1
|
Chen S, Fan J, Xie Q, Qin Y, Xie H, Xiao C, Wang W, Liu B. Bufotalin loaded biomimetic nanodrug for combined chemo/photodynamic therapy of cancer. Mater Today Bio 2025; 32:101684. [PMID: 40206143 PMCID: PMC11979420 DOI: 10.1016/j.mtbio.2025.101684] [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: 01/13/2025] [Revised: 03/03/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
The combination of chemotherapy and photodynamic therapy (PDT) for enhancing cancer therapeutic efficiency has attracted tremendous attention recently. However, limitations, such as low local concentration and uncontrollable release of therapeutic agents, reduce combined treatment efficacy. In the present study, we engineered a biomimetic nanodrug employing hollow Prussian blue nanoparticles (HPB NPs) to co-load the chemical agent bufotalin (CS-5) and the photosensitizer chlorin e6 (Ce6) for combined chemo/PDT therapy against cancer. HPB NPs with catalase (CAT)-mimetic activity significantly improved the efficacy of PDT by catalyzing the decomposition of H2O2 into O2, thus alleviating hypoxia, which conversely amplified the efficiency of combination therapy. In vivo assay demonstrated that the encapsulation of a hybrid membrane on the HPB NPs prolonged blood circulation life 3.4-fold compared to free drug. Additionally, this strategy of combinational chemo/PDT therapy exhibits a remarkable cytotoxic effect against gastric cancer (BGC-823) and breast cancer (4T1) through the induction of ferroptosis and pyroptosis while simultaneously activating the immune response, with minimal adverse effects on normal organs. Thus, the co-delivery system based on biomimetic nanocarriers appears to be a promising platform for combined chemo/PDT therapy in tumor treatment.
Collapse
Affiliation(s)
- Simin Chen
- College of Biology, Hunan University, Changsha, 410082, China
| | - Jialong Fan
- College of Biology, Hunan University, Changsha, 410082, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, 410219, China
| | - Qian Xie
- Hunan Provincial Maternal and Child Health Care Hospital, Hunan Province, Changsha 410008, China
| | - Yan Qin
- College of Biology, Hunan University, Changsha, 410082, China
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Hailong Xie
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, 421001, China
| | - Chang Xiao
- College of Biology, Hunan University, Changsha, 410082, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, China
| |
Collapse
|
2
|
Nedaeinia R, Dianat-Moghadam H, Movahednasab M, Khosroabadi Z, Keshavarz M, Amoozgar Z, Salehi R. Therapeutic and prognostic values of ferroptosis signature in glioblastoma. Int Immunopharmacol 2025; 155:114597. [PMID: 40239336 DOI: 10.1016/j.intimp.2025.114597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/15/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025]
Abstract
Ferroptosis is a regulated cell death process that results in decreased tumor growth and aggressiveness when targeted in various cancer cells. Studying the impact of ferroptosis in glioblastoma (GBM) will provide important knowledge about tumor biology and potential treatment strategies. The high metabolic activity resulting in ROS production, iron content and active lipid metabolism of glioblastoma cells make them particularly susceptible to ferroptosis. Single-cell RNA sequencing reveals the molecular signature of GBM and its tumor microenvironment, introducing ferroptosis-related biomarkers pathways and drug resistance mechanisms to enhance treatment outcomes for GBM patients. The relationship between ferroptosis and the immune landscape in GBM is complex and can have either positive or negative effects. These effects can be identified through single-cell RNA sequencing to develop targeted chemo-, radio- and immuno- therapies against glioma stem cells and tumor-supportive immune cells. Additionally, the implication of oncolytic virotherapy in combination with ferroptosis induction can lead to improved treatment of GBM in a clinical setting.
Collapse
Affiliation(s)
- Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hassan Dianat-Moghadam
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maedeh Movahednasab
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Khosroabadi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
3
|
Jawed R, Bhatti H, Khan A. Genetic profile of ferroptosis in non-small cell lung carcinoma and pharmaceutical options for ferroptosis induction. Clin Transl Oncol 2025; 27:1867-1886. [PMID: 39460894 DOI: 10.1007/s12094-024-03754-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths and the second most commonly diagnosed malignancy worldwide. Lung adenocarcinoma (LUAD) and lung squamous cell LC (LUSCC) are the most common subtypes of non-small cell LC (NSCLC). Early diagnosis of LC can be challenging due to a lack of biomarkers. The overall survival (OS) of patients with NSCLC is still poor despite the enormous efforts that have been made to develop novel treatments. Understanding fundamental molecular and genetic mechanisms is necessary to develop new therapeutic approaches for NSCLC. A recently identified type of programmed cell death known as ferroptosis is one potential approach. Ferroptosis causes oxidative damage and the death of cancerous cells by peroxidizing unsaturated phospholipids and accumulating reactive oxygen species (ROS) in an iron-dependent manner. Ferroptosis-related gene (FRG) signatures have recently been evaluated for their ability to predict patient OS and prognosis. These analyses show FRGs are involved in cancer progression, and may serve as promising biomarkers for tumor diagnosis and therapy. Moreover, we summarize the current pharmaceutical options of ferroptosis induction and their underlying molecular mechanism in LC. Therefore, this review aims to provide a comprehensive summary of FRG-based prognostic models, their associated metabolic and signaling pathways, and promising therapeutic options for ferroptosis induction in NSCLC.
Collapse
Affiliation(s)
- Rohil Jawed
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, 210023, China.
| | - Huma Bhatti
- School of Chemistry and Chemical Engineering, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, 210023, China
| | - Adnan Khan
- Clinical and Molecular Labs, Karachi Institute of Radiotherapy and Nuclear Medicine (KIRAN), KDA Scheme 33 Near Safoora Chowk, Karachi, Pakistan
| |
Collapse
|
4
|
Wang C, Zha YL, Wang H, Sun B, Qiang WG, Yuan Y, Shi HB, Hu WW. Carfilzomib promotes Iodine-125 seed radiation-induced apoptosis, paraptosis, and ferroptosis in esophageal squamous cell carcinoma by aggravating endoplasmic reticulum stress. Transl Oncol 2025; 57:102393. [PMID: 40315760 DOI: 10.1016/j.tranon.2025.102393] [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: 09/03/2024] [Revised: 02/09/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025] Open
Abstract
Iodine-125 (125I) seed brachytherapy has been applied to treat various malignant tumors such as esophageal cancer, however, radioresistance can reduce its efficacy. Endoplasmic reticulum stress (ERS) and subsequent unfolded protein response (UPR) is one of the core mechanisms of 125I seed radiation-induced cell death, thus aggravating ERS has been considered a promising sensitization strategy. Herein, we show that combination therapy of an irreversible proteasome inhibitor carfilzomib (CFZ) and 125I seed radiation displayed strong anti-tumor effect on esophageal squamous cell carcinoma (ESCC). Mechanistically, ERS and UPR regulated multiple cell death modalities induced by the combination therapy, including apoptosis, paraptosis, and ferroptosis. 125I seed radiation induced reactive oxygen species (ROS) production, DNA damage, p53 activation, and apoptosis. CFZ promoted ROS production, and augmented 125I seed radiation-induced apoptosis via the mitochondrial pathway, which was mediated by the UPR-C/EBP homologous protein (CHOP) pathway and was independent of the p53 pathway. CFZ enhanced 125I seed radiation-induced intracellular Ca2+ overload, protein ubiquitination, ERS, and UPR, consequently promoting paraptosis. 125I seed radiation induced accumulation of intracellular Fe2+ and lipid peroxides but upregulated the expression of ferroptosis inhibitors, SLC7A11 and glutathione peroxidase 4 (GPX4). The combination therapy promoted ferroptosis by enhancing the accumulation of intracellular Fe2+ and downregulating GPX4 expression. The mouse experiment demonstrated that CFZ can promote the efficacy of 125I seed radiation with good tolerance. Our findings suggest that combination therapy of 125I seed radiation and CFZ is associated with multiple cell death modalities and may serve as a promising therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Chao Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Yin-Lin Zha
- Department of Radiation Oncology, the Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Hao Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Bai Sun
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Wei-Guang Qiang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Ye Yuan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China
| | - Hong-Bing Shi
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China.
| | - Wen-Wei Hu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, PR China.
| |
Collapse
|
5
|
Jiang P, Jiang Z, Li S, Li YX, Chen Y, Li X. The suppressive role of GLS in radiosensitivity and irradiation-induced immune response in LUAD: integrating bioinformatics and experimental insights. Front Immunol 2025; 16:1582587. [PMID: 40308578 PMCID: PMC12040943 DOI: 10.3389/fimmu.2025.1582587] [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: 02/24/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Background Radiotherapy elicits immune activation, thereby synergistically enhancing systemic tumor control when combined with immunotherapy. Glutaminase (GLS), a key enzyme for glutamine metabolism, has been found to regulate glutamine availability within tumor microenvironment (TME). However, the precise mechanisms through which GLS modulates radiosensitivity and irradiation-induced immune responses in lung adenocarcinoma (LUAD) and its clinical value remain to be fully elucidated. Methods We employed bulk RNA-seq and single-cell transcriptomics to explore the role of GLS expression in radiosensitivity and immune infiltration. The bioinformatic results were validated by in vitro and in vivo experiments. Co-culture assays and flow cytometry were used to validate the impact of GLS expression on CD8+ T cell activation and cytotoxicity. Moreover, a GLS-DSBr (double strand break repair) prognostic model was developed using machine learning with data from 2,066 LUAD patients. Results In vitro and in vivo experiments demonstrated that GLS silence inhibited DSB repair and promoted ferroptosis, therefore enhancing radiosensitivity. Single-cell and spatial transcriptomics revealed the immunomodulatory effects of GLS expression in the TME. Further, Co-culture assays and flow cytometry experiments indicated that silencing GLS in LUAD cells potentiated the activation and cytotoxicity of CD8+ T cells in the context of radiotherapy. The GLS-DSBr model demonstrated robust predictive performance for overall survival, as well as the efficacy of radiotherapy and immunotherapy in LUAD. The applicability of GLS-DSBr model was further validated through pan-cancer analysis. Conclusion In the contexts of radiotherapy, GLS downregulation exerts dual regulatory effects by modulating ferroptosis and remodeling the immune landscapes, particularly enhancing CD8+ T cell cytotoxicity. Our work suggests that strategies preferentially targeting GLS in tumor cells may represent promising and translatable therapeutic approaches to promote antitumor efficacy of radiotherapy plus immune checkpoint blockade in LUAD patients. Furthermore, the established GLS-DSBr model serves as a robust predictive tool for prognosis and effects of radiotherapy and immunotherapy, which assists personalized treatment optimization in LUAD.
Collapse
Affiliation(s)
- Peicheng Jiang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhifeng Jiang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Su Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
- Department of Cardiology, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Ye-Xiong Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuqiong Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Xinyan Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Matesanz-Sánchez R, Peitzsch M, Lange I, Mircetic J, Seifert M, Cordes N, Vehlow A. A novel role of exostosin glycosyltransferase 2 (EXT2) in glioblastoma cell metabolism, radiosensitivity and ferroptosis. Cell Death Differ 2025:10.1038/s41418-025-01503-w. [PMID: 40234611 DOI: 10.1038/s41418-025-01503-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/25/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
Glioblastoma (GBM) employs various strategies to resist therapy, resulting in poor patient survival. A key aspect of its survival mechanisms lies in metabolic regulation, maintaining rapid growth and evading cell death. Recent studies revealed the connection between therapy resistance and ferroptosis, a lipid peroxidation-dependent cell death mechanism triggered by metabolic dysfunction. Our aim was to identify novel regulators of therapy resistance in GBM cells. We conducted a comprehensive analysis combining RNA-sequencing data from a panel of human GBM cell models and TCGA GBM patient datasets. We focused on the top-12 differentially expressed gene candidates associated with poor survival in GBM patients and performed an RNA interference-mediated screen to uncover the radiochemosensitizing potential of these molecules and their impact on metabolic activity, DNA damage, autophagy, and apoptosis. We identified exostosin glycosyltransferase 2 (EXT2), an enzyme previously described in heparan sulfate biosynthesis, as the most promising candidate. EXT2 depletion elicited reduced cell viability and proliferation as well as radiochemosensitization in various GBM cell models. Mechanistically, we explored EXT2 function by conducting untargeted and targeted metabolomics and detected that EXT2-depleted GBM cells exhibit a differential abundance of metabolites belonging to S-adenosylmethionine (SAM) metabolism. Considering these metabolic changes, we determined lipid peroxidation and found that the diminished antioxidant capacity resulting from decreased levels of metabolites in the transsulfuration pathway induces ferroptosis. Moreover, modifications of specific SAM and transsulfuration metabolism associated enzymes revealed a prosurvival and ferroptosis-reducing function when EXT2 is depleted. Collectively, our results uncover a novel role of EXT2 in GBM cell survival and response to X-ray radiation, which is controlled by modulation of ferroptosis. These findings expand our understanding of how GBM cells respond to radio(chemo)therapy and may contribute to the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Rocío Matesanz-Sánchez
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität, Dresden, 01307, Germany
| | - Inga Lange
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, 01328, Germany
| | - Jovan Mircetic
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, 69192, Germany
| | - Michael Seifert
- Institute for Medical Informatics and Biometry (IMB), Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Nils Cordes
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany.
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, 01328, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, 69192, Germany.
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany.
| | - Anne Vehlow
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany.
| |
Collapse
|
7
|
Zhang X, Zhou Y, Li H, Chen M, Peng F, Li N. Integration of Transcriptomic and Single-Cell Data to Uncover Senescence- and Ferroptosis-Associated Biomarkers in Sepsis. Biomedicines 2025; 13:942. [PMID: 40299574 PMCID: PMC12025025 DOI: 10.3390/biomedicines13040942] [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: 03/07/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025] Open
Abstract
Background: Sepsis is a life-threatening condition characterized by organ dysfunction due to an imbalanced immune response to infection, with high mortality. Ferroptosis, an iron-dependent cell death process, and cellular senescence, which exacerbates inflammation, have recently been implicated in sepsis pathophysiology. Methods: Weighted gene co-expression network analysis (WGCNA) was used to identify ferroptosis- and senescence-related gene modules in sepsis. Differentially expressed genes (DEGs) were analyzed using public datasets (GSE57065, GSE65682, and GSE26378). Receiver operating characteristic (ROC) analysis was performed to evaluate their diagnostic potential, while single-cell RNA sequencing (scRNA-seq) was used to assess their immune-cell-specific expression. Molecular docking was conducted to predict drug interactions with key proteins. Results: Five key genes (CD82, MAPK14, NEDD4, TXN, and WIPI1) were significantly upregulated in sepsis patients and highly correlated with immune cell infiltration. MAPK14 and TXN exhibited strong diagnostic potential (AUC = 0.983, 0.978). Molecular docking suggested potential therapeutic interactions with diclofenac, flurbiprofen, and N-acetyl-L-cysteine. Conclusions: This study highlights ferroptosis and senescence as critical mechanisms in sepsis and identifies promising biomarkers for diagnosis and targeted therapy. Future studies should focus on clinical validation and precision medicine applications.
Collapse
Affiliation(s)
- Xiangqian Zhang
- Department of Blood Transfusion, Clinical Transfusion Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yiran Zhou
- Department of Blood Transfusion, Clinical Transfusion Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Hang Li
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, 07743 Jena, Germany
| | - Mengru Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Fang Peng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ning Li
- Department of Blood Transfusion, Clinical Transfusion Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
8
|
Chen X, Cui H, Qin L, Liu R, Fang F, Wang Z. Soybean Lecithin-Gallic Acid Complex Sensitizes Lung Cancer Cells to Radiation Through Ferroptosis Regulated by Nrf2/SLC7A11/GPX4 Pathway. Nutrients 2025; 17:1262. [PMID: 40219018 PMCID: PMC11990552 DOI: 10.3390/nu17071262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Radioresistance remains a significant obstacle in lung cancer radiotherapy, necessitating novel strategies to enhance therapeutic efficacy. This study investigated the radiosensitizing potential of a soybean lecithin-gallic acid complex (SL-GAC) in non-small cell lung cancer (NSCLC) cells and explored its underlying ferroptosis-related mechanisms. SL-GAC was synthesized to improve the bioavailability of gallic acid (GA), a polyphenol with anticancer properties. Methods: NSCLC cell lines (A549 and H1299) and normal bronchial epithelial cells (BEAS-2B) were treated with SL-GAC, ionizing radiation (IR), or their combination. Through a series of in vitro experiments, including cell viability assays, scratch healing assays, flow cytometry, and Western blot analysis, we comprehensively evaluated the effects of SL-GAC on NSCLC cell proliferation, migration, oxidative stress, and ferroptosis induction. Results: SL-GAC combined with IR synergistically suppressed NSCLC cell proliferation and migration, exacerbated oxidative stress via elevated ROS and malondialdehyde levels, and induced mitochondrial dysfunction marked by reduced membrane potential and structural damage, whereas no significant ROS elevation was observed in BEAS-2B cells. Mechanistically, the combination triggered ferroptosis in NSCLC cells, evidenced by iron accumulation and downregulation of Nrf2, SLC7A11, and GPX4, alongside upregulated ACSL4. Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, reversed these effects and restored radiosensitivity. Conclusions: Our findings demonstrate that SL-GAC enhances NSCLC radiosensitivity by promoting ferroptosis via the Nrf2/SLC7A11/GPX4 axis, highlighting its potential as a natural radiosensitizer for clinical translation.
Collapse
Affiliation(s)
- Xingyang Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun 130021, China
| | - Hongli Cui
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Lijing Qin
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Rongrong Liu
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Fang Fang
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun 130021, China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| |
Collapse
|
9
|
Zheng J, Conrad M. Ferroptosis: when metabolism meets cell death. Physiol Rev 2025; 105:651-706. [PMID: 39661331 DOI: 10.1152/physrev.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
We present here a comprehensive update on recent advancements in the field of ferroptosis, with a particular emphasis on its metabolic underpinnings and physiological impacts. After briefly introducing landmark studies that have helped to shape the concept of ferroptosis as a distinct form of cell death, we critically evaluate the key metabolic determinants involved in its regulation. These include the metabolism of essential trace elements such as selenium and iron; amino acids such as cyst(e)ine, methionine, glutamine/glutamate, and tryptophan; and carbohydrates, covering glycolysis, the citric acid cycle, the electron transport chain, and the pentose phosphate pathway. We also delve into the mevalonate pathway and subsequent cholesterol biosynthesis, including intermediate metabolites like dimethylallyl pyrophosphate, squalene, coenzyme Q (CoQ), vitamin K, and 7-dehydrocholesterol, as well as fatty acid and phospholipid metabolism, including the biosynthesis and remodeling of ester and ether phospholipids and lipid peroxidation. Next, we highlight major ferroptosis surveillance systems, specifically the cyst(e)ine/glutathione/glutathione peroxidase 4 axis, the NAD(P)H/ferroptosis suppressor protein 1/CoQ/vitamin K system, and the guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin/dihydrofolate reductase axis. We also discuss other potential anti- and proferroptotic systems, including glutathione S-transferase P1, peroxiredoxin 6, dihydroorotate dehydrogenase, glycerol-3-phosphate dehydrogenase 2, vitamin K epoxide reductase complex subunit 1 like 1, nitric oxide, and acyl-CoA synthetase long-chain family member 4. Finally, we explore ferroptosis's physiological roles in aging, tumor suppression, and infection control, its pathological implications in tissue ischemia-reperfusion injury and neurodegeneration, and its potential therapeutic applications in cancer treatment. Existing drugs and compounds that may regulate ferroptosis in vivo are enumerated.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, Garching, Germany
| |
Collapse
|
10
|
Fang Y, Chen H, Liu Y, Jiang K, Qian Y, Wei J, Fu D, Yang H, Dai S, Jin T, Bu T, Ding K. NUPR1 Promotes Radioresistance in Colorectal Cancer Cells by Inhibiting Ferroptosis. J Cell Mol Med 2025; 29:e70519. [PMID: 40176685 PMCID: PMC11965884 DOI: 10.1111/jcmm.70519] [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: 05/06/2024] [Revised: 01/20/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
Radioresistance is a major clinical challenge and the underlying mechanism has not been thoroughly elucidated. In this study, a radioresistant (RR) cell line is established to explore the transcriptomic signatures of radioresistance in colorectal cancer (CRC). KEGG enriched pathway analysis demonstrated that ferroptosis is inactivated in RR cells. Further detection confirmed that radiotherapy can promote ferroptosis, and ferroptosis inactivation is one of the hallmarks of radioresistance in CRC. What's more, induction of ferroptosis can restore the radiosensitivity of CRC cells. Then, we performed RNA sequencing to compare gene expression between parental and RR cells, and cells pretreated with or without RSL3. Via high-throughput screening, NUPR1 was identified as a potential candidate for ferroptosis-mediated radioresistance in CRC. CRC cells can acquire radiation resistance by NUPR1-mediated ferroptosis suppression in the NUPR1-overexpressing cell line. More importantly, ZZW-115, an NUPR1 inhibitor, can sensitise RR cells to radiotherapy. Overall, our findings identify ferroptosis inactivation linked with resistance to radiotherapy. Besides, NUPR1 can promote radiation resistance by inhibiting ferroptosis, and targeting NUPR1 may be a potential strategy to relieve radioresistance associated with ferroptosis in CRC.
Collapse
Affiliation(s)
- Yimin Fang
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Haiyan Chen
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Department of Radiation Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Yunhua Liu
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Kai Jiang
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Yucheng Qian
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Jingsun Wei
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Dongliang Fu
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Hang Yang
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Siqi Dai
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Tian Jin
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Tongtong Bu
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| |
Collapse
|
11
|
Li M, Jin S, Ma H, Yang X, Zhang Z. Reciprocal regulation between ferroptosis and STING-type I interferon pathway suppresses head and neck squamous cell carcinoma growth through dendritic cell maturation. Oncogene 2025:10.1038/s41388-025-03368-2. [PMID: 40164871 DOI: 10.1038/s41388-025-03368-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 02/23/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) presents a serious clinical challenge mainly due to its resistance to conventional therapies and its complex, immunosuppressive tumor microenvironment. While recent studies have identified ferroptosis as a new therapeutic option, its impact on the immune microenvironment in HNSCC remains controversial, which may hinder its translational application. Although the role of the stimulator of interferon genes (STING)-type I interferon (IFN-I) pathway in antitumor immune responses has been widely investigated, its relationship with ferroptosis in HNSCC has not been fully explored. In this study, we discovered that ferroptosis in HNSCC inhibited tumor growth, activated STING-IFN-I pathway and subsequently improved recruitment and maturation of dendritic cells. We further demonstrated that IFN-I could enhance ferroptosis by inhibiting xCT-glutathione peroxidase 4 (GPX4) antioxidant system. To harness this positive feedback loop, we treated HNSCC tumors with both ferroptosis inducer and STING agonist, resulting in significant tumor suppression, elevated ferroptosis levels and enhanced dendritic cell infiltration. Overall, our findings reveal a mutually regulatory relationship between ferroptosis and the intrinsic STING-IFN-I pathway, providing novel insights into immune-mediated tumor suppression and suggesting its potential as therapeutic approach in HNSCC.
Collapse
Affiliation(s)
- Mingyu Li
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China
| | - Shufang Jin
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Hailong Ma
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Xi Yang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| | - Zhiyuan Zhang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology. No. 639, Zhizaoju Rd, Shanghai, 200011, China.
- Shanghai Research Institute of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639, Zhizaoju Rd, Shanghai, 200011, China.
| |
Collapse
|
12
|
Kim JH, Rho JR, Choi JH. Marine Sponge-Derived Gukulenin A Sensitizes Ovarian Cancer Cells to PARP Inhibition via Ferroptosis Induction. Mar Drugs 2025; 23:138. [PMID: 40278259 PMCID: PMC12028354 DOI: 10.3390/md23040138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Resistance to PARP inhibitors (PARPi), such as olaparib (OLA), is a major challenge in ovarian cancer treatment. In this study, we investigated the combination effect of PARPi and gukulenin A (GUA), a bis-tropolone tetraterpenoid isolated from the marine sponge Phorbas gukhulensis. We found that GUA at a mildly cytotoxic dose synergistically enhanced OLA-induced cytotoxicity in human ovarian cancer cells. The combination treatment significantly increased reactive oxygen species (ROS) levels and lipid peroxidation, leading to ferroptotic rather than apoptotic cell death. Network pharmacology and gene ontology (GO) enrichment analyses revealed oxidative stress-related pathways as key mediators of this effect. Inhibition of NADPH oxidase (NOX) reversed combination-induced cell death, while ferrostatin-1 (FER-1), a ferroptosis inhibitor, significantly reduced lipid peroxidation and cytotoxicity. Additionally, GUA and OLA treatment suppressed ERK1/2 activation, and ERK overexpression attenuated the combination-induced cell death. Collectively, these findings suggest that marine-derived GUA enhances PARPi efficacy in ovarian cancer cells by inducing ferroptosis through oxidative stress and ERK pathway modulation.
Collapse
Affiliation(s)
- Jin-Hyung Kim
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Jung-Rae Rho
- Department of Oceanography, Kunsan National University, Jeonbuk 54150, Republic of Korea;
| | - Jung-Hye Choi
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Integrated Drug Development and Natural Products, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
13
|
Cao Y, Li J, Chen Y, Wang Y, Liu Z, Huang L, Liu B, Feng Y, Yao S, Zhou L, Yin Y, Huang Z. Monounsaturated fatty acids promote cancer radioresistance by inhibiting ferroptosis through ACSL3. Cell Death Dis 2025; 16:184. [PMID: 40102409 PMCID: PMC11920413 DOI: 10.1038/s41419-025-07516-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Abstract
Radioresistance is a major challenge in tumor radiotherapy and involves in a mixture of cellular events, including ferroptosis, a new type of programmed cell death characterized by the excess accumulation of iron-dependent lipid peroxides. In the present study, we observed that surviving cancer tissues and cells after radiotherapy had significantly greater glutathione to oxidized glutathione (GSH/GSSG) ratios and lower lipid reactive oxygen species (ROS) and malondialdehyde (MDA) levels than nonirradiated tumors and cells. Untargeted lipidomic analyses revealed that oleic acid (OA) and palmitoleic acid (POA) were the most significantly upregulated unsaturated fatty acids in irradiated surviving cancer cells compared with those in control cancer cells irradiated with IR. Both OA and POA could protect cancer cells from the killing effects of the ferroptosis inducer erastin and RSL3, and OA had a stronger protective effect than POA, resulting in lower lipid ROS production than POA. Mechanistically, OA protected cells from ferroptosis caused by the accumulation of polyunsaturated fatty acid-containing phospholipids in an ACSL3-dependent manner. A mouse model demonstrated that ACSL3 knockdown combined with imidazole ketone erastin synergistically enhanced antitumor effects in radiation-resistant tumors in vivo. Our study reveals previously undiscovered associations between radiation and fatty acid metabolism and ferroptosis, providing a novel treatment strategy for overcoming cancer radioresistance.
Collapse
Affiliation(s)
- Yulin Cao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Jiuming Li
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Ying Chen
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuanben Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhiang Liu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Liuying Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Bingxin Liu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuyang Feng
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
| | - Surui Yao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Leyuan Zhou
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yuan Yin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China.
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214062, China.
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
14
|
Luo Y, He Y, Xu S, Chen Y, Qin F, Hu W. Ferroptosis: a potential target for non-surgical treatment of laryngeal cancer. Eur Arch Otorhinolaryngol 2025:10.1007/s00405-025-09279-y. [PMID: 40087171 DOI: 10.1007/s00405-025-09279-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/17/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Laryngeal cancer (LC) is among the most prevalent tumors of the respiratory tract. In recent years, the implementation of non-surgical treatments like radiotherapy and chemotherapy has significantly enhanced the therapeutic outcomes for LC. Nevertheless, the underlying therapeutic mechanisms remain unclear, posing a hindrance to the progression of subsequent treatment strategies. OBJECTIVES To explore the potential mechanisms from existing effective treatments for LC and identify relevant targets, thereby providing guidance for subsequent therapeutic research on LC. METHODS This study focuses on ferroptosis, a common type of non-apoptotic cell death that is closely linked to various malignancies. It examines the relationship between ferroptosis and LC by analyzing how regulating ferroptosis-related targets in LC cells can influence the development of the cancer. RESULTS There is a strong association between ferroptosis and LC. Regulating the targets related to ferroptosis in LC cells can effectively counteract the progression of LC. CONCLUSIONS Taking ferroptosis as an entry point, analyzing its potential mechanism in inhibiting LC can provide a direction for the treatment of laryngeal cancer, which may contribute to the improvement of therapeutic strategies for this disease.
Collapse
Affiliation(s)
- Yang Luo
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yuzhu He
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Shuang Xu
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yuxiang Chen
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Fengfeng Qin
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wenjian Hu
- Department of Otolaryngology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University of Luzhou, NO. 182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
15
|
Tian K, Wu P, Gao S, Xu C, Xu W, Jia Z, Wang Y, Sheng L, Zhou X, Wu S, Wu L. A Deferoxamine-Loaded Microneedle Patch Enhances Healing of Radiation-Induced Skin Injury: Potential Involvement of Ferroptosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:15035-15049. [PMID: 40025663 DOI: 10.1021/acsami.4c21589] [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: 03/04/2025]
Abstract
Radiation-induced skin injury (RSI) presents a significant challenge in wound care due to its complex pathophysiology, which includes increased oxidative stress, impaired angiogenesis, and delayed re-epithelialization. Transcriptomic analysis reveals significant alterations in genes associated with the ferroptosis pathway following radiation exposure. In this study, we introduce microneedles composed of silk fibroin hydrogel loaded with deferoxamine (SF+MNs+DFO) to inhibit ferroptosis. SF+MNs+DFO exhibits optimal mechanical properties and drug release kinetics. Histopathological analysis shows reduced inflammation, oxidative stress, and collagen deposition in RSI treated with SF+MNs+DFO, leading to accelerated tissue regeneration and decreased scarring. Molecular biology studies indicate that SF+MNs+DFO inhibits ferroptosis by reducing the concentration of free Fe2+ in the body, thereby decreasing the generation of reactive oxygen species (ROS) and lipid peroxidation. Immunofluorescence studies further confirm the increased neovascularization and reduced fibrosis in SF+MNs+DFO-treated RSI, indicating enhanced tissue repair. SF+MNs+DFO not only inhibits ferroptosis but also promotes angiogenesis and tissue regeneration, offering a promising therapeutic strategy for RSI. In conclusion, DFO-loaded SF hydrogel microneedles provide precise drug delivery, iron chelation, and improved wound healing, demonstrating an effective approach for treating RSI.
Collapse
Affiliation(s)
- Kai Tian
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu China
| | - Pingfan Wu
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu China
| | - Suyue Gao
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu China
| | - Changzhi Xu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wushuang Xu
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu China
| | - Zou Jia
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu China
| | - Yifan Wang
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu China
| | - Lei Sheng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Shuwang Wu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lijun Wu
- Department of Plastic and Aesthetic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu China
| |
Collapse
|
16
|
Scattolin T, Cavarzerani E, Alessi D, Mauceri M, Botter E, Tonon G, Caligiuri I, Repetto O, Kamensek U, Brezar SK, Dalla Pozza M, Palazzolo S, Cemazar M, Canzonieri V, Demitri N, Nolan SP, Gasser G, Visentin F, Rizzolio F. Unlocking the potential of organopalladium complexes for high-grade serous ovarian cancer therapy. Dalton Trans 2025; 54:4685-4696. [PMID: 39967474 DOI: 10.1039/d5dt00194c] [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: 02/20/2025]
Abstract
High-Grade Serous Ovarian Cancer (HGSOC) is the most common and lethal subtype of ovarian cancer, known for its high aggressiveness and extensive genomic alterations. Typically diagnosed at an advanced stage, HGSOC presents formidable challenges in drug therapy. The limited efficacy of standard treatments, development of chemoresistance, scarcity of targeted therapies, and significant tumor heterogeneity render this disease incurable with current treatment options, highlighting the urgent need for novel therapeutic approaches to improve patient outcomes. In this study we report a straightforward and stereoselective synthetic route to novel Pd(II)-vinyl and -butadienyl complexes bearing a wide range of monodentate and bidentate ligands. Most of the synthesized complexes exhibited good to excellent in vitro anticancer activity against ovarian cancer cells. Particularly promising is the water-soluble complex bearing two PTA (1,3,5-triaza-7-phosphaadamantane) ligands and the Pd(II)-butadienyl fragment. This compound combines excellent cytotoxicity towards cancer cells with substantial inactivity towards non-cancerous ones. This derivative was selected for further studies on ex vivo tumor organoids and in vivo mouse models, which demonstrate its remarkable efficacy with surprisingly low collateral toxicity even at high dosages. Moreover, this class of compounds appears to operate through a ferroptotic mechanism, thus representing the first such example for an organopalladium compound.
Collapse
Affiliation(s)
- Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Enrico Cavarzerani
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Dario Alessi
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Matteo Mauceri
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Eleonora Botter
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Giovanni Tonon
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Ombretta Repetto
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Simona Kranjc Brezar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Maria Dalla Pozza
- ChimieParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, Università degli Studi di Trieste, Strada di Fiume 447, Trieste, Italy
| | - Nicola Demitri
- Area Science Park, Elettra-Sincrotrone Trieste, S.S. 14 Km 163.5, Basovizza, 34149, Trieste, Italy
| | - Steven P Nolan
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281, S-3, 9000 Ghent, Belgium
| | - Gilles Gasser
- ChimieParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Fabiano Visentin
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Flavio Rizzolio
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| |
Collapse
|
17
|
Ma X, He C, Wang Y, Cao X, Jin Z, Ge Y, Cao Z, An M, Hao L. Mechanisms and Applications of Manganese-Based Nanomaterials in Tumor Diagnosis and Therapy. Biomater Res 2025; 29:0158. [PMID: 40026879 PMCID: PMC11868662 DOI: 10.34133/bmr.0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/30/2025] [Accepted: 02/09/2025] [Indexed: 03/05/2025] Open
Abstract
Tumors are the second most common cause of mortality globally, ranking just below heart disease. With continuous advances in diagnostic technology and treatment approaches, the survival rates of some cancers have increased. Nevertheless, due to the complexity of the mechanisms underlying tumors, cancer remains a serious public health issue that threatens the health of the population globally. Manganese (Mn) is an essential trace element for the human body. Its regulatory role in tumor biology has received much attention in recent years. Developments in nanotechnology have led to the emergence of Mn-based nanoparticles that have great potential for use in the diagnosis and treatment of cancers. Mn-based nanomaterials can be integrated with conventional techniques, including chemotherapy, radiation therapy, and gene therapy, to augment their therapeutic effectiveness. Further, Mn-based nanomaterials can play a synergistic role in emerging treatment strategies for tumors, such as immunotherapy, photothermal and photodynamic therapy, electromagnetic hyperthermia, sonodynamic therapy, chemodynamic therapy, and intervention therapy. Moreover, Mn-based nanomaterials can enhance both the precision of tumor diagnostics and the capability for combined diagnosis and treatment. This article examines the roles and associated mechanisms of Mn in the field of physiology and tumor biology, with a focus on the application prospects of Mn-based nanomaterials in tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Xiaowen Ma
- Department of Chemistry, School of Forensic Medicine,
China Medical University, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang 110122, China
- China Medical University Center of Forensic Investigation, Shenyang 110122, China
- First Department of Clinical Medicine,
China Medical University, Shenyang 110122, China
| | - Chuan He
- Department of Laboratory Medicine,
the First Hospital of China Medical University, Shenyang 110001, China
| | - Yang Wang
- Department of Chemistry, School of Forensic Medicine,
China Medical University, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang 110122, China
- China Medical University Center of Forensic Investigation, Shenyang 110122, China
| | - Xingrui Cao
- Department of Chemistry, School of Forensic Medicine,
China Medical University, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang 110122, China
- China Medical University Center of Forensic Investigation, Shenyang 110122, China
| | - Zikai Jin
- First Department of Clinical Medicine,
China Medical University, Shenyang 110122, China
| | - Yi Ge
- School of Pharmacy,
Queen’s University Belfast, Belfast BT9 7BL, UK
| | - Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine,
China Medical University, Shenyang 110122, China
| | - Mingxin An
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education,
China Medical University, Shenyang 110122, China
| | - Liang Hao
- Department of Chemistry, School of Forensic Medicine,
China Medical University, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang 110122, China
- China Medical University Center of Forensic Investigation, Shenyang 110122, China
| |
Collapse
|
18
|
Xu W, Guan G, Yue R, Dong Z, Lei L, Kang H, Song G. Chemical Design of Magnetic Nanomaterials for Imaging and Ferroptosis-Based Cancer Therapy. Chem Rev 2025; 125:1897-1961. [PMID: 39951340 DOI: 10.1021/acs.chemrev.4c00546] [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: 02/27/2025]
Abstract
Ferroptosis, an iron-dependent form of regulatory cell death, has garnered significant interest as a therapeutic target in cancer treatment due to its distinct characteristics, including lipid peroxide generation and redox imbalance. However, its clinical application in oncology is currently limited by issues such as suboptimal efficacy and potential off-target effects. The advent of nanotechnology has provided a new way for overcoming these challenges through the development of activatable magnetic nanoparticles (MNPs). These innovative MNPs are designed to improve the specificity and efficacy of ferroptosis induction. This Review delves into the chemical and biological principles guiding the design of MNPs for ferroptosis-based cancer therapies and imaging-guided therapies. It discusses the regulatory mechanisms and biological attributes of ferroptosis, the chemical composition of MNPs, their mechanism of action as ferroptosis inducers, and their integration with advanced imaging techniques for therapeutic monitoring. Additionally, we examine the convergence of ferroptosis with other therapeutic strategies, including chemodynamic therapy, photothermal therapy, photodynamic therapy, sonodynamic therapy, and immunotherapy, within the context of nanomedicine strategies utilizing MNPs. This Review highlights the potential of these multifunctional MNPs to surpass the limitations of conventional treatments, envisioning a future of drug-resistance-free, precision diagnostics and ferroptosis-based therapies for treating recalcitrant cancers.
Collapse
Affiliation(s)
- Wei Xu
- School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Guoqiang Guan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Renye Yue
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, PR China
| | - Zhe Dong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Lingling Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Heemin Kang
- Department of Materials Science and Engineering and College of Medicine, Korea University, 12 Seoul 02841, Republic of Korea
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| |
Collapse
|
19
|
Sant’Angelo D, Descamps G, Lecomte V, Stanicki D, Penninckx S, Dragan T, Van Gestel D, Laurent S, Journe F. Therapeutic Approaches with Iron Oxide Nanoparticles to Induce Ferroptosis and Overcome Radioresistance in Cancers. Pharmaceuticals (Basel) 2025; 18:325. [PMID: 40143107 PMCID: PMC11945075 DOI: 10.3390/ph18030325] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
The emergence of nanotechnology in medicine, particularly using iron oxide nanoparticles (IONPs), may impact cancer treatment strategies. IONPs exhibit unique properties, such as superparamagnetism, biocompatibility, and ease of surface modification, making them ideal candidates for imaging, and therapeutic interventions. Their application in targeted drug delivery, especially with traditional chemotherapeutic agents like cisplatin, has shown potential in overcoming limitations such as low bioavailability and systemic toxicity of chemotherapies. Moreover, IONPs, by releasing iron ions, can induce ferroptosis, a form of iron-dependent cell death, which offers a promising pathway to reverse radio- and chemoresistance in cancer therapy. In particular, IONPs demonstrate significant potential as radiosensitisers, enhancing the effects of radiotherapy by promoting reactive oxygen species (ROS) generation, lipid peroxidation, and modulating the tumour microenvironment to stimulate antitumour immune responses. This review explores the multifunctional roles of IONPs in radiosensitisation through ferroptosis induction, highlighting their promise in advancing treatment for head and neck cancers. Additional research is crucial to fully addressing their potential in clinical settings, offering a novel approach to personalised cancer treatment.
Collapse
Affiliation(s)
- Dorianne Sant’Angelo
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Géraldine Descamps
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Valentin Lecomte
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Dimitri Stanicki
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Sébastien Penninckx
- Department of Medical Physics, Institut Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Tatiana Dragan
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Dirk Van Gestel
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Fabrice Journe
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
20
|
Zhao Y, Chen Z, Xie S, Xiao F, Hu Q, Ju Z. The emerging role and therapeutical implications of ferroptosis in wound healing. BURNS & TRAUMA 2025; 13:tkae082. [PMID: 39958433 PMCID: PMC11827611 DOI: 10.1093/burnst/tkae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/11/2024] [Accepted: 12/03/2024] [Indexed: 02/18/2025]
Abstract
Wound healing is a complex biological process involving multiple steps, including hemostasis, inflammation, proliferation, and remodeling. A novel form of regulated cell death, ferroptosis, has garnered attention because of its involvement in these processes. Ferroptosis is characterized by the accumulation of lipid peroxides and is tightly regulated by lipid metabolism, iron metabolism, and the lipid-peroxide repair network, all of which exert a significant influence on wound healing. This review highlights the current findings and emerging concepts regarding the multifaceted roles of ferroptosis throughout the stages of normal and chronic wound healing. Additionally, the potential of targeted interventions aimed at modulating ferroptosis to improve wound-healing outcomes is discussed.
Collapse
Affiliation(s)
- Yanan Zhao
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Shenghao Xie
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Feng Xiao
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Qian Hu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, No. 601, Huangpu Avenue West, Tianhe District, Guangzhou, 510632, China
| |
Collapse
|
21
|
Pan G, Xia Y, Hao M, Guan J, Zhu Q, Zha T, Sheng L, Zhao Z, Pan H, Fang W, Xu X, Chen X, Zhou S, Tong Z. EZH2 suppresses IR-induced ferroptosis by forming a co-repressor complex with HIF-1α to inhibit ACSL4: Targeting EZH2 enhances radiosensitivity in KDM6A-deficient esophageal squamous carcinoma. Cell Death Differ 2025:10.1038/s41418-025-01451-5. [PMID: 39920286 DOI: 10.1038/s41418-025-01451-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/29/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
The mutation status of the lysine demethylase 6 A (KDM6A), a gene antagonist to Enhancer of zeste homolog 2 (EZH2), is closely related to the therapeutic efficacy of EZH2 inhibitors in several malignancies. However, the mutational landscape of KDM6A and the therapeutic targetability of EZH2 inhibitors in esophageal squamous carcinoma (ESCC) remain unreported. Here, we found that approximately 9.18% (9/98) of our study ESCC tissues had KDM6A mutations of which 7 cases resulted in a complete loss of expression and consequent loss of demethylase function. We found that KDM6A-deficient ESCC cells exhibited increased sensitivity to EZH2 inhibitor, and the radiosensitizing activity of EZH2 inhibitor was evident in KDM6A-dficient ESCC cells. Further transcriptome analysis revealed that ferroptosis is implicated in the radiosensitizing effect exerted by EZH2 inhibition on KDM6A-deficient ESCC cells. The following Chromatin Immunoprecipitation (ChIP), co-immunoprecipitation, and luciferase reporter assays demonstrated that in KDM6A-deficient ESCC cells, (1) Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4) is the target gene for EZH2 to regulate ferroptosis; (2) The IR-induced hypoxia inducible factor 1 subunit alpha (HIF-1α) is a predominant mediator of EZH2 to repress ACSL4; (3) the HRE7-8 regions of the ACSL4 promoter are required for the repressive function of EZH2 on ACSL4; (4) EZH2 regulates ACSL4 by forming a co-repressive complex with HIF-1α. Our study provides preclinical evidence supporting that EZH2 inhibitors may confer therapeutic benefit in KDM6A-deficient ESCC patients.
Collapse
Affiliation(s)
- Guizhen Pan
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yeye Xia
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Mengyu Hao
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jiahao Guan
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qianqian Zhu
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Radiation Oncology, Fuyang Tumour Hospital, Fuyang, China
| | - Tianqi Zha
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lei Sheng
- Department of Radiation Oncology, the Chest Hospital of Anhui Province, Hefei, Anhui, China
| | - Zhenfeng Zhao
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Huaguang Pan
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Weiyang Fang
- Department of Electrocardiography, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyong Xu
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiangcun Chen
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shuguang Zhou
- The Fifth Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Zhuting Tong
- Department of Radiation Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
22
|
Neitzel LR, Fuller DT, Cornell J, Rea S, de Aguiar Ferreira C, Williams CH, Hong CC. Inhibition of GPR68 induces ferroptosis and radiosensitivity in diverse cancer cell types. Sci Rep 2025; 15:4074. [PMID: 39900965 PMCID: PMC11791087 DOI: 10.1038/s41598-025-88357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
Radioresistance is thought to be a major consequence of tumor milieu acidification resulting from the Warburg effect. Previously, using ogremorphin (OGM), a small molecule inhibitor of GPR68, an extracellular proton sensing receptor, we demonstrated that GPR68 is a key pro-survival pathway in glioblastoma cells. Here, we demonstrate that GPR68 inhibition also induces ferroptosis in lung cell carcinoma (A549) and pancreatic ductal adenocarcinoma (Panc02) cells. Moreover, OGM synergized with ionizing radiation to induce lipid peroxidation, a hallmark of ferroptosis, as well as reduce colony size in 2D and 3D cell culture. GPR68 inhibition is not acutely detrimental but increases intracellular free ferrous iron, which is known to trigger reactive oxygen species (ROS) generation. In summary, GPR68 inhibition induces lipid peroxidation in cancer cells and sensitizes them to ionizing radiation in part through the mobilization of intracellular free ferrous iron. Our results suggest that GPR68 is a key mediator of cancer cell radioresistance activated by acidic tumor microenvironment.
Collapse
Affiliation(s)
- Leif R Neitzel
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI, USA
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI, USA
| | - Daniela T Fuller
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jessica Cornell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Samantha Rea
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Carolina de Aguiar Ferreira
- The Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Department of Radiology, Michigan State University, East Lansing, MI, 48824, USA
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, 48824, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Charles H Williams
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI, USA.
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI, USA.
| | - Charles C Hong
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI, USA.
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI, USA.
| |
Collapse
|
23
|
Bedolla N, Liu L, Liu X, Xie Q, Ren Y. Ursolic acid enhances radiosensitivity in esophageal squamous cell carcinoma by modulating p53/SLC7A11/GPX4 pathway-mediated ferroptosis. Toxicon 2025; 255:108233. [PMID: 39788329 DOI: 10.1016/j.toxicon.2025.108233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
BACKGROUND Radiotherapy is essential for the management of esophageal squamous cell carcinoma (ESCC). However, ESCC cells are highly susceptible to developing resistance to radiotherapy, leading to poor prognosis. Ursolic acid (UA) is a herbal monomer, has multiple medicinal benefits like anti-tumor. The impact of UA on the sensitivity of ESCC cells to radiotherapy is currently unclear. METHODS The impact of UA and ionizing radiation (IR) on the viability of TE-1 and KYSE30 cells was assessed by the MTT assay. EdU staining, flow cytometry, clone formation, Wound healing and Transwell assay detected the biological properties of ESCC cells. FerroOrange, DCFH-DA, and kits to detect the influences of UA and/or IR treatment on cellular ferroptosis. The levels of p53/solute carrier family 7a member 11 (SLC7A11)/glutathione peroxidase 4 (GPX4) pathway proteins were detected by Western blot. Additionally, a subcutaneous graft tumor model was constructed in nude mice. RESULTS 10 μM UA reduced the viability and induced death of ESCC cells. UA enhanced the impacts of IR by suppressing cell proliferation, migration and invasion, inducing cell death, and causing cell cycle arrest. Ferroptosis inhibitor impaired the inhibitory impacts of UA and IR on the biological properties of ESCC cells. The combination of UA and IR led to ferroptosis through the modulation of the p53/SLC7A11/GPX4 pathway, and UA enhanced the responsiveness of ESCC cells to IR both in vitro and in vivo. CONCLUSION UA inhibits the malignant biological behavior of ESCC by modulating ferroptosis through the p53/SLC7A11/GPX4 pathway, and enhances the sensitivity of ESCC cells to IR.
Collapse
Affiliation(s)
- Nuran Bedolla
- College of Biological Sciences and Technology, YiLi Normal University, China
| | - Linyu Liu
- College of Biological Sciences and Technology, YiLi Normal University, China
| | - Xueting Liu
- College of Biological Sciences and Technology, YiLi Normal University, China
| | - Qiuxian Xie
- College of Biological Sciences and Technology, YiLi Normal University, China
| | - Yanli Ren
- College of Biological Sciences and Technology, YiLi Normal University, China.
| |
Collapse
|
24
|
Noh JK, Lee MK, Lee Y, Bae M, Min S, Kong M, Lee JW, Kim SI, Lee YC, Ko S, Woo SR, Eun Y. Targeting ferroptosis for improved radiotherapy outcomes in HPV-negative head and neck squamous cell carcinoma. Mol Oncol 2025; 19:540-557. [PMID: 39297393 PMCID: PMC11792990 DOI: 10.1002/1878-0261.13720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/16/2024] [Accepted: 08/01/2024] [Indexed: 02/05/2025] Open
Abstract
To enhance the efficacy of radiotherapy (RT) in human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC), we explored targeting ferroptosis, a regulated cell death process. We developed a gene signature associated with ferroptosis using Cox proportional hazard modeling in HPV-negative HNSCC patients who underwent RT. This ferroptosis-related gene signature (FRGS) was a significant predictor of overall survival and recurrence-free survival in HPV-negative HNSCC patients who received RT. Subtype B of the FRGS, characterized by decreased expression of ferroptosis inducers [nuclear receptor coactivator 4 (NCOA4) and natural resistance-associated macrophage protein 2 homolog/divalent metal transporter 1 (NRAMP2/DMT1)] and increased expression of suppressors [phospholipid hydroperoxide glutathione peroxidase (GPX4) and ferritin heavy chain (FTH1)], was associated with poorer prognosis, potentially indicating the inhibition of ferroptosis. Furthermore, our in vitro and in vivo studies demonstrated that treatment with statins, such as atorvastatin and simvastatin, induced ferroptosis and sensitized radioresistant HNSCC cells to irradiation, improving radiosensitivity and potentially enhancing the response to RT. Additionally, in xenograft models, the combination of statins and RT led to a significant reduction in tumor initiation. These findings provide valuable insights for enhancing treatment and improving prognosis in HPV-negative HNSCC by targeting ferroptosis and utilizing statins to sensitize tumors to RT-induced cell death.
Collapse
Affiliation(s)
- Joo Kyung Noh
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Min Kyeong Lee
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Yeonseo Lee
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Minji Bae
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Soonki Min
- Department of Radiation OncologyKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Moonkyoo Kong
- Department of Radiation OncologyKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Jung Woo Lee
- Department of Oral and Maxillofacial Surgery, School of DentistryKyung Hee UniversitySeoulKorea
| | - Su Il Kim
- Department of Otolaryngology‐Head and Neck SurgeryKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Young Chan Lee
- Department of Otolaryngology‐Head and Neck SurgeryKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Seong‐Gyu Ko
- Department of Preventive Medicine, College of Korean MedicineKyung Hee UniversitySeoulKorea
| | - Seon Rang Woo
- Department of Otolaryngology‐Head and Neck SurgeryKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| | - Young‐Gyu Eun
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
- Department of Otolaryngology‐Head and Neck SurgeryKyung Hee University School of Medicine, Kyung Hee University Medical CenterSeoulKorea
| |
Collapse
|
25
|
Chen X, Wang X, Zou Y, Wang Y, Duan T, Zhou Z, Huang Y, Ye Q. EMC2 suppresses ferroptosis via regulating TFRC in nasopharyngeal carcinoma. Transl Oncol 2025; 52:102251. [PMID: 39709720 PMCID: PMC11832954 DOI: 10.1016/j.tranon.2024.102251] [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: 11/20/2023] [Revised: 11/15/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is an epithelial malignancy with poorly understood underlying molecular mechanisms. Ferroptosis, a form of programmed cell death, is not fully elucidated in NPC. METHOD We conducted quantitative proteomics to detect dysregulated proteins in NPC tissues. The levels of endoplasmic reticulum membrane protein complex 2 (EMC2) in NPC tissue microarrays were evaluated by immunohistochemistry, and the prognostic value of EMC2 was analyzed in NPC patients. The role of EMC2 in ferroptosis and carcinogenesis was determined through in vitro and in vivo experiments. Quantitative proteomics, protease inhibition, ubiquitin detection, and rescue experiments were performed to explore the mechanism of EMC2-regulated ferroptosis. RESULTS Significantly upregulated EMC2 was detected in NPC, and it was closely related to the characteristics of tumor progression. Elevated EMC2 was obviously correlated with poor survival in patients with NPC. EMC2 knockdown promoted ferroptosis, inhibiting cell viability, migration, and invasion, and enhancing the efficacy of cisplatin in NPC cells. Conversely, EMC2 overexpression contributed to ferroptosis repression, malignant progression, and reduced the efficacy of cisplatin. In addition, EMC2 knockdown suppressed xenograft tumor growth and enhanced ferroptosis in nude mice. Mechanistically, we identified transferrin receptor (TFRC) as a critical downstream protein. EMC2 interacted with TFRC and promoted its ubiquitin-proteasomal degradation. EMC2 regulated ferroptosis by mediating the level of TFRC. CONCLUSIONS EMC2 suppresses ferroptosis and promotes tumor progression, and the EMC2-TFRC axis is a novel ferroptosis regulatory pathway. EMC2 is a potentially biomarker and therapeutic target for NPC.
Collapse
Affiliation(s)
- Xianghui Chen
- Shengli Clinical Medical College of Fujian Medical University, Department of Otolaryngology, Head and Neck Surgery, Fujian Provincial Hospital, Fuzhou 350001, China; Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, China
| | - Xiaoyan Wang
- Shengli Clinical Medical College of Fujian Medical University, Department of Otolaryngology, Head and Neck Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yuxia Zou
- Shengli Clinical Medical College of Fujian Medical University, Department of Otolaryngology, Head and Neck Surgery, Fujian Provincial Hospital, Fuzhou 350001, China; Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 571199, China
| | - Yan Wang
- Shengli Clinical Medical College of Fujian Medical University, Department of Otolaryngology, Head and Neck Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Tingting Duan
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 571199, China
| | - Zijie Zhou
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Yi Huang
- Shengli Clinical Medical College of Fujian Medical University, Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Qing Ye
- Shengli Clinical Medical College of Fujian Medical University, Department of Otolaryngology, Head and Neck Surgery, Fujian Provincial Hospital, Fuzhou 350001, China.
| |
Collapse
|
26
|
Gao G, Zhang X. Broadening horizons: research on ferroptosis in lung cancer and its potential therapeutic targets. Front Immunol 2025; 16:1542844. [PMID: 39917300 PMCID: PMC11799241 DOI: 10.3389/fimmu.2025.1542844] [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: 12/10/2024] [Accepted: 01/03/2025] [Indexed: 02/09/2025] Open
Abstract
Ferroptosis is a novel form of cell death distinct from traditional mechanisms, characterized by the accumulation of iron ions and the production of lipid peroxides. It not only affects the survival of tumor cells but is also closely linked to changes in the tumor microenvironment. Lung cancer is one of the leading malignancies worldwide in terms of incidence and mortality, and its complex biological mechanisms and resistance make treatment challenging. Recent studies have shown that ferroptosis plays a key role in the onset and progression of lung cancer, with its intricate regulatory mechanisms influencing tumor development and response to therapy. As research into ferroptosis deepens, related molecular pathways, such as glutamate metabolism, iron metabolism, and antioxidant defense, have been gradually revealed. However, in clinical practice, ferroptosis-based therapeutic strategies for lung cancer are still in their early stages. Challenges remain, including the incomplete understanding of the specific mechanisms of ferroptosis, insufficient research on related regulatory factors, and limited insight into the interactions within the tumor microenvironment. Therefore, effective modulation of ferroptosis to enhance lung cancer treatment remains an urgent issue. This review summarizes the biological mechanisms of ferroptosis, analyzes the regulatory factors of ferroptosis in lung cancer cells and their interaction with the tumor microenvironment, and further explores potential therapeutic strategies targeting ferroptosis. By synthesizing the latest research, this paper aims to provide new perspectives and directions for lung cancer treatment, with the goal of advancing clinical applications.
Collapse
Affiliation(s)
| | - Xindi Zhang
- Department of Pulmonary Disease (Department of Respiratory and Critical Care Medicine), Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
27
|
Li Q, Wu Z, Chen S, Liang Y, Zhu K, Su N, Liu T, Zhao B. Enhancing Ferroptosis-Mediated Radiosensitization via Synergistic Disulfidptosis Induction. ACS NANO 2025; 19:1757-1770. [PMID: 39727204 DOI: 10.1021/acsnano.4c15932] [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: 12/28/2024]
Abstract
Ferroptosis plays an important role in radiotherapy (RT), and the induction of ferroptosis can effectively sensitize radiotherapy. However, the therapeutic efficiency is always affected by ferroptosis resistance, especially SLC7A11 (Solute Carrier Family 7 Member 11)-cystine-cysteine-GSH (glutathione)-GPX4 (glutathione peroxidase 4) pathway-mediated resistance. In this study, tumor-microenvironment self-activated high-Z element-containing nanoferroptosis inducers, PEGylated Fe-Bi-SS metal-organic frameworks (FBSP MOFs), were developed to sensitize RT. Unexpectedly, ferroptosis-resistant SLC7A11 would be self-adaptively upregulated, leading to self-responsive ferroptosis resistance. Since the conversion from SLC7A11-transported cystine to cysteine is highly glucose-dependent, glucose oxidase (GOx) was incorporated in the MOFs, causing the depletion of NADPH (nicotinamide adenine dinucleotide phosphate) to terminate the conversion from cystine to cysteine, relieving the self-adaptive ferroptosis resistance. Excitingly, the accumulation of cystine would synergistically lead to disulfide stress and trigger disulfidptosis, making a new contribution to enhance therapeutic efficiency. Moreover, the hydrogen peroxide produced during glucose oxidation could also cascade-react with the Fenton reaction, therefore enhancing ferropotosis. Both in vitro and in vivo results suggested that therapeutic efficiency of ferroptosis-mediated radiosensitization could be enhanced benefiting from synergistic disulfidptosis induction, indicating that disulfidptosis might be an efficient strategy to relieve ferroptosis resistance and enhance RT efficiency.
Collapse
Affiliation(s)
- Qiuyu Li
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Zede Wu
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P.R. China
| | - Siwen Chen
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
| | - Yu Liang
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kai Zhu
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P.R. China
| | - Ning Su
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, P.R. China
| | - Bingxia Zhao
- Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, P.R. China
- Experiment Education/Administration Center, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, P.R. China
| |
Collapse
|
28
|
Imam M, Ji J, Zhang Z, Yan S. Targeting the initiator to activate both ferroptosis and cuproptosis for breast cancer treatment: progress and possibility for clinical application. Front Pharmacol 2025; 15:1493188. [PMID: 39867656 PMCID: PMC11757020 DOI: 10.3389/fphar.2024.1493188] [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: 09/08/2024] [Accepted: 11/12/2024] [Indexed: 01/28/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide. Metal metabolism is pivotal for regulating cell fate and drug sensitivity in breast cancer. Iron and copper are essential metal ions critical for maintaining cellular function. The accumulation of iron and copper ions triggers distinct cell death pathways, known as ferroptosis and cuproptosis, respectively. Ferroptosis is characterized by iron-dependent lipid peroxidation, while cuproptosis involves copper-induced oxidative stress. They are increasingly recognized as promising targets for the development of anticancer drugs. Recently, compelling evidence demonstrated that the interplay between ferroptosis and cuproptosis plays a crucial role in regulating breast cancer progression. This review elucidates the converging pathways of ferroptosis and cuproptosis in breast cancer. Moreover, we examined the value of genes associated with ferroptosis and cuproptosis in the clinical diagnosis and treatment of breast cancer, mainly outlining the potential for a co-targeting approach. Lastly, we delve into the current challenges and limitations of this strategy. In general, this review offers an overview of the interaction between ferroptosis and cuproptosis in breast cancer, offering valuable perspectives for further research and clinical treatment.
Collapse
Affiliation(s)
| | | | | | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
29
|
Wang Y, He Z, Dong X, Yao Y, Chen Q, Shi Y, Deng Y, Zhang Q, Yu L, Wang C. Regulation and therapy: the role of ferroptosis in DLBCL. Front Pharmacol 2025; 15:1458412. [PMID: 39834804 PMCID: PMC11743434 DOI: 10.3389/fphar.2024.1458412] [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: 07/02/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of B-cell non-Hodgkin's lymphoma (NHL), up to 30%-40% of patients will relapse and 10%-15% of patients have primary refractory disease, so exploring new treatment options is necessary. Ferroptosis is a non-apoptotic cell death mode discovered in recent years. Its occurrence pathway plays an essential impact on the therapeutic effect of tumors. Numerous studies have shown that modulating critical factors in the ferroptosis pathway can influence the growth of tumor cells in hematological malignancies including DLBCL. This review highlights recent advances in ferroptosis-related genes (FRGs), including STAT3, Nrf2, and ZEB1, and focuses on the clinical potential of ferroptosis inducers such as IKE, α-KG, DMF, and APR-246, which are currently being explored in clinical studies for their therapeutic effects in DLBCL. Correlational studies provide a novel idea for the research and treatment of ferroptosis in DLBCL and other hematological malignancies and lay a solid foundation for future studies.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Zhengmei He
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Xinyu Dong
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| | - Yiming Yao
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| | - Qiuni Chen
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yuye Shi
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Yuan Deng
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Quane Zhang
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Liang Yu
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| | - Chunling Wang
- Department of Hematology, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai’an, China
- Northern Jiangsu Institute of Clinical Medicine, Nanjing Medical University, Nanjing, China
- Department of Hematology, The Huaian Clinical College of Xuzhou Medical University, Huai’an, China
| |
Collapse
|
30
|
Geirnaert F, Kerkhove L, Montay-Gruel P, Gevaert T, Dufait I, De Ridder M. Exploring the Metabolic Impact of FLASH Radiotherapy. Cancers (Basel) 2025; 17:133. [PMID: 39796760 PMCID: PMC11720285 DOI: 10.3390/cancers17010133] [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: 12/16/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
FLASH radiotherapy (FLASH RT) is an innovative modality in cancer treatment that delivers ultrahigh dose rates (UHDRs), distinguishing it from conventional radiotherapy (CRT). FLASH RT has demonstrated the potential to enhance the therapeutic window by reducing radiation-induced damage to normal tissues while maintaining tumor control, a phenomenon termed the FLASH effect. Despite promising outcomes, the precise mechanisms underlying the FLASH effect remain elusive and are a focal point of current research. This review explores the metabolic and cellular responses to FLASH RT compared to CRT, with particular focus on the differential impacts on normal and tumor tissues. Key findings suggest that FLASH RT may mitigate damage in healthy tissues via altered reactive oxygen species (ROS) dynamics, which attenuate downstream oxidative damage. Studies indicate the FLASH RT influences iron metabolism and lipid peroxidation pathways differently than CRT. Additionally, various studies indicate that FLASH RT promotes the preservation of mitochondrial integrity and function, which helps maintain apoptotic pathways in normal tissues, attenuating damage. Current knowledge of the metabolic influences following FLASH RT highlights its potential to minimize toxicity in normal tissues, while also emphasizing the need for further studies in biologically relevant, complex systems to better understand its clinical potential. By targeting distinct metabolic pathways, FLASH RT could represent a transformative advance in RT, ultimately improving the therapeutic window for cancer treatment.
Collapse
Affiliation(s)
- Febe Geirnaert
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| | - Lisa Kerkhove
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| | - Pierre Montay-Gruel
- Radiation Oncology Department, Iridium Netwerk, 2610 Antwerp, Belgium;
- Antwerp Research in Radiation Oncology (AreRO), Center for Oncological Research (CORE), University of Antwerp, 2020 Antwerp, Belgium
| | - Thierry Gevaert
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| | - Inès Dufait
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| | - Mark De Ridder
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| |
Collapse
|
31
|
Su Z, Liu Y, Wang L, Gu W. Regulation of SLC7A11 as an unconventional checkpoint in tumorigenesis through ferroptosis. Genes Dis 2025; 12:101254. [PMID: 39569390 PMCID: PMC11577153 DOI: 10.1016/j.gendis.2024.101254] [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: 12/21/2023] [Revised: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 11/22/2024] Open
Abstract
Although cell-cycle arrest, senescence, and apoptosis are well accepted as the classic barriers in tumorigenesis, recent studies indicate that metabolic regulation is equally important as a major checkpoint in cancer development. It is well accepted that ferroptosis, an iron-dependent programmed cell death, acts as a new type of tumor suppression mechanism tightly linked with numerous metabolic pathways. SLC7A11 is a transmembrane cystine/glutamate transporter protein that plays a vital role in controlling ferroptosis in vivo. The levels of SLC7A11 are dynamically regulated by various types of stresses, such as oxidative stress, nutrient deprivation, endoplasmic reticulum stress, radiation, oncogenic stress, DNA damage, and immune stress. SLC7A11 can be transcriptionally regulated by both activators such as ATF4, NRF2, and ETS1, and repressors including BACH1, p53, ATF3, and STAT1 during stress responses. Moreover, SLC7A11 activity and its protein stability and cellular localization are also modulated upon stress. Patients' data show that SLC7A11 is overexpressed in various types of human cancers, and higher levels of SLC7A11 predict poorer overall survival. Growing evidence also suggests that targeting SLC7A11 is a promising approach in cancer therapy by effectively inhibiting tumor proliferation, invasion, and metastasis, as well as counteracting cancer stem cells and overcoming chemoresistance. This review highlights the regulation of SLC7A11 as an unconventional checkpoint in tumorigenesis through modulating ferroptotic responses under various types of stress.
Collapse
Affiliation(s)
- Zhenyi Su
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yanqing Liu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lin Wang
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
32
|
Christopher JA, Breckels LM, Crook OM, Vazquez-Chantada M, Barratt D, Lilley KS. Global Proteomics Indicates Subcellular-Specific Anti-Ferroptotic Responses to Ionizing Radiation. Mol Cell Proteomics 2025; 24:100888. [PMID: 39617061 PMCID: PMC11780130 DOI: 10.1016/j.mcpro.2024.100888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 01/11/2025] Open
Abstract
Cells have many protective mechanisms against background levels of ionizing radiation orchestrated by molecular changes in expression, post-translational modifications, and subcellular localization. Radiotherapeutic treatment in oncology attempts to overwhelm such mechanisms, but radioresistance is an ongoing challenge. Here, global subcellular proteomics combined with Bayesian modeling identified 544 differentially localized proteins in A549 cells upon 6 Gy X-ray exposure, revealing subcellular-specific changes of proteins involved in ferroptosis, an iron-dependent cell death, suggestive of potential radioresistance mechanisms. These observations were independent of expression changes, emphasizing the utility of global subcellular proteomics and the promising prospect of ferroptosis-inducing therapies for combating radioresistance.
Collapse
Affiliation(s)
- Josie A Christopher
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, UK.
| | - Lisa M Breckels
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Oliver M Crook
- Department of Statistics, University of Oxford, Oxford, UK
| | | | | | - Kathryn S Lilley
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
33
|
Chen H, Lyu F, Gao X. Advances in ferroptosis for castration-resistant prostate cancer treatment: novel drug targets and combination therapy strategies. Prostate Cancer Prostatic Dis 2024:10.1038/s41391-024-00933-w. [PMID: 39733054 DOI: 10.1038/s41391-024-00933-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Metastatic prostate cancer (PCa) has much lower survival and ultimately develops castration resistance, which expects novel targets and therapeutic approaches. As a result of iron-dependent lipid peroxidation, ferroptosis triggers programmed cell death and has been associated with castration-resistant prostate cancer (CRPC). SUBJECTS To better understand how ferroptosis can be used to treat CRPC, we reviewed the following: First, ferroptosis mechanisms and characteristics. We then pay attention to ferroptosis effects on CRPC, and the relationship between ferroptosis and CRPC treatment. Finally, we'd like to figure out if ferroptosis could predict the prognosis of CRPC thus screening early for populations that may benefit from appropriate therapies. RESULTS The review demonstrated that ferroptosis regulators like PI3K/AKT/mTOR, DECR1 et al., have a significant role in the development of CRPC and that several inducers of ferroptosis, such as erastin, BSO, RSL3, and FIN56, have already demonstrated their effects in that area. What's more, ferroptosis is crucial for radiation-induced anticancer effects by inducing lipid peroxidation and regulating p53, AMPK, and others. Additionally, it has been discovered that certain GPX4 and SLC7A11 inhibitors can increase radiosensitivity, which brings new combination strategies. Finally, among the genes associated with ferroptosis, which may be excellent predictors of prostate cancer prognosis, several risk models have been developed and shown promising predictive capabilities. CONCLUSIONS Ferroptosis can serve as a potential therapeutic target for CRPC, and could be a new strategy for combination therapy. Moreover, ferroptosis-related genes may be great indicators of PCa prognosis. Further research on ferroptosis in CRPC therapy can benefit from the frameworks provided by this review.
Collapse
Affiliation(s)
- Huizhu Chen
- Department of Radiation Oncology, Peking University First Hospital, 100034, Beijing, China
| | - Feng Lyu
- Department of Radiation Oncology, Peking University First Hospital, 100034, Beijing, China
| | - Xianshu Gao
- Department of Radiation Oncology, Peking University First Hospital, 100034, Beijing, China.
| |
Collapse
|
34
|
Li Y, Liu C, Fang B, Chen X, Wang K, Xin H, Wang K, Yang SM. Ferroptosis, a therapeutic target for cardiovascular diseases, neurodegenerative diseases and cancer. J Transl Med 2024; 22:1137. [PMID: 39710702 DOI: 10.1186/s12967-024-05881-6] [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: 09/02/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The identification of ferroptosis represents a pivotal advancement in the field of cell death research, revealing an entirely novel mechanism of cellular demise and offering new insights into the initiation, progression, and therapeutic management of various diseases. Ferroptosis is predominantly induced by intracellular iron accumulation, lipid peroxidation, or impairments in the antioxidant defense system, culminating in membrane rupture and consequent cell death. Studies have associated ferroptosis with a wide range of diseases, and by enhancing our comprehension of its underlying mechanisms, we can formulate innovative therapeutic strategies, thereby providing renewed hope for patients.
Collapse
Affiliation(s)
- Yinghui Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Cuiyun Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bo Fang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xinzhe Chen
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Kai Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China.
| | - Kun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Su-Min Yang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
35
|
Zhang P, Liu W, Wang S, Wang Y, Han H. Ferroptosisand Its Role in the Treatment of Sepsis-Related Organ Injury: Mechanisms and Potential Therapeutic Approaches. Infect Drug Resist 2024; 17:5715-5727. [PMID: 39720615 PMCID: PMC11668052 DOI: 10.2147/idr.s496568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/12/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis is a complicated clinical disease caused by a defective host response to infection, leading to elevated morbidity and fatality globally. Sepsis patients have a significant risk of life-threatening organ damage, including hearts, brains, lungs, kidneys, and livers. Nevertheless, the molecular pathways driving organ injury in sepsis are not well known. Ferroptosis, a non-apoptotic cell death, occurs due to iron metabolism disturbance and lipid peroxide buildup. Multiple studies indicate that ferroptosis has a significant role in decreasing inflammation and lipid peroxidation during sepsis. Ferroptosis inhibitors and medications, aimed at the most studied ferroptosis process, including Xc-system, Nrf2/GPX4 axis, and NCOA4-FTH1-mediated ferritinophagy, alleviating sepsis effectively. However, few clinical trials demonstrated ferroptosis-targeted drugs's effectiveness in sepsis. Our study examines ferroptosis-targeted medicinal agents and their potential benefits for treating sepsis-associated organ impairment. This review indicates that ferroptosis suppression by pharmaceutical means may be a useful therapy for sepsis-associated organ injury.
Collapse
Affiliation(s)
- Pengyu Zhang
- The Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Wendi Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Shu Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yuan Wang
- Department of Histology and Embryology, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Han Han
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
36
|
Han JM, Mwiti G, Yeom SJ, Lim J, Kim WS, Lim S, Lim ST, Byun EB. Radiation-Resistant Bacteria Deinococcus radiodurans-Derived Extracellular Vesicles as Potential Radioprotectors. Adv Healthc Mater 2024:e2403192. [PMID: 39676336 DOI: 10.1002/adhm.202403192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/26/2024] [Indexed: 12/17/2024]
Abstract
The increasing use of radiation presents a risk of radiation exposure, making the development of radioprotectors necessary. In the previous study, it is investigated that Deinococcus radiodurans (R1-EVs) exert the antioxidative properties. However, the radioprotective activity of R1-EVs remains unclear. In the present study, the protective effects of R1-EVs against total body irradiation (TBI)-induced acute radiation syndrome (ARS) are investigated. To assess R1-EVs' radioprotective efficacy, ARS is induced in mice with 8 Gy of TBI, and protection against hematopoietic (H)- and gastrointestinal (GI)-ARS is evaluated. The survival rate of irradiated mice group decreases substantially after irradiation. In contrast, pretreatment with R1-EVs increases the survival rates of the mice. The administration of R1-EVs provides effective protection against radiation-induced death of bone marrow cells and splenocytes by scavenging reactive oxygen species (ROS). Additionally, R1-EVs protect both intestinal stem and epithelial cells from radiation-induced apoptosis. R1-EVs stimulate the production of short-chain fatty acids in the gastrointestinal tract, suppress proinflammatory cytokines, and increase regulatory T cells in pretreated mice versus the irradiation-only group. Proteomic analysis shows that the R1-EV proteome is significantly enriched with proteins involved in oxidative stress response. These findings highlight R1-EVs as potent radioprotectors with applications against radiation damage and ROS-mediated diseases.
Collapse
Affiliation(s)
- Jeong Moo Han
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 03080, South Korea
- Institute for Data Innovation in Science, Seoul National University, Seoul, 08826, South Korea
| | - Godfrey Mwiti
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, 3000, Australia
- Department of Food and Nutrition, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Seo-Joon Yeom
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jaeyoon Lim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Food and Nutrition, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Woo Sik Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Sangyong Lim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Radiation Science, University of Science and Technology, Daejeon, 01812, Republic of Korea
| | - Seung-Taik Lim
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Eui-Baek Byun
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| |
Collapse
|
37
|
Wang X, Shi W, Li M, Xin Y, Jiang X. RSL3 sensitizes glioma cells to ionizing radiation by suppressing TGM2-dependent DNA damage repair and epithelial-mesenchymal transition. Redox Biol 2024; 78:103438. [PMID: 39580966 PMCID: PMC11625373 DOI: 10.1016/j.redox.2024.103438] [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: 10/18/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
RAS-selective lethal small molecule 3 (RSL3) is a small-molecule compound that triggers ferroptosis by inactivating glutathione peroxidase 4. However, its effect on the radioresistance of glioma cells and the underlying mechanisms remains unclear. In this study, we found that RSL3 sensitized glioma cells to ionizing radiation (IR) and enhanced IR-induced DNA double-strand breaks (DSBs). Inhibition of ferroptosis pathways partly prevented the clonogenic death caused by the IR/RSL3 combination but did not alleviate the DNA DSBs, indicating that RSL3 promotes IR-induced DNA DSBs via a non-ferroptotic mechanism. We demonstrated that transglutaminase 2 (TGM2) plays a vital role in the radiosensitization effect of RSL3 on glioma cells. Treatment with RSL3 downregulated TGM2 in a dose-dependent manner. Overexpression of TGM2 not only alleviated DNA DSBs but also inhibited clonogenic death caused by the IR/RSL3 combination. Mechanistically, RSL3 triggered oxidative stress in glioma cells, which promoted the S-gluthathionylation of TGM2 via upregulation of glutathione S-transferase P1(GSTP1), culminating in the proteasomal degradation of TGM2. This process resulted in the suppression of DNA repair mechanisms by impeding the nuclear accumulation of TGM2 and disrupting the interaction between TGM2 and topoisomerase IIα after irradiation. We also found that RSL3 inhibited glioma cell epithelial-mesenchymal transition (EMT) in both IR-treated and non-IR-treated cells. Overexpression of TGM2 prevented, while knockdown of TGM2 aggravated the EMT inhibition caused by RSL3, indicating that RSL3 also sensitized glioma cells to IR by inhibiting EMT via a TGM2-dependent mechanism. Furthermore, in mice bearing human U87 tumor xenografts, RSL3 administration synergized with IR to inhibit tumor growth, accompanied by TGM2 inhibition, DNA DSBs, and EMT inhibition in tumor tissues. Taken together, we demonstrated that RSL3 sensitizes glioma cells to IR by suppressing TGM2-mediated DNA repair and EMT.
Collapse
Affiliation(s)
- Xuanzhong Wang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| | - Weiyan Shi
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| | - Mengxin Li
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| |
Collapse
|
38
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [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: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
39
|
Jin Y, Pan S, Wang M, Huang S, Ke Y, Li D, Luo H, Kou Z, Shi D, Kou W, Fu H, Pan J. The m 6A modification of ACSL4 mRNA sensitized esophageal squamous cell carcinoma to irradiation via accelerating ferroptosis. Cell Biol Int 2024; 48:1877-1890. [PMID: 39285560 DOI: 10.1002/cbin.12245] [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: 06/18/2024] [Revised: 08/12/2024] [Accepted: 08/23/2024] [Indexed: 11/15/2024]
Abstract
Radioresistance is a major obstacle for the therapy of esophageal squamous cell carcinoma (ESCC) and lead to a poor prognosis. Ferroptosis is supposed to be responsible for radioresistance. However, the ferroptosis-induced radioresistance in ESCC and its related regulatory mechanisms are not yet fully understood. In this study, human ESCC cell line and the corresponding radioresistance cells were irradiated with 6 megavolts (MV) X-ray. It was showed that irradiation led to less ferroptosis in radioresistant ESCC cells as compared to the parental cells, as depicted by transmission electron microscopy, intracellular Fe2+ iron contents, lipid peroxidation, and expression of COX2. The increase of ASCL4 expression levels in radioresistant cells after radiotherapy was smaller than that in the parental cells. ACSL4 overexpression significantly enhanced ferroptosis. The fold increase in ACSL4 m6A modification in the radioresistant cells was significantly smaller than that in the parental cells as detected by methylated RNA immunoprecipitation with qRT-PCR. METTL14 overexpression accelerated ferroptosis induced by irradiation via upregulating m6A modification of ACSL4 mRNA. In conclusions, ferroptosis ablation was responsible for the radioresistant of ESCC. The METTL14-mediated m6A modification of ACSL4 mRNA sensitized ESCC to irradiation via accelerating ferroptosis. This study sheds new light on our understanding of radioresistant in ESCC, and provides potential strategies for ESCC radiotherapy.
Collapse
Affiliation(s)
- Yingying Jin
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shupei Pan
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mincong Wang
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shan Huang
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yue Ke
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dan Li
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hen Luo
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhanfeng Kou
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dongwen Shi
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weihua Kou
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hongxiao Fu
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiyuan Pan
- Department of Radiation Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
40
|
Zheng Y, Sun J, Luo Z, Li Y, Huang Y. Emerging mechanisms of lipid peroxidation in regulated cell death and its physiological implications. Cell Death Dis 2024; 15:859. [PMID: 39587094 PMCID: PMC11589755 DOI: 10.1038/s41419-024-07244-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
Regulated cell death (RCD) refers to the form of cell death that can be regulated by various biomacromolecules. Each cell death modalities have their distinct morphological changes and molecular mechanisms. However, intense evidences suggest that lipid peroxidation can be the common feature that initiates and propagates the cell death. Excessive lipid peroxidation alters the property of membrane and further damage the proteins and nucleic acids, which is implicated in various human pathologies. Here, we firstly review the classical chain process of lipid peroxidation, and further clarify the current understanding of the myriad roles and molecular mechanisms of lipid peroxidation in various RCD types. We also discuss how lipid peroxidation involves in diseases and how such intimate association between lipid peroxidation-driven cell death and diseases can be leveraged to develop rational therapeutic strategies.
Collapse
Affiliation(s)
- Yongxin Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
| | - Junlu Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Zhiting Luo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Yimin Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China.
- State Key Laboratory of Respiratory Diseases, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Yongbo Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou Institute of Respiratory Health Guangzhou, Guangzhou, China.
- State Key Laboratory of Respiratory Diseases, Guangzhou, China.
| |
Collapse
|
41
|
Gao Q, Wang X, Zhang Y, Wen J, Wang F, Lin Z, Feng Y, Huang J, Li Q, Luo H, Liu X, Zhai X, Li L, He S, Mi Z, Zhang L, Niu T, Xu C, Zheng Y. Ferroptosis-related prognostic model of mantle cell lymphoma. Open Med (Wars) 2024; 19:20241090. [PMID: 39588389 PMCID: PMC11587922 DOI: 10.1515/med-2024-1090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024] Open
Abstract
Background Mantle cell lymphoma (MCL) is a B-cell non-Hodgkin's lymphoma. Ferroptosis, an iron-dependent programmed cell death, is closely related to cancer prognosis. In this study, we established a model of ferroptosis related genes for prognostic evaluation of patients with MCL. Methods Using the single-cell RNA sequencing datasets GSE184031 and mRNA sequencing data GSE32018 from the Gene Expression Omnibus, we identified 139 ferroptosis-related genes in MCL. Next a prognostic model was constructed by Cox regression and Least absolute selection and shrinkage Operator regression analysis. Finally, we used CIBERSORT to analyze the immune microenvironment and the "oncoPredict" package to predict potential drugs. Results In our model, the prognosis of MCL patients was assessed by risk scoring using 7 genes ANXA1, IL1B, YBX1, CCND1, MS4A1, MFHAS1, and RILPL2. The patients were divided into high-risk and low-risk groups based on our model, and the high-risk patients had inferior overall survival. Finally, according to our model and computational drug sensitivity analysis, four small molecule compounds, BMS-754807, SB216763, Doramapimod, and Trametinib, were identified as potential therapeutic agents for patients with MCL. Conclusion In summary, we provide a prognostic model with ferroptosis-related gene signature for MCL. This study provides a prognostic model with ferroptosis-related gene signature for MCL. The results show that the model helps predict prognosis in MCL.
Collapse
Affiliation(s)
- Qianwen Gao
- Department of Biology, School of Life Science, Sichuan University, Chengdu, China
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xin Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yue Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingjing Wen
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Hematology, Mian-yang Central Hospital, Mianyang, China
| | - Fangfang Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhimei Lin
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Hematology, The Affiliated Hospital of Chengdu University, Chengdu, China
| | - Yu Feng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingcao Huang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qian Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongmei Luo
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Liu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Zhai
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Linfeng Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Siyao He
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziyue Mi
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Caigang Xu
- Department of Hematology, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China
| |
Collapse
|
42
|
Gotorbe C, Segui F, Echavidre W, Durivault J, Blanchard T, Vial V, Pagnuzzi-Boncompagni M, Villeneuve R, Amblard R, Garnier N, Ortholan C, Serrano B, Picco V, Pouysségur J, Vucetic M, Montemagno C. Exploiting Integrin-αVβ3 to Enhance Radiotherapy Efficacy in Medulloblastoma via Ferroptosis. Curr Oncol 2024; 31:7390-7402. [PMID: 39590175 PMCID: PMC11592711 DOI: 10.3390/curroncol31110545] [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: 09/20/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Medulloblastoma, a malignant pediatric brain tumor, has a poor prognosis upon relapse, highlighting a critical clinical need. Our previous research linked medulloblastoma cell radioresistance to integrin-αvβ3 expression. β3-depleted (β3_KO) medulloblastoma cells exhibit lipid hydroxyperoxide accumulation after radiotherapy, indicating ferroptosis, a regulated cell death induced by ROS and inhibited by antioxidants such as cysteine, glutathione (GSH), and glutathione peroxidase 4 (GPx4). However, the link between αvβ3 expression, ferroptosis inhibition, and sensitivity to radiotherapy remains unclear. We showed that irradiated β3_KO medulloblastoma cells primarily die by ferroptosis, with β3-subunit expression correlating with radiotherapy sensitivity and anti-ferroptotic protein levels. Our findings suggest that integrin-αvβ3 signaling boosts oxidative stress resilience via mTORC1. Thus, targeting integrin-αvβ3 could enhance radiotherapy efficacy in medulloblastoma by inducing ferroptotic cell death.
Collapse
Affiliation(s)
- Célia Gotorbe
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Fabien Segui
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - William Echavidre
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Jérôme Durivault
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Thays Blanchard
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Valérie Vial
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Marina Pagnuzzi-Boncompagni
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Rémy Villeneuve
- Radiophysics Department, Princess Grace Hospital, 98000 Monaco, Monaco; (R.V.); (R.A.); (N.G.); (B.S.)
| | - Régis Amblard
- Radiophysics Department, Princess Grace Hospital, 98000 Monaco, Monaco; (R.V.); (R.A.); (N.G.); (B.S.)
| | - Nicolas Garnier
- Radiophysics Department, Princess Grace Hospital, 98000 Monaco, Monaco; (R.V.); (R.A.); (N.G.); (B.S.)
| | - Cécile Ortholan
- Radiotherapy Department, Princess Grace Hospital, 98000 Monaco, Monaco;
| | - Benjamin Serrano
- Radiophysics Department, Princess Grace Hospital, 98000 Monaco, Monaco; (R.V.); (R.A.); (N.G.); (B.S.)
| | - Vincent Picco
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Jacques Pouysségur
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
- CNRS, INSERM, Centre A. Lacassagne, Institute for Research on Cancer & Aging (IRCAN), University Côte d’Azur, 06107 Nice, France
| | - Milica Vucetic
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| | - Christopher Montemagno
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (C.G.); (F.S.); (W.E.); (J.D.); (T.B.); (V.V.); (M.P.-B.); (V.P.); (J.P.)
| |
Collapse
|
43
|
De Leon-Oliva D, Boaru DL, Minaya-Bravo AM, De Castro-Martinez P, Fraile-Martinez O, Garcia-Montero C, Cobo-Prieto D, Barrena-Blázquez S, Lopez-Gonzalez L, Albillos A, Alvarez-Mon M, Saez MA, Diaz-Pedrero R, Ortega MA. Improving understanding of ferroptosis: Molecular mechanisms, connection with cellular senescence and implications for aging. Heliyon 2024; 10:e39684. [PMID: 39553553 PMCID: PMC11564042 DOI: 10.1016/j.heliyon.2024.e39684] [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: 06/27/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
In the face of cell damage, cells can initiate a response ranging from survival to death, the balance being crucial for tissue homeostasis and overall health. Cell death, in both accidental and regulated forms, plays a fundamental role in maintaining tissue homeostasis. Among the regulated mechanisms of cell death, ferroptosis has garnered attention for its iron-dependent phospholipid (PL) peroxidation and its implications in aging and age-related disorders, as well as for its therapeutic relevance. In this review, we provide an overview of the mechanisms, regulation, and physiological and pathological roles of ferroptosis. We present new insights into the relationship between ferroptosis, cellular senescence and aging, emphasizing how alterations in ferroptosis pathways contribute to aging-related tissue dysfunction. In addition, we examine the therapeutic potential of ferroptosis in aging-related diseases, offering innovative insights into future interventions aimed at mitigating the effects of aging and promoting longevity.
Collapse
Affiliation(s)
- Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Ana M. Minaya-Bravo
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - David Cobo-Prieto
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Immune System Diseases-Rheumatology Service, Central University Hospital of Defence-UAH Madrid, 28801, Alcala de Henares, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala de Henares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Agustín Albillos
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Gastroenterology and Hepatology Service, Ramón y Cajal University Hospital, University of Alcalá, IRYCIS, Network Biomedical Research Center for Liver and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806, Alcala de Henares, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, 28801, Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala de Henares, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| |
Collapse
|
44
|
Zhou Z, Liao J, Wang Y, Zhou M. A ferroptosis-associated prognostic model correlated with immune landscape and radiotherapy response in low-grade gliomas (LGGs). J Neuroimmunol 2024; 396:578444. [PMID: 39357132 DOI: 10.1016/j.jneuroim.2024.578444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/11/2024] [Accepted: 09/01/2024] [Indexed: 10/04/2024]
Abstract
Despite receiving comprehensive treatment, the prognosis for low-grade gliomas (LGGs) patients varies considerably. Recent studies have focused extensively on ferroptosis, across a range of tumor types. Nevertheless, methodologies to evaluate the efficacy of radiotherapy for LGGs, from the perspective of ferroptosis-related genes (FRGs), remain strikingly rare. In this study, we conducted a retrospective study on the transcriptional profiles of LGG patients from the public databases and a local cohort. An FRG model was developed and validated, exhibits heightened robustness when contrasted with the traditional ssGSEA model. Patients demonstrating higher FRG scores were identified as a high-risk group, displaying a worse prognosis. By incorporating the FRG score alongside other prognosis-associated clinical indicators, we formulated an enhanced nomogram to achieve a higher level of prediction performance. Additionally, among LGG patients receiving radiotherapy, a poorer prognosis was observed in the high-risk group. Further investigation revealed that samples from the high-risk group generally exhibit a TME in an immuno-suppressive state. Collectively, we developed an FRG model and a robust nomogram for LGG prognostication. This study suggests that a high FRG score, indicative of an immunosuppressive TME, could potentially lead to a less favorable prognosis for certain LGG patients receiving radiotherapy.
Collapse
Affiliation(s)
- Zhaoming Zhou
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, China; Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jing Liao
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yinghui Wang
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Meijuan Zhou
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, China; Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China.
| |
Collapse
|
45
|
Scarmelotto A, Delprat V, Michiels C, Lucas S, Heuskin AC. The oxygen puzzle in FLASH radiotherapy: A comprehensive review and experimental outlook. Clin Transl Radiat Oncol 2024; 49:100860. [PMID: 39381632 PMCID: PMC11458961 DOI: 10.1016/j.ctro.2024.100860] [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: 05/21/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
FLASH radiotherapy is attracting increasing interest because it maintains tumor control while inflicting less damage to normal tissues compared to conventional radiotherapy. This sparing effect, the so-called FLASH effect, is achieved when radiation is delivered at ultra-high dose rates (≥40 Gy/s). Although the FLASH effect has already been demonstrated in several preclinical models, a complete mechanistic description explaining why tumors and normal tissues respond differently is still missing. None of the current hypotheses fully explains the experimental evidence. A common point between many of these is the role of oxygen, which is described as a major factor, either through transient hypoxia in the form of dissolved molecules, or reactive oxygen species (ROS). Therefore, this review focuses on both forms of this molecule, retracing old and more recent theories, while proposing new mechanisms that could provide a complete description of the FLASH effect based on preclinical and experimental evidence. In addition, this manuscript describes a set of experiments designed to provide the FLASH community with new tools for exploring the post-irradiation fate of ROS and their potential biological implications.
Collapse
Affiliation(s)
- Andrea Scarmelotto
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Victor Delprat
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Carine Michiels
- Unité de Recherche en Biologie Cellulaire (URBC), Namur Research Institute For Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Stéphane Lucas
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
- Ion Beam Application (IBA), Chemin du Cyclotron, 6, B-1348 Louvain-La-Neuve, Belgium
| | - Anne-Catherine Heuskin
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| |
Collapse
|
46
|
Nissi L, Tuominen S, Routila J, Huusko T, Ketonen P, Sundvall M, Leivo I, Irjala H, Minn H, Grönroos T, Ventelä S. xCT as a Predictor for Survival in a Population-Based Cohort of Head and Neck Squamous Cell Carcinoma. Cancer Med 2024; 13:e70371. [PMID: 39487701 PMCID: PMC11530867 DOI: 10.1002/cam4.70371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND xCT, also known as SLC7A11 (solute carrier Family 7 Member 11), is a cystine/glutamate antiporter protein that mediates regulated cell death and antioxidant defense. The aim of this study was to investigate the effect of xCT on the outcome of patients diagnosed with new head and neck squamous cell carcinoma (HNSCC). METHODS This retrospective cohort study utilized a population-based dataset, comprising all patients (n = 1033) diagnosed with new HNSCC during 2005-2015 in a population of 697,000 people. All patients (n = 585) with a tumor tissue sample available for immunohistochemical (IHC) staining were included. The follow-up rates were 97% and 81% at 3 and 5 years, respectively. Also, the specificity of the anti-xCT antibody was validated. RESULTS The expression level and prognostic significance of xCT were strongly dependent on tumor location. In oropharyngeal squamous cell carcinoma (OPSCC) patients, xCT expression was a significant prognostic factor for 5-year overall survival (OAS) (HR: 2.71; 95% CI 1.67-4.39; p < 0.001), disease-specific survival (DSS) (HR: 2.58; 95% CI 1.47-4.54; p = 0.001), and disease-free survival (DFS) (HR: 2.69; 95% CI 1.55-4.64; p < 0.001). Five-year survival rates for OPSCC patients with high and low levels of xCT were OAS 34% versus 62%; DSS 51% versus 73%; DFS 43% versus 73%, respectively. According to a multivariate model adjusted for age, T-class, nodal positivity, and tobacco consumption, xCT was an independent prognostic factor for 3-year survival, in which it outperformed p16 IHC. Similar associations were not observed in squamous cell carcinomas of oral cavity or larynx. Regarding treatment modalities, xCT was most predictive in HNSCC patients who received radiotherapy. CONCLUSIONS High xCT expression was associated with poor prognosis in OPSCC. Our findings suggest that joint analysis of xCT and p16 may add significant value in OPSCC treatment stratification.
Collapse
Affiliation(s)
- Linda Nissi
- Department of Oncology, and FICAN West Cancer CentreTurku University Hospital and University of TurkuTurkuFinland
| | - Sanni Tuominen
- Preclinical Imaging Turku PET CentreUniversity of TurkuTurkuFinland
- Medicity Research LaboratoryUniversity of TurkuTurkuFinland
| | - Johannes Routila
- Department of Otorhinolaryngology—Head and Neck SurgeryTurku University Hospital and University of TurkuTurkuFinland
| | - Teemu Huusko
- Department of Otorhinolaryngology—Head and Neck SurgeryTurku University Hospital and University of TurkuTurkuFinland
| | - Petra Ketonen
- Preclinical Imaging Turku PET CentreUniversity of TurkuTurkuFinland
- Medicity Research LaboratoryUniversity of TurkuTurkuFinland
| | - Maria Sundvall
- Department of Oncology, and FICAN West Cancer CentreTurku University Hospital and University of TurkuTurkuFinland
- Cancer Research UnitInstitute of BiomedicineUniversity of TurkuTurkuFinland
| | - Ilmo Leivo
- Department of PathologyTurku University Hospital and University of TurkuTurkuFinland
| | - Heikki Irjala
- Department of Otorhinolaryngology—Head and Neck SurgeryTurku University Hospital and University of TurkuTurkuFinland
| | - Heikki Minn
- Department of Oncology, and FICAN West Cancer CentreTurku University Hospital and University of TurkuTurkuFinland
| | - Tove J. Grönroos
- Department of Oncology, and FICAN West Cancer CentreTurku University Hospital and University of TurkuTurkuFinland
- Preclinical Imaging Turku PET CentreUniversity of TurkuTurkuFinland
- Medicity Research LaboratoryUniversity of TurkuTurkuFinland
| | - Sami Ventelä
- Department of Otorhinolaryngology—Head and Neck SurgeryTurku University Hospital and University of TurkuTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| |
Collapse
|
47
|
Wen L, Zhang W, Hu J, Chen T, Wang Y, Lv C, Li M, Wang L, Xiao F. Luteolin target HSPB1 regulates endothelial cell ferroptosis to protect against radiation vascular injury. PLoS One 2024; 19:e0311922. [PMID: 39392831 PMCID: PMC11469493 DOI: 10.1371/journal.pone.0311922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/26/2024] [Indexed: 10/13/2024] Open
Abstract
Vascular endothelial damage due to ionizing radiation is the main pathological process of radiation injury and the main cause of damage to various organs in nuclear accidents. Ferroptosis plays an important role in ionizing radiation-induced cell death. We have previously reported that luteolin is highly resistant to ferroptosis. In the present study, body weight, microvessel count, H&E, and Masson staining results showed that luteolin rescued radial vascular injury in vivo. Cell Counting Kit 8 (CCK8), Giemsa staining clarified the anti-ferroptosis ability of luteolin with low toxicity. Malondialdehyde (MDA), superoxide dismutase (SOD), NADP+/NADPH, Fe2+ staining, dihydroethidium (DHE) and MitoTracker assays for ferroptosis-related metrics, we found that luteolin enhances human umbilical vein endothelial cells (HUVECs) antioxidant damage capacity. Drug affinity responsive target stability (DARTS), surface plasmon resonance (SPR), computer simulated docking and western blot showed that heat shock protein beta-1 (HSPB1) is one of the targets of luteolin action. Luteolin inhibits ferroptosis by promoting the protein expression of HSPB1/solute carrier family 7 member 11 (SLC7A11)/ glutathione peroxidase 4 (GPX4). In conclusion, we have preliminarily elucidated the antioxidant damage ferroptosis ability and the target of action of luteolin to provide a theoretical basis for the application of luteolin in radiation injury diseases.
Collapse
Affiliation(s)
- Li Wen
- School of Nursing, Jilin University, Changchun, P. R. China
| | - Weiyuan Zhang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, P. R. China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, P. R. China
| | - Jia Hu
- Department of Cardiovascular, The Sixth Medical Center of Chinese PLA General Hospital, Haidian District, Beijing, China
| | - Tao Chen
- Department of Cardiovascular, The Sixth Medical Center of Chinese PLA General Hospital, Haidian District, Beijing, China
| | - Yiming Wang
- School of Nursing, Jilin University, Changchun, P. R. China
| | - Changchang Lv
- School of Nursing, Jilin University, Changchun, P. R. China
| | - Min Li
- Beijing Institute of Radiation Medicine, Beijing, P. R. China
| | - Lisheng Wang
- School of Nursing, Jilin University, Changchun, P. R. China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, P. R. China
| | - Fengjun Xiao
- Beijing Institute of Radiation Medicine, Beijing, P. R. China
| |
Collapse
|
48
|
Geronikolou S, Pavlopoulou A, Koutelekos I, Kalogirou D, Bacopoulou F, Cokkinos DV. Polycystic Ovary Syndrome and Ferroptosis: Following Ariadne's Thread. Biomedicines 2024; 12:2280. [PMID: 39457593 PMCID: PMC11505293 DOI: 10.3390/biomedicines12102280] [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: 08/03/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Recent literature suggests that ferroptosis (FPT) may be a key player in polycystic ovary syndrome (PCOS) pathogenesis, but the underlying mechanism(s) remain(s) unclear. Aim: Therefore, herein, we made an effort to reproduce the molecular signature of the syndrome by including FPT and exploring novel drug targets for PCOS. Methods: (a) Our previously constructed PCOS interactions molecular network was extended with the addition of FPT-associated genes (interaction score above 0.7) and (b) gene set enrichment analysis was performed so as to detect over-represented KEGG pathways. Results: The updated interactome includes 140 molecules, 20 of which are predicted/novel, with an interaction score of 7.3, and 12 major hubs. Moreover, we identified 16 over-represented KEGG pathways, with FPT being the most overexpressed pathway. The FPT subnetwork is connected with the PCOS network through KDM1A. Conclusions: FPT cell death is involved in PCOS development, as its major hub TP53 was shown to be the most important hub in the whole PCOS interactome, hence representing a prioritized drug target.
Collapse
Affiliation(s)
- Styliani Geronikolou
- Clinical, Translational and Experimental Surgery Research Center, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center (IBG), 35340 Izmir, Turkey;
- Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, 35340 Izmir, Turkey
| | - Ioannis Koutelekos
- Department of Nursing, School of Health and Care Sciences, University of West Attica, 12243 Athens, Greece;
| | - Dimitrios Kalogirou
- Department of Public and Community Health, School of Public Health, University of West Attica, 11521 Athens, Greece;
| | - Flora Bacopoulou
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dennis V. Cokkinos
- Clinical, Translational and Experimental Surgery Research Center, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
49
|
Han J, Zheng D, Liu PS, Wang S, Xie X. Peroxisomal homeostasis in metabolic diseases and its implication in ferroptosis. Cell Commun Signal 2024; 22:475. [PMID: 39367496 PMCID: PMC11451054 DOI: 10.1186/s12964-024-01862-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: 07/31/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024] Open
Abstract
Peroxisomes are dynamic organelles involved in various cellular processes, including lipid metabolism, redox homeostasis, and intracellular metabolite transfer. Accumulating evidence suggests that peroxisomal homeostasis plays a crucial role in human health and disease, particularly in metabolic disorders and ferroptosis. The abundance and function of peroxisomes are regulated by a complex interplay between biogenesis and degradation pathways, involving peroxins, membrane proteins, and pexophagy. Peroxisome-dependent lipid metabolism, especially the synthesis of ether-linked phospholipids, has been implicated in modulating cellular susceptibility to ferroptosis, a newly discovered form of iron-dependent cell death. This review discusses the current understanding of peroxisome homeostasis, its roles in redox regulation and lipid metabolism, and its implications in human diseases. We also summarize the main mechanisms of ferroptosis and highlight recent discoveries on how peroxisome-dependent metabolism and signaling influence ferroptosis sensitivity. A better understanding of the interplay between peroxisomal homeostasis and ferroptosis may provide new insights into disease pathogenesis and reveal novel therapeutic strategies for peroxisome-related metabolic disorders and ferroptosis-associated diseases.
Collapse
Affiliation(s)
- Jiwei Han
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China
| | - Daheng Zheng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China
| | - Pu-Ste Liu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Shanshan Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangdong, China
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang, China.
| |
Collapse
|
50
|
Cao PHA, Dominic A, Lujan FE, Senthilkumar S, Bhattacharya PK, Frigo DE, Subramani E. Unlocking ferroptosis in prostate cancer - the road to novel therapies and imaging markers. Nat Rev Urol 2024; 21:615-637. [PMID: 38627553 DOI: 10.1038/s41585-024-00869-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/19/2024]
Abstract
Ferroptosis is a distinct form of regulated cell death that is predominantly driven by the build-up of intracellular iron and lipid peroxides. Ferroptosis suppression is widely accepted to contribute to the pathogenesis of several tumours including prostate cancer. Results from some studies reported that prostate cancer cells can be highly susceptible to ferroptosis inducers, providing potential for an interesting new avenue of therapeutic intervention for advanced prostate cancer. In this Perspective, we describe novel molecular underpinnings and metabolic drivers of ferroptosis, analyse the functions and mechanisms of ferroptosis in tumours, and highlight prostate cancer-specific susceptibilities to ferroptosis by connecting ferroptosis pathways to the distinctive metabolic reprogramming of prostate cancer cells. Leveraging these novel mechanistic insights could provide innovative therapeutic opportunities in which ferroptosis induction augments the efficacy of currently available prostate cancer treatment regimens, pending the elimination of major bottlenecks for the clinical translation of these treatment combinations, such as the development of clinical-grade inhibitors of the anti-ferroptotic enzymes as well as non-invasive biomarkers of ferroptosis. These biomarkers could be exploited for diagnostic imaging and treatment decision-making.
Collapse
Affiliation(s)
- Pham Hong Anh Cao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Abishai Dominic
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fabiola Ester Lujan
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Sanjanaa Senthilkumar
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Mayo Clinic Alix School of Medicine, Rochester, MN, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Center for Nuclear Receptors and Cell Signalling, University of Houston, Houston, TX, USA.
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
| | - Elavarasan Subramani
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|