated with APG once a week. CD34 levels were tested 7 days after the skin damage. The levels of glutathione peroxidase 4 (GPX4), Nuclear Receptor Coactivator 4 (NCOA4), Light chain 3 (LC3), and Masson staining were quantified on 14 days. The wound area and wound healing rate were separately measured at 0 and 14 days after the injury, regardless of DF patients or diabetic rats. Results: The wound healing rate was higher in the APG group than in the non-APG group, regardless of DF patients or diabetic rats. The APG group had a greater ΔEPCs% in DF patients than the non-APG group. Regarding rat experiment, the APG group exhibited lower levels of NCOA4, and LC3 expressions and a shorter wound healing time. However, the APG group showed higher levels of CD34 expression, GPX4 protein, and collagen fibers than the non-APG group. Conclusions: Autologous platelet-rich gel accelerated the wound healing rate in diabetic populations and rats. Autologous platelet-rich gel promoted EPCs counts, collagen fiber volume, and vessel numbers. Autologous platelet-rich gel decreased LC3 and NCOA4 expression, but increased GPX4 protein expression. The possible mechanism was the inhibition of ferritinophagy.
Collapse
Affiliation(s)
- Daoai Wu
- Department of Endocrinology, Guangzhou Red Cross Hospital Affiliated to Jinan University, Guangzhou, China
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Ping Zhu
- Department of Endocrinology, Guangzhou Red Cross Hospital Affiliated to Jinan University, Guangzhou, China
| | - Zhaoming Shi
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Chen Li
- Department of Blood Transfusion, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Chenchen Wu
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Weihua Sun
- Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jianmin Ran
- Department of Endocrinology, Guangzhou Red Cross Hospital Affiliated to Jinan University, Guangzhou, China
| |
Collapse
|
50
|
Liu Y, Meng Y, Zhu Y, Gu L, Ma A, Liu R, Liu D, Shen S, Zhang S, Xu C, Zhang J, Wang J. Comparative time-dependent proteomics reveal the tolerance of cancer cells to magnetic iron oxide nanoparticles. Regen Biomater 2024; 11:rbae065. [PMID: 38933085 PMCID: PMC11199825 DOI: 10.1093/rb/rbae065] [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: 02/22/2024] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer is one of the most challenging diseases in the world. Recently, iron oxide nanoparticles (IONPs) are emerging materials with rapid development and high application value, and have shown great potential on tumor therapy due to their unique magnetic and biocompatible properties. However, some data hint us that IONPs were toxic to normal cells and vital organs. Thus, more data on biosafety evaluation is urgently needed. In this study, we compared the effects of silicon-coated IONPs (Si-IONPs) on two cell types: the tumor cells (Hela) and the normal cells (HEK293T, as 293 T for short), compared differences of protein composition, allocation and physical characteristics between these two cells. The major findings of our study pointed out that 293 T cells death occurred more significant than that of Hela cells after Si-IONPs treatment, and the rate and content of endocytosis of Si-IONPs in 293 T cells was more prominent than in Hela cells. Our results also showed Si-IONPs significant promoted the production of reactive oxygen species and disturbed pathways related to oxidative stress, iron homeostasis, apoptosis and ferroptosis in both two types of cells, however, Hela cells recovered from these disturbances more easily than 293 T. In conclusion, compared with Hela cells, IONPs are more likely to induce 293 T cells death and Hela cells have their own unique mechanisms to defense invaders, reminding scientists that future in vivo and in vitro studies of nanoparticles need to be cautious, and more safety data are needed for further clinical treatment.
Collapse
Affiliation(s)
- Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liwei Gu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ang Ma
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Rui Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Dandan Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shengnan Shen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shujie Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chengchao Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Department of Nephrology, Shenzhen key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, China
| |
Collapse
|