1
|
Yen CC, Chen PCH, Chen SC, Wu WC, Yen CH, Lin YC, Wu PK, Chen CM, Wang JY, Chao TC, Yang MH, Fletcher JA. Ferroptosis as a therapeutic vulnerability in MDM2 inhibition in dedifferentiated liposarcoma. Oncol Lett 2025; 29:269. [PMID: 40247991 PMCID: PMC12005077 DOI: 10.3892/ol.2025.15015] [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: 10/24/2024] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Ferroptosis is a form of necrotic cell death characterized by phospholipid oxidation. The cystine-glutamate antiporter (xCT), composed of solute carrier family 7 member 11 (SLC7A11) and SLC3A2, imports cystine for glutathione synthesis. Glutathione peroxidase 4 (GPX4) requires glutathione to counteract lipid peroxidation and prevent ferroptosis. Erastin, an xCT inhibitor, and Ras-selective lethal small molecule 3 (RSL3), a GPX4 inhibitor, suppress GPX4 function and induce ferroptosis. Tumor protein p53 (TP53) has a paradoxical role in ferroptosis regulation. Mouse double minute 2 homolog (MDM2), a negative regulator of TP53, is a key oncogene in well-differentiated liposarcoma (WDLPS) and dedifferentiated liposarcoma (DDLPS). Therefore, the present study explored the role of ferroptosis in DDLPS treatment response and resistance. Publicly available expression profiles of WDLPS, DDLPS and adipose tissue were analyzed, and the differential expression of ferroptosis-related genes regulated by the MDM2-TP53 pathway was identified in WDLPS and DDLPS. In vitro experiments were performed to assess the effects of erastin and RSL3 on the viability, lipid peroxidation and apoptosis of DDLPS cell lines. The results revealed that erastin and RSL3 induced lipid peroxidation and apoptosis, thereby exerting cytotoxic effects. In addition, nutlin-3, an MDM2 inhibitor, was demonstrated to increase lipid peroxidation and cytotoxicity when applied prior to erastin treatment. Notably, nutlin-3 also upregulated SLC3A2 expression in DDLPS cell lines, thereby enhancing cystine uptake. This increase in cystine uptake was suppressed by erastin. In addition, nutlin-3-induced SLC3A2 upregulation was abolished by TP53 knockdown. Nutlin-3 combined with erastin or RSL3 reduced absolute p-4EBP-1 levels in NDDLS-1 cells and p-p70S6 levels in both cell lines, with no significant impact on the p-4EBP-1/4EBP-1 and p-p70S6/p70S6 ratios. These results indicate that ferroptosis is a therapeutic vulnerability in the response to MDM2 inhibition in DDLPS. Furthermore, combining MDM2 inhibitors with ferroptosis-inducing agents may provide a potential therapeutic strategy for DDLPS and the role of mTOR in the pro-apoptotic effect of these combinations deserve further investigation.
Collapse
Affiliation(s)
- Chueh-Chuan Yen
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Paul Chih-Hsueh Chen
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - San-Chi Chen
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Wen-Chi Wu
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Chiao-Han Yen
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Yung-Chan Lin
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Po-Kuei Wu
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Chao-Ming Chen
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Jir-You Wang
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Ta-Chung Chao
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Muh-Hwa Yang
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | | |
Collapse
|
2
|
Yan W, Xiang S, Feng J, Zu X. Role of ubiquitin-specific proteases in programmed cell death of breast cancer cells. Genes Dis 2025; 12:101341. [PMID: 40083330 PMCID: PMC11904532 DOI: 10.1016/j.gendis.2024.101341] [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: 01/20/2024] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 03/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignant tumor and the leading cause of cancer-related deaths among women worldwide. Great progress has been recently achieved in controlling breast cancer; however, mortality from breast cancer remains a substantial challenge, and new treatment mechanisms are being actively sought. Programmed cell death (PCD) is associated with the progression and treatment of many types of human cancers. PCD can be divided into multiple pathways including autophagy, apoptosis, mitotic catastrophe, necroptosis, ferroptosis, pyroptosis, and anoikis. Ubiquitination is a post-translational modification process in which ubiquitin, a 76-amino acid protein, is coupled to the lysine residues of other proteins. Ubiquitination is involved in many physiological events and promotes cancer development and progression. This review elaborates the role of ubiquitin-specific protease (USP) in programmed cell death, which is common in breast cancer cells, and lays the foundation for tumor diagnosis and targeted therapy.
Collapse
Affiliation(s)
| | | | - Jianbo Feng
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| | - Xuyu Zu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001 Hunan, China
| |
Collapse
|
3
|
Alarcón-Veleiro C, López-Calvo I, Berjawi L, Lucio-Gallego S, Mato-Basalo R, Quindos-Varela M, Lesta-Mellid R, Santamarina-Caínzos I, Varela-Rodríguez S, Fraga M, Quintela M, Vizoso-Vázquez A, Arufe MC, Fafián-Labora J. Ferroptosis: An emerging strategy for managing epithelial ovarian cancer. Biomed Pharmacother 2025; 187:118065. [PMID: 40306179 DOI: 10.1016/j.biopha.2025.118065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/30/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
Ferroptosis is a regulated form of cell death characterised by iron-dependent lipid peroxidation, a process intricately linked to cellular redox homeostasis. This form of cell death is induced by the accumulation of intracellular iron and the subsequent generation of reactive oxygen species (ROS), which leads to lipid peroxidation and ultimately cell death. Ferroptosis is distinct from traditional forms of cell death, such as apoptosis, and holds significant therapeutic potential, particularly in cancers harboring rat sarcoma virus (RAS) mutations, such as epithelial ovarian cancer (EOC). EOC is notoriously resistant to conventional therapies and is associated with a poor prognosis. In this review, we examine recent progress in the understanding of ferroptosis, with a particular focus on its redox biology and the complex regulatory networks involved. We also propose a novel classification system for ferroptosis modulators, grouping them into six categories (I, II, III, IV, V and VI) based on their mechanisms of action and their roles in modulating cellular redox status. By refining these categories, we aim to provide deeper insights into the role of ferroptosis in cancer biology, especially in EOC, and to identify potential therapeutic avenues. We propose that further investigation of ferroptosis in the context of redox biology could reveal novel biomarkers and therapeutic targets, offering promising strategies to overcome resistance mechanisms and improve clinical outcomes for patients with EOC and other treatment-resistant cancers.
Collapse
Affiliation(s)
- C Alarcón-Veleiro
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - I López-Calvo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain; Centro Interdisciplinar de Química de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Rúa as Xubias 84, A Coruña 15006, Spain
| | - L Berjawi
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - S Lucio-Gallego
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - R Mato-Basalo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - M Quindos-Varela
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - R Lesta-Mellid
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - I Santamarina-Caínzos
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - S Varela-Rodríguez
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - M Fraga
- Department of Anatomical Pathology, University Hospital Complex A Coruña, As Xubias 84, A Coruña 15006, Spain
| | - M Quintela
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - A Vizoso-Vázquez
- Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain
| | - M C Arufe
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| | - J Fafián-Labora
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| |
Collapse
|
4
|
Cao P, Jaeschke H, Ni HM, Ding WX. The Ways to Die: Cell Death in Liver Pathophysiology. Semin Liver Dis 2025. [PMID: 40199509 DOI: 10.1055/a-2576-4332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Liver diseases are closely associated with various cell death mechanisms, including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis. Each process contributes uniquely to the pathophysiology of liver injury and repair. Importantly, these mechanisms are not limited to hepatocytes; they also significantly involve nonparenchymal cells. This review examines the molecular pathways and regulatory mechanisms underlying these forms of cell death in hepatocytes, emphasizing their roles in several liver diseases, such as ischemia-reperfusion injury, metabolic dysfunction-associated steatotic liver disease, drug-induced liver injury, and alcohol-associated liver disease. Recent insights into ferroptosis and pyroptosis may reveal novel therapeutic targets for managing liver diseases. This review aims to provide a comprehensive overview of these cell death mechanisms in the context of liver diseases, detailing their molecular signaling pathways and implications for potential treatment strategies.
Collapse
Affiliation(s)
- Peng Cao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
- Division of Gastroenterology, Hepatology and Mobility, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
5
|
Hong Y, He J, Deng D, Liu Q, Zu X, Shen Y. Targeting kinases that regulate programmed cell death: a new therapeutic strategy for breast cancer. J Transl Med 2025; 23:439. [PMID: 40229646 PMCID: PMC11995514 DOI: 10.1186/s12967-025-06367-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/08/2025] [Indexed: 04/16/2025] Open
Abstract
Breast cancer is one of the most prevalent malignant tumors among women and ranks as the second leading cause of cancer-related deaths in females, primarily due to delays in diagnosis and shortcomings in treatment strategies. Consequently, there is a pressing need to identify reliable therapeutic targets and strategies. In recent years, the identification of effective biomarkers-particularly novel molecular therapeutic targets-has become a focal point in breast cancer research, aimed at predicting disease aggressiveness and monitoring treatment responses. Simultaneously, advancements in understanding the molecular mechanisms underlying cellular programmed death have opened new avenues for targeting kinase-regulated programmed cell death as a viable therapeutic strategy. This review summarizes the latest research progress regarding kinase-regulated programmed death (including apoptosis, pyroptosis, autophagy, necroptosis, and ferroptosis) in breast cancer treatment. It covers the key kinases involved in this mechanism, their roles in the onset and progression of breast cancer, and strategies for modulating these kinases through pharmacological interventions.
Collapse
Affiliation(s)
- Yun Hong
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-Incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, China
| | - Dan Deng
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-Incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qinyue Liu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hunan Province Clinical Research Center for Accurate Diagnosis and Treatment of High-Incidence Sexually Transmitted Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
- Hunan Provincial Clinical Medical Research Center for Drug Evaluation of major chronic diseases, Hengyang, China.
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
- Hunan Provincial Clinical Medical Research Center for Drug Evaluation of major chronic diseases, Hengyang, China.
| |
Collapse
|
6
|
Wang T, Sun N, Ma Y, Zhang S. Recent Advances in the Development of Sigma Receptor (Radio)Ligands and Their Application in Tumors. ACS Pharmacol Transl Sci 2025; 8:951-977. [PMID: 40242588 PMCID: PMC11997895 DOI: 10.1021/acsptsci.4c00711] [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: 12/06/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 04/18/2025]
Abstract
Cancer ranks among the top triumvirate leading causes of human deaths worldwide. The pathological mechanisms are notably intricate, demonstrating proliferative and metastatic capabilities, which complicate therapeutic interventions. The sigma-1 receptor (σ1R) plays a crucial role in tumor survival and migration, while the sigma-2 receptor (σ2R) is intimately associated with tumor proliferation. This review encapsulated the investigation concerning σ1R and σ2R in neoplasms and rigorously summarized the ligands and radio-ligands development and their tumor applications, such as antitumor cell proliferation and PET/SPECT imaging in tumors. A comprehensive classification discussion was undertaken regarding the chemical structures and emphasized the possibility of dual/multitargeted ligands. Ultimately, we discussed the effects of chiral structures and the pharmacological characteristics of ligands on affinity and pharmacokinetic features in vivo, particularly concerning radiopharmaceuticals. This review functions as a beneficial resource, fostering ligand deployment and stimulating the generation of innovative ideas for developing innovative radiopharmaceuticals.
Collapse
Affiliation(s)
- Tao Wang
- Department
of Nuclear Medicine, Xinqiao Hospital, Army
Medical University, Chongqing 400037, China
- School
of Medical Imaging, North Sichuan Medical
College, NanChong 637100, China
- Department
of Nuclear Medicine, Affiliated Hospital
of North Sichuan Medical College, North Sichuan Medical College, NanChong 637000, China
| | - Na Sun
- Department
of Nuclear Medicine, Xinqiao Hospital, Army
Medical University, Chongqing 400037, China
| | - Yanxi Ma
- Department
of Nuclear Medicine, Xinqiao Hospital, Army
Medical University, Chongqing 400037, China
| | - Song Zhang
- Department
of Nuclear Medicine, Xinqiao Hospital, Army
Medical University, Chongqing 400037, China
| |
Collapse
|
7
|
Song W, Sun P, Zhao T, Zang Y, Dong P, Tang Q, Chen W, Chen W, Wang Z, Zhang Q, Wang Y, Yin C, Yu M. Unveiling the therapeutic potential of ferroptosis in lung cancer: a comprehensive bibliometric analysis and future therapeutic insights. Discov Oncol 2025; 16:508. [PMID: 40208519 PMCID: PMC11985706 DOI: 10.1007/s12672-025-02234-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Lung cancer remains the leading cause of cancer-related deaths worldwide, with increasing attention being given to novel therapeutic strategies that target the mechanisms underlying tumor growth and drug resistance. Among these, ferroptosis, a regulated cell death driven by iron-dependent lipid peroxidation, has become a key focus in cancer research. Despite extensive research, the exact role of ferroptosis in lung cancer progression and treatment remains unclear, especially regarding its interaction with immune cells and the tumor microenvironment. OBJECTIVE AND METHODS To address these limitations, this study utilizes a comprehensive bibliometric analysis to explore the current landscape of ferroptosis research in lung cancer. We collected data from the Web of Science Core Collection, covering articles published between 2015 and 2025, and analyzed them using advanced tools such as VOSviewer and CiteSpace. RESULTS This study uses a comprehensive bibliometric analysis to uncover key trends and emerging areas related to lung cancer in ferroptosis research. Recently, the focus has shifted from basic mechanisms to clinical applications, particularly in developing GPX4-targeted therapies and combination treatments. With increasing international collaboration, the United States and China have become key players. Interdisciplinary research, especially on ferroptosis and the cancer-immune system, offers new insights into its role in the tumor microenvironment and immunotherapy. Ferroptosis shows excellent promise in overcoming drug resistance by regulating iron-dependent lipid peroxidation and enhancing treatment efficacy. Future research should focus on ferroptosis' clinical translation, particularly in personalized medicine and overcoming resistance, offering broad prospects for lung cancer treatment. CONCLUSION This paper provides valuable insights into the trends, key contributors, and emerging frontiers of ferroptosis research in lung cancer. It identifies important developments that can serve as a foundation for translating ferroptosis-based therapies into clinical practice, particularly to address drug resistance in lung cancer.
Collapse
Affiliation(s)
- Wenhuan Song
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
- Binzhou Medical University, Binzhou, China
| | - Peipei Sun
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Tongzhen Zhao
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Yunxue Zang
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Pengpeng Dong
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Qi Tang
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Wenyu Chen
- Binzhou Medical University, Binzhou, China
| | - Wenyi Chen
- Binzhou Medical University, Binzhou, China
| | | | | | | | - Chunhui Yin
- Shandong Academy of Medical Sciences (SDAMS), Jinan, China
| | - Mingkun Yu
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China.
- Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
8
|
Zhang Y, Su F, Zhu E, Sun Y, Kuang H, Wang Q. A systematical review on traditional Chinese medicine treating chronic diseases via regulating ferroptosis from the perspective of experimental evidence and clinical application. CHINESE HERBAL MEDICINES 2025; 17:246-260. [PMID: 40256717 PMCID: PMC12009076 DOI: 10.1016/j.chmed.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/12/2024] [Accepted: 01/20/2025] [Indexed: 04/22/2025] Open
Abstract
Ferroptosis is a unique regulated form of cell death that is distinct from apoptosis, necrosis, and other well-characterized regulated cell death types, and plays an important role in the occurrence and development of chronic metabolic diseases, including diabetes, hypertension, hyperlipidemia, and non-alcoholic steatohepatitis. Recently, increasing evidence has supported traditional Chinese medicine (TCM) as a new hot spot for the treatment of chronic metabolic diseases by mediating ferroptosis. Unfortunately, few systematic reviews have described the importance of TCM in treating chronic metabolic diseases through the ferroptosis pathway. In the current review, the mechanism of ferroptosis and the roles of ferroptosis in chronic metabolic diseases are summarized. Additionally, this review illustrates that the regulation of ferroptosis by TCM could be an effective approach for treating chronic metabolic diseases based on experimental evidence and clinical application. In summary, this work will improve the understanding of ferroptosis and the ability of TCM to regulate ferroptosis in chronic metabolic diseases, thereby promoting the development and application of natural TCM.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Fazhi Su
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Enlin Zhu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Qiuhong Wang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 511400, China
| |
Collapse
|
9
|
Lan J, Cai D, Gou S, Bai Y, Lei H, Li Y, Chen Y, Zhao Y, Shen J, Wu X, Li M, Chen M, Li X, Sun Y, Gu L, Li W, Wang F, Cho CH, Zhang Y, Zheng X, Xiao Z, Du F. The dynamic role of ferroptosis in cancer immunoediting: Implications for immunotherapy. Pharmacol Res 2025; 214:107674. [PMID: 40020885 DOI: 10.1016/j.phrs.2025.107674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Currently, cancer immunotherapy strategies are primarily formulated based on the patient's present condition, representing a "static" treatment approach. However, cancer progression is inherently "dynamic," as the immune environment is not fixed but undergoes continuous changes. This dynamism is characterized by the ongoing interactions between tumor cells and immune cells, which ultimately lead to alterations in the tumor immune microenvironment. This process can be effectively elucidated by the concept of cancer immunoediting, which divides tumor development into three phases: "elimination," "equilibrium," and "escape." Consequently, adjusting immunotherapy regimens based on these distinct phases may enhance patient survival and improve prognosis. Targeting ferroptosis is an emerging area in cancer immunotherapy, and our findings reveal that the antioxidant systems associated with ferroptosis possess dual roles, functioning differently across the three phases of cancer immunoediting. Therefore, this review delve into the dual role of the ferroptosis antioxidant system in tumor development and progression. It also propose immunotherapy strategies targeting ferroptosis at different stages, ultimately aiming to illuminate the significant implications of targeting ferroptosis at various phases for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarui Lan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Dan Cai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Shuang Gou
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Yulin Bai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Huaqing Lei
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yan Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Zhang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China
| | - Xin Zheng
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China.
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| |
Collapse
|
10
|
Liang Y, Zhao Y, Qi Z, Li X, Zhao Y. Ferroptosis: CD8 +T cells' blade to destroy tumor cells or poison for self-destruction. Cell Death Discov 2025; 11:128. [PMID: 40169575 PMCID: PMC11962101 DOI: 10.1038/s41420-025-02415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
Ferroptosis represents an emerging, iron-dependent form of cell death driven by lipid peroxidation. In recent years, it has garnered significant attention in the realm of cancer immunotherapy, particularly in studies involving immune checkpoint inhibitors. This form of cell death not only enhances our comprehension of the tumor microenvironment but is also considered a promising therapeutic strategy to address tumor resistance, investigate immune activation mechanisms, and facilitate the development of cancer vaccines. The combination of immunotherapy with ferroptosis provides innovative targets and fresh perspectives for advancing cancer treatment. Nevertheless, tumor cells appear to possess a wider array of ferroptosis evasion strategies compared to CD8+T cells, which have been conclusively shown to be more vulnerable to ferroptosis. Furthermore, ferroptosis in the TME can create a favorable environment for tumor survival and invasion. Under this premise, both inducing tumor cell ferroptosis and inhibiting T cell ferroptosis will impact antitumor immunity to some extent, and even make the final result run counter to our therapeutic purpose. This paper systematically elucidates the dual-edged sword role of ferroptosis in the antitumor process of T cells, briefly outlining the complexity of ferroptosis within the TME. It explores potential side effects associated with ferroptosis-inducing therapies and critically considers the combined application of ferroptosis-based therapies with ICIs. Furthermore, it highlights the current challenges faced by this combined therapeutic approach and points out future directions for development.
Collapse
Affiliation(s)
- Yuan Liang
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhaoyang Qi
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinru Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
11
|
Zhu L, Hu J, Wu X, Zhang J, Xu X, Huang X, Tian B, Zhao CX, Du Y, Wu L. Programmed enhancement of endogenous iron-mediated lysosomal membrane permeabilization for tumor ferroptosis/pyroptosis dual-induction. Nat Commun 2025; 16:3017. [PMID: 40148335 PMCID: PMC11950380 DOI: 10.1038/s41467-025-58124-7] [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: 09/11/2024] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Ferroptosis and pyroptosis, as emerging regulated forms of cell death capable of overcoming apoptotic resistance, demonstrate promising potential in tumor therapy. Given that iron manipulation and reactive oxygen species elevation serve as common stimuli for both processes, inducing lysosomal membrane permeabilization (LMP) with ensuing release of lysosomal contents (including iron ions and cathepsins) is anticipated to realize dual induction of ferroptosis/pyroptosis. Herein, we report a folic acid and croconaine molecule-functionalized upconversion nanoparticle (UCNP-Cro/FA) that is able to mobilize intracellular stores of endogenous iron and spatiotemporally control the lysosome-intrinsic Fenton chemistry, thereby triggering LMP-associated cell death. The process of endogenous iron mobilization occurs through two key steps: Cro-mediated coordination of abundant Fe3+ ions within lysosomes, followed by UV-emitting upconversion core-mediated photoreduction, resulting in Fe2+ ions release. Both in vitro and in vivo experiments show that UCNP-Cro/FA + NIR treatment effectively boost LMP by endogenous iron-mediated •OH production, ultimately triggering irreversible tumor cell death via ferroptosis and Caspase-1/GSDMD-dependent pyroptosis pathways. Moreover, this process potentiates tumor immunogenicity, holding promise for tumor immunotherapy. Overall, this work proposes a feasible tumor therapy strategy that integrates ferroptosis and pyroptosis through the efficient application and activation of endogenous iron.
Collapse
Affiliation(s)
- Luwen Zhu
- Department of General Surgery, Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, P. R. China
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jiahao Hu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xiaochuan Wu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jucong Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xinyi Xu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xiajie Huang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Bing Tian
- Institute of Biophysics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China.
| | - Chun-Xia Zhao
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA, Australia.
| | - Yongzhong Du
- Department of General Surgery, Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, P. R. China.
- State Key Laboratory of Advanced Drug Delivery and Release Systems, Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, P. R. China.
| | - Liming Wu
- Department of General Surgery, Center for Metabolism Research, The Fourth Affiliated Hospital of School of Medicine and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, P. R. China.
| |
Collapse
|
12
|
Tan L, He G, Shen C, He S, Chen Y, Guo X. Construction of a ferroptosis-based prediction model for the prognosis of MYCN-amplified neuroblastoma and screening and verification of target sites. Hereditas 2025; 162:41. [PMID: 40108662 PMCID: PMC11921587 DOI: 10.1186/s41065-025-00413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Neuroblastoma (NB) is a prevalent extracranial solid tumor in pediatric patients. Of these, the MYCN-amplified type has a poor treatment response and prognosis. To enhance therapeutic efficacy and prognostic outcomes, numerous research teams have undertaken extensive investigations through various pathways and directions. Among these, ferroptosis has recently emerged as a significant area of research focus.Ferroptosis, a type of iron-dependent cell death, is primarily caused by lipid peroxides. This study intends to develop a prognosis model based on MYCN-amplified NB and ferroptosis-related genes (FGs). METHODS Data for this study were sourced from the TARGET and FerrDb databases. Lasso regression algorithms and univariate COX analysis were leveraged to determine feature genes; multivariate COX analysis was employed to develop a prediction model and risk scores; and receiver operating characteristic (ROC) curves and Kaplan-Meier analysis were utilized to assess the predictive ability of the model. Furthermore, discrepancies in immune cell infiltration (ICI) between the high-risk (HR) and low-risk (LR) populations were assessed via CIBERSORT analysis. Finally, experiments were conducted on MYCN-amplified and MYCN non-amplified cells so as to validate the differential expression of the gene. RESULTS A prediction model was constructed and risk scores were calculated based on 4 genes (LIFR, TP53, NRAS, and OSBPL9). The HR group, which was stratified by the median score, had a lower overall survival rate than the LR group.The differences in expression of each gene between MYCN-amplified and MYCN non-amplified cells were further confirmed through cell experiments and qPCR. CONCLUSION The prediction model in this study can be employed to forecast the prognosis of MYCN-amplified NB. These genes may represent promising new ferroptosis-related intervention targets (FITs) in treating MYCN-amplified NB, with the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Linjun Tan
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China
| | - Guoqian He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Chengqi Shen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Sijia He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China
| | - Yan Chen
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
- Department of Pediatrics, Guizhou Children's Hospital, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
- Collaborative Innovation Center for Tissue Injury Repairand Regenerative Medicine of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
| | - Xia Guo
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, No.149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, China.
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Section 3, South Renmin Road, Chengdu, 610041, China.
| |
Collapse
|
13
|
Qiu Y, Su Y, Sai W, Feng G. Research progress on ferroptosis in head and neck squamous cell carcinoma. J Mol Histol 2025; 56:109. [PMID: 40095205 DOI: 10.1007/s10735-025-10381-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/20/2025] [Indexed: 03/19/2025]
Abstract
Ferroptosis, a regulated iron-dependent cell death pathway driven by lipid peroxidation and mitochondrial dysfunction, has emerged as a critical player in diseases characterized by dysregulated iron metabolism and redox imbalance. In recent years, its therapeutic potential has garnered significant attention in head and neck squamous cell carcinoma (HNSCC), a malignancy notorious for its high incidence, frequent recurrence, and poor prognosis. This review systematically delineates the molecular underpinnings of ferroptosis in HNSCC pathogenesis and therapy, focusing on four interconnected axes: (1) iron homeostasis disruption, exemplified by dysregulation of the iron efflux channel ferroportin (FPN); (2) lipid peroxidation dynamics, mediated through key regulators such as SLC7A11; (3) mitochondrial remodeling, including structural and functional alterations during ferroptosis execution; and (4) critical signaling cascades, notably the PI3K-AKT-mTOR pathway, which orchestrates cellular survival and death decisions. Therapeutic exploration has identified ferroptosis inducers (e.g., erastin) as promising agents to disrupt redox equilibrium in HNSCC cells, while pharmacological inhibitors offer potential for mitigating off-target toxicity. Notably, combination strategies integrating ferroptosis modulation with conventional therapies or other programmed cell death mechanisms demonstrate synergistic efficacy, highlighting a paradigm shift in precision oncology. This study aims to provide a mechanistic framework for ferroptosis in HNSCC, bridging preclinical insights with translational opportunities. By elucidating context-dependent regulatory networks and optimizing therapeutic targeting, we propose novel strategies to overcome treatment resistance, ultimately improving clinical outcomes and quality of life for HNSCC patients.
Collapse
Affiliation(s)
- Yi Qiu
- Nantong University Medical College, Nantong, 226001, China
| | - Yuyuan Su
- Nantong University Medical College, Nantong, 226001, China
| | - Wenli Sai
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, West Temple Rd.20, Nantong, 226001, China.
| | - Guijuan Feng
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, 226001, China.
| |
Collapse
|
14
|
Guo W, Duan Z, Wu J, Zhou BP. Epithelial-mesenchymal transition promotes metabolic reprogramming to suppress ferroptosis. Semin Cancer Biol 2025; 112:20-35. [PMID: 40058616 DOI: 10.1016/j.semcancer.2025.02.013] [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: 08/10/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular de-differentiation process that provides cells with the increased plasticity and stem cell-like traits required during embryonic development, tissue remodeling, wound healing and metastasis. Morphologically, EMT confers tumor cells with fibroblast-like properties that lead to the rearrangement of cytoskeleton (loss of stiffness) and decrease of membrane rigidity by incorporating high level of poly-unsaturated fatty acids (PUFA) in their phospholipid membrane. Although large amounts of PUFA in membrane reduces rigidity and offers capabilities for tumor cells with the unbridled ability to stretch, bend and twist in metastasis, these PUFA are highly susceptible to lipid peroxidation, which leads to the breakdown of membrane integrity and, ultimately results in ferroptosis. To escape the ferroptotic risk, EMT also triggers the rewiring of metabolic program, particularly in lipid metabolism, to enforce the epigenetic regulation of EMT and mitigate the potential damages from ferroptosis. Thus, the interplay among EMT, lipid metabolism, and ferroptosis highlights a new layer of intricated regulation in cancer biology and metastasis. Here we summarize the latest findings and discuss these mutual interactions. Finally, we provide perspectives of how these interplays contribute to cellular plasticity and ferroptosis resistance in metastatic tumor cells that can be explored for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Wenzheng Guo
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Zhibing Duan
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Jingjing Wu
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Binhua P Zhou
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
15
|
Wan M, Pan S, Shan B, Diao H, Jin H, Wang Z, Wang W, Han S, Liu W, He J, Zheng Z, Pan Y, Han X, Zhang J. Lipid metabolic reprograming: the unsung hero in breast cancer progression and tumor microenvironment. Mol Cancer 2025; 24:61. [PMID: 40025508 PMCID: PMC11874147 DOI: 10.1186/s12943-025-02258-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Aberrant lipid metabolism is a well-recognized hallmark of cancer. Notably, breast cancer (BC) arises from a lipid-rich microenvironment and depends significantly on lipid metabolic reprogramming to fulfill its developmental requirements. In this review, we revisit the pivotal role of lipid metabolism in BC, underscoring its impact on the progression and tumor microenvironment. Firstly, we delineate the overall landscape of lipid metabolism in BC, highlighting its roles in tumor progression and patient prognosis. Given that lipids can also act as signaling molecules, we next describe the lipid signaling exchanges between BC cells and other cellular components in the tumor microenvironment. Additionally, we summarize the therapeutic potential of targeting lipid metabolism from the aspects of lipid metabolism processes, lipid-related transcription factors and immunotherapy in BC. Finally, we discuss the possibilities and problems associated with clinical applications of lipid‑targeted therapy in BC, and propose new research directions with advances in spatiotemporal multi-omics.
Collapse
Affiliation(s)
- Mengting Wan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Haizhou Diao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Ziqi Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Shuya Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wan Liu
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaying He
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Graduate School of Bengbu Medical University, Bengbu, Anhui Province, China
| | - Zihan Zheng
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
16
|
Qi X, Zhou J, Wang X, Shen Y, Cao Y, Jiang L, Shen M, Zhang H, Wang T, Wei P, Xu R, Yang Y, Ding X, Wang C, Jia X, Yan Q, Li W, Lu C. HPV E6/E7-Induced Acetylation of a Peptide Encoded by a Long Non-Coding RNA Inhibits Ferroptosis to Promote the Malignancy of Cervical Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414018. [PMID: 39836502 PMCID: PMC11905060 DOI: 10.1002/advs.202414018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/05/2025] [Indexed: 01/23/2025]
Abstract
Although a fraction of functional peptides concealed within long non-coding RNAs (lncRNAs) is identified, it remains unclear whether lncRNA-encoded peptides are involved in the malignancy of cervical cancer (CC). Here, a 92-amino acid peptide is discovered, which is named TUBORF, encoded by lncRNA TUBA3FP and highly expressed in CC tissues. TUBORF inhibits ferroptosis to promote the malignant proliferation of CC cells. Mechanistically, human papillomavirus (HPV) oncogenes E6 and E7 upregulate TUBORF through CREB-binding protein (CBP)/E1A-binding protein p300 (p300)-mediated histone H3 lysine 27 acetylation (H3K27ac) of lncTUBA3FP enhancer. Furthermore, E6 and E7 elevate and recruit acetyltransferase establishment of sister chromatid cohesion N-acetyltransferase 1 (ESCO1) to bind to and acetylate TUBORF, which facilitates the degradation of immunity-related GTPase Q (IRGQ) via a ubiquitin-proteasome pathway, resulting in the inhibition of ferroptosis and promotion of the malignant proliferation of CC cells. Importantly, silencing ESCO1 or TURORF amplifies anticancer effects by paclitaxel both in CC cells and in vivo. These novel findings reveal oncopeptide TUBORF and its acetyltransferase ESCO1 as important regulators of ferroptosis and tumorigenesis during cervical cancer pathogenesis and establish the scientific basis for targeting these molecules for treating CC.
Collapse
Affiliation(s)
- Xiaoyu Qi
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Jing Zhou
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Xinyue Wang
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yan Shen
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yuxun Cao
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Liangzi Jiang
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Miaomiao Shen
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Haoran Zhang
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Tianjiao Wang
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Pengjun Wei
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Ruoqi Xu
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yue Yang
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Xiangya Ding
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Cong Wang
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China
| | - Qin Yan
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Wan Li
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Infectious Diseases, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Chun Lu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
- Department of Infectious Diseases, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, 211166, P. R. China
| |
Collapse
|
17
|
Consoli V, Sorrenti V, Gulisano M, Spampinato M, Vanella L. Navigating heme pathways: the breach of heme oxygenase and hemin in breast cancer. Mol Cell Biochem 2025; 480:1495-1518. [PMID: 39287890 PMCID: PMC11842487 DOI: 10.1007/s11010-024-05119-5] [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: 08/06/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024]
Abstract
Breast cancer remains a significant global health challenge, with diverse subtypes and complex molecular mechanisms underlying its development and progression. This review comprehensively examines recent advances in breast cancer research, with a focus on classification, molecular pathways, and the role of heme oxygenases (HO), heme metabolism implications, and therapeutic innovations. The classification of breast cancer subtypes based on molecular profiling has significantly improved diagnosis and treatment strategies, allowing for tailored approaches to patient care. Molecular studies have elucidated key signaling pathways and biomarkers implicated in breast cancer pathogenesis, shedding light on potential targets for therapeutic intervention. Notably, emerging evidence suggests a critical role for heme oxygenases, particularly HO-1, in breast cancer progression and therapeutic resistance, highlighting the importance of understanding heme metabolism in cancer biology. Furthermore, this review highlights recent advances in breast cancer therapy, including targeted therapies, immunotherapy, and novel drug delivery systems. Understanding the complex interplay between breast cancer subtypes, molecular pathways, and innovative therapeutic approaches is essential for improving patient outcomes and developing more effective treatment strategies in the fight against breast cancer.
Collapse
Affiliation(s)
- Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy
| | - Maria Gulisano
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Mariarita Spampinato
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy.
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy.
| |
Collapse
|
18
|
Xie Y, Tao Z, Wang B, Zhao Y, Chen X, Li B, Wang J, Chen G, Hu X. Ferroptosis-related genes as prognostic markers for survival and immunotherapy in triple-negative breast cancer: analysis of public databases and a single institution. Ther Adv Med Oncol 2025; 17:17588359251322291. [PMID: 40034604 PMCID: PMC11873862 DOI: 10.1177/17588359251322291] [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: 10/25/2024] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
Background Ferroptosis plays a vital role in cancer development and treatment. The relationship between ferroptosis-related genes and breast cancer prognosis, as well as immunotherapy outcomes, remains unknown. Objectives To evaluate the prognostic value of ferroptosis-related genes in breast cancer. Methods We conducted differential expressions and prognostic analysis for ferroptosis-related genes on public databases and breast cancer patients in our center and analyzed their predictive value for immunotherapy of breast cancer patients. Results We identified prognostic ferroptosis-related genes, constructed a nomogram, and validated key genes using patient data from our center. We also investigated ferroptosis-related genes significantly associated with immune infiltration and identified FTH1 as a promising biomarker for triple-negative breast cancer immunotherapy. Conclusion Ferroptosis-related genes had potential prognostic value and predictive value for breast cancer immunotherapy.
Collapse
Affiliation(s)
- Yizhao Xie
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhonghua Tao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Biyun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yannan Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyu Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinyan Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guangliang Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Luo JY, Deng YL, Lu SY, Chen SY, He RQ, Qin DY, Chi BT, Chen G, Yang X, Peng W. Current Status and Future Directions of Ferroptosis Research in Breast Cancer: Bibliometric Analysis. Interact J Med Res 2025; 14:e66286. [PMID: 40009842 PMCID: PMC11904379 DOI: 10.2196/66286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Ferroptosis, as a novel modality of cell death, holds significant potential in elucidating the pathogenesis and advancing therapeutic strategies for breast cancer. OBJECTIVE This study aims to comprehensively analyze current ferroptosis research and future trends, guiding breast cancer research advancements and innovative treatment strategies. METHODS This research used the R package Bibliometrix (Department of Economic and Statistical Sciences at the University of Naples Federico II), VOSviewer (Centre for Science and Technology Studies at Leiden University), and CiteSpace (Drexel University's College of Information Science and Technology), to conduct a bibliometric analysis of 387 papers on breast cancer and ferroptosis from the Web of Science Core Collection. The analysis covers authors, institutions, journals, countries or regions, publication volumes, citations, and keywords. RESULTS The number of publications related to this field has surged annually, with China and the United States collaborating closely and leading in output. Sun Yat-sen University stands out among the institutions, while the journal Frontiers in Oncology and the author Efferth T contribute significantly to the field. Highly cited papers within the domain primarily focus on the induction of ferroptosis, protein regulation, and comparisons with other modes of cell death, providing a foundation for breast cancer treatment. Keyword analysis highlights the maturity of glutathione peroxidase 4-related research, with breast cancer subtypes emerging as motor themes and the tumor microenvironment, immunotherapy, and prognostic models identified as basic themes. Furthermore, the application of nanoparticles serves as an additional complement to the basic themes. CONCLUSIONS The current research status in the field of ferroptosis and breast cancer primarily focuses on the exploration of relevant theoretical mechanisms, whereas future trends and mechanisms emphasize the investigation of therapeutic strategies, particularly the clinical application of immunotherapy related to the tumor microenvironment. Nanotherapy has demonstrated significant clinical potential in this domain. Future research directions should deepen the exploration in this field and accelerate the clinical translation of research findings to provide new insights and directions for the innovation and development of breast cancer treatment strategies.
Collapse
Affiliation(s)
- Jia-Yuan Luo
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yu-Long Deng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shang-Yi Lu
- Department of Hepatological and Gland Surgery, Wuzhou Gongren Hospital/The Seventh Affiliated Hospital of Guangxi Medical University, Wuzhou, China
| | - Si-Yan Chen
- Day Chemotherapy Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rong-Quan He
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Di-Yuan Qin
- Department of Computer Science and Technology, School of Computer and Electronic Information, Guangxi University, Nanning, China
| | - Bang-Teng Chi
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xia Yang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wei Peng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
20
|
Ding Z, Li Z, Sun K, Liu Y, Fang Z, Sun S, Li C, Wang Z. Mitochondrial Regulation of Ferroptosis in Cancer Cells. Int J Biol Sci 2025; 21:2179-2200. [PMID: 40083691 PMCID: PMC11900798 DOI: 10.7150/ijbs.105446] [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: 10/18/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Ferroptosis is an iron-dependent nonapoptotic regulated cell death modality characterized by lethal levels of lipid peroxide accumulation and disrupted antioxidant systems. An increasing number of studies have revealed correlations between ferroptosis and the pathophysiology and treatment of cancer. Given the intricate involvement of mitochondria in ferroptosis, as suggested by previous studies, here, we review advances in understanding the roles of mitochondrial quality control and mitochondrial metabolism (including the roles of the TCA cycle, reactive oxygen species, iron metabolism, and lipid metabolism) in cancer-related ferroptosis and outline the molecular mechanism and clinical translation of mitochondria-related ferroptosis in cancer treatment. with the aim of promoting the precise utilization and prevention of ferroptosis in cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Shengrong Sun
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Chenyuan Li
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Zhong Wang
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
21
|
Wang W, Chen J, Zhan L, Zou H, Wang L, Guo M, Gao H, Xu J, Wu W. Iron and ferroptosis in kidney disease: molecular and metabolic mechanisms. Front Immunol 2025; 16:1531577. [PMID: 39975561 PMCID: PMC11835690 DOI: 10.3389/fimmu.2025.1531577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Maintaining iron homeostasis is necessary for kidney functioning. There is more and more research indicating that kidney disease is often caused by iron imbalance. Over the past decade, ferroptosis' role in mediating the development and progression of renal disorders, such as acute kidney injury (renal ischemia-reperfusion injury, drug-induced acute kidney injury, severe acute pancreatitis induced acute kidney injury and sepsis-associated acute kidney injury), chronic kidney disease (diabetic nephropathy, renal fibrosis, autosomal dominant polycystic kidney disease) and renal cell carcinoma, has come into focus. Thus, knowing kidney iron metabolism and ferroptosis regulation may enhance disease therapy. In this review, we discuss the metabolic and molecular mechanisms of iron signaling and ferroptosis in kidney disease. We also explore the possible targets of ferroptosis in the therapy of renal illness, as well as their existing limitations and future strategies.
Collapse
Affiliation(s)
- Wenjie Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jingdi Chen
- Department of orthopedics, The Airborne Military Hospital, Wuhan, Hubei, China
| | - Liying Zhan
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Handong Zou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lu Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Mengmeng Guo
- The First Clinical College of Wuhan University, Wuhan, Hubei, China
| | - Hang Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Xu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Wu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
22
|
Jang N, Kim IK, Jung D, Chung Y, Kang YP. Regulation of Ferroptosis in Cancer and Immune Cells. Immune Netw 2025; 25:e6. [PMID: 40078787 PMCID: PMC11896659 DOI: 10.4110/in.2025.25.e6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
Ferroptosis, an iron-dependent form of regulated cell death, is driven by lipid peroxidation and shaped by metabolic and antioxidant pathways. In immune cells, ferroptosis susceptibility varies by cell types, lipid composition, and metabolic demands, influencing immune responses in cancer, infections, and autoimmune diseases. Therapeutically, targeting ferroptosis holds promise in cancer immunotherapy by enhancing antitumor immunity or inhibiting immunosuppressive cells. This review highlights the metabolic pathways underlying ferroptosis, its regulation in immune cells, its dual role in tumor progression and antitumor immunity, and its context-dependent therapeutic implications for optimizing cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Yeonseok Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
23
|
Yan HX, Zhang YZ, Niu YQ, Wang YW, Liu LH, Tang YP, Huang JM, Leung ELH. Investigating the interaction between calcium signaling and ferroptosis for novel cancer treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156377. [PMID: 39798340 DOI: 10.1016/j.phymed.2025.156377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/02/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Drug resistance in cancer is steadily rising, making the development of new therapeutic targets increasingly critical for improving treatment outcomes. PURPOSE The mutual regulation of ions is essential for cell growth. Based on this concept, ion interference strategies offer a highly effective approach for cancer treatment. Calcium ions (Ca2+), as major second messengers, are closely associated with ion exchange and homeostasis. Disruptions in this balance can lead to cell death. However, while iron ions are also crucial, the connection between Ca2+and iron-induced cell death (ferroptosis) has not been well established. Therefore, this study suggests that Ca2+ may play a role in the induction of ferroptosis, presenting a novel and efficient target for cancer therapy. STUDY DESIGN PubMed, Google Scholar, and Web of Science databases were systematically searched for articles published in the past 15 years on the mechanisms of calcium ion-induced ferroptosis in cancer and related drugs. RESULTS The analysis highlights how Ca2+regulate ferroptosis. The mechanisms by which Ca2+influence ferroptosis are summarized based on existing literature, and relevant drugs that act on Ca2+/ferroptosis axis are outlined. CONCLUSION Ca2+ regulate ferroptosis primarily through the modulation of reactive oxygen species (ROS) and glutathione (GSH) levels, a mechanism that applies to a wide range of cancer cells as well as paracancerous and normal cells in cancer treatment. Furthermore, plant-derived active compounds exhibit potent anticancer properties and often act on the Ca2+/ferroptosis axis. These natural compounds could play a significant role in the development of new cancer treatment strategies.
Collapse
Affiliation(s)
- Hao-Xin Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), PR China
| | - Yi-Zhong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), PR China
| | - Yu-Qing Niu
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China
| | - Yu-Wei Wang
- Key Laboratory of Shanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shanxi University of Chinese Medicine, Xianyang 712046, Shanxi Province, PR China
| | - Li-Hua Liu
- Economics and Management Yanbian University, Yanji, PR China
| | - Yu-Ping Tang
- Key Laboratory of Shanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shanxi University of Chinese Medicine, Xianyang 712046, Shanxi Province, PR China.
| | - Ju-Min Huang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China.
| |
Collapse
|
24
|
Guo Z, Liu Y, Chen D, Sun Y, Li D, Meng Y, Zhou Q, Zeng F, Deng G, Chen X. Targeting regulated cell death: Apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis in anticancer immunity. J Transl Int Med 2025; 13:10-32. [PMID: 40115032 PMCID: PMC11921819 DOI: 10.1515/jtim-2025-0004] [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] [Indexed: 03/22/2025] Open
Abstract
In the evolving landscape of cancer treatment, the strategic manipulation of regulated cell death (RCD) pathways has emerged as a crucial component of effective anti-tumor immunity. Evidence suggests that tumor cells undergoing RCD can modify the immunogenicity of the tumor microenvironment (TME), potentially enhancing its ability to suppress cancer progression and metastasis. In this review, we first explore the mechanisms of apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis, along with the crosstalk between these cell death modalities. We then discuss how these processes activate antigen-presenting cells, facilitate the cross-priming of CD8+ T cells, and trigger anti-tumor immune responses, highlighting the complex effects of novel forms of tumor cell death on TME and tumor biology. Furthermore, we summarize potential drugs and nanoparticles that can induce or inhibit these emerging RCD pathways and their therapeutic roles in cancer treatment. Finally, we put forward existing challenges and future prospects for targeting RCD in anti-cancer immunity. Overall, this review enhances our understanding of the molecular mechanisms and biological impacts of RCD-based therapies, providing new perspectives and strategies for cancer treatment.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, 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 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yihuang Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, 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 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Danyao Chen
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yuming Sun
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, 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 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, 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 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, 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 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Furong Zeng
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, 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 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan Province, China
- Furong Laboratory, Changsha 410008, 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 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
25
|
Arkan C, Akcora-Yildiz D. FDA-approved disulfiram induces ferroptosis via alteration of redox balance and lipid peroxidation and overcomes carfilzomib resistance in multiple myeloma. Leuk Lymphoma 2025; 66:250-261. [PMID: 39527722 DOI: 10.1080/10428194.2024.2422843] [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: 05/13/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Multiple myeloma (MM), a malignant plasma cell disorder, remains incurable due to inevitable chemo-resistance development, including carfilzomib (CFZ) leading to relapse and poor patient outcomes. Therefore, new treatment strategies, including using FDA-approved alcohol deterrent disulfiram (DSF) are under investigation. The present study investigated the effect of DSF on ferroptosis and CFZ resistance in MM cells. Our findings indicate that DSF increases the production of cytosolic and mitochondrial reactive oxygen species, and causes a loss of mitochondrial Δψ and an elevation in lipid peroxidation in MM cells. DSF treatment in MM cell lines led to the significant downregulation of ferroptotic genes, including glutathione peroxidase 4. Moreover, ferroptosis inhibitor liproxstatin-1rescued DSF-induced ferroptosis by promoting glutathione peroxidase 4 upregulation. DSF alone overcomes CFZ resistance through lipid peroxidation elevation and acts synergistically with CFZ in CFZ-resistant MM cell lines. Our results suggest that DSF is a promising anti-myeloma agent for overcoming CFZ resistance in MM through ferroptosisinduction.
Collapse
Affiliation(s)
- Caglar Arkan
- Department of Biology, Art & Science Faculty, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Dilara Akcora-Yildiz
- Department of Biology, Art & Science Faculty, Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
26
|
Wang Y, Xu Y, Qu Y, Jin Y, Cao J, Zhan J, Li Z, Chai C, Huang C, Li M. Ferroptosis: A novel cell death modality as a synergistic therapeutic strategy with photodynamic therapy. Photodiagnosis Photodyn Ther 2025; 51:104463. [PMID: 39736368 DOI: 10.1016/j.pdpdt.2024.104463] [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: 09/03/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/01/2025]
Abstract
Although there has been significant progress in current comprehensive anticancer treatments centered on surgery, postoperative recurrence and tumor metastasis still significantly affect both prognosis and quality of life of the patient. Hence, the development of precisely targeted tumor therapies and exploration of immunotherapy represent additional strategies for tumor treatment. Photodynamic therapy (PDT) is a relatively safe treatment modality that not only induces multiple modes of tumor cell death but also mediates the secondary immunological responses against tumor resistance and metastasis. Ferroptosis, an iron-dependent type of programmed cell death characterized by accumulation of reactive oxygen species and lipid peroxidation products to lethal levels, has emerged as an attractive target trigger for tumor therapies. Recent research has revealed a close association between PDT and ferroptosis, suggesting that combining ferroptosis inducers with PDT could strengthen their synergistic anti-tumor efficiency. Here in this review, we discuss the rationale for combining PDT with ferroptosis inducers and highlight the progress of single-molecule photosensitizers to induce ferroptosis, as well as the applications of photosensitizers combined with other therapeutic drugs for collaborative therapy. Furthermore, given the current research dilemma, we propose potential therapeutic strategies to advance the combined usage of PDT and ferroptosis inducers, providing the basis and guidelines for prospective clinical translation and research directionality with regard to PDT.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiting Xu
- Central Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan 430022, China
| | - Yong Qu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yifang Jin
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Juanmei Cao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Dermatology, First Affiliated Hospital, Shihezi University, Shihezi 832008, China
| | - Jinshan Zhan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhuoxia Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chuxing Chai
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Changzheng Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
27
|
Pei S, Wang Y, Yao R, Zhang Z, Yin W, Li N. Whole blood exchange ameliorates acute hemolytic anemia by reducing inflammation and oxidative stress in rats. FASEB J 2025; 39:e70358. [PMID: 39878699 PMCID: PMC11777199 DOI: 10.1096/fj.202401748rr] [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: 07/30/2024] [Revised: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 01/31/2025]
Abstract
Hemolytic anemia (HA) is characterized by massive destruction of red blood cells (RBCs) and insufficient oxygen supply, which can lead to shock, organ failure, even death. Recent studies have preliminarily demonstrated the therapeutic effectiveness of whole blood exchange (WBE) in the management of acute hemolytic anemia and exhibited potential for reducing the duration of corticosteroid treatment, while the underlying mechanism of WBE therapy was not investigated in preclinical study. Hence, we investigate the therapeutic mechanisms of WBE in HA through established continued WBE therapy in rats creatively. This study aims to examine the mechanism of WBE on phenylhydrazine hydrochloride-induced hemolytic anemia in SD rats to aid the development of therapeutics for drug-induced hemolytic anemia (DIHA). Research results demonstrated the efficacy of WBE therapy in reducing mortality and ameliorating anemia in DIHA, as evidenced by significant improvements in representative hematological parameters such as RBCs, hemoglobin, and lactate dehydrogenase levels. Additionally, WBE indicated the ability to suppress oxidative stress and inflammation, and it mitigated organ damage and biochemical function by stabilizing hepatic ferroportin levels and decreasing organ iron content. These results highlighted the effectiveness of WBE as an innovative treatment for HA, furnishing evidence to prioritize it over traditional blood transfusion for severe anemias.
Collapse
Affiliation(s)
- Siya Pei
- Department of Blood Transfusion, Xiangya HospitalCentral South UniversityChangshaChina
- Clinical Transfusion Research Centre, Xiangya HospitalCentral South UniversityChangshaChina
- Department of Clinical Laboratory, Xiangya HospitalCentral South UniversityChangshaChina
| | - Yanjie Wang
- Department of Blood Transfusion, Xiangya HospitalCentral South UniversityChangshaChina
- Clinical Transfusion Research Centre, Xiangya HospitalCentral South UniversityChangshaChina
- Department of Clinical Laboratory, Xiangya HospitalCentral South UniversityChangshaChina
| | - Run Yao
- Department of Blood Transfusion, Xiangya HospitalCentral South UniversityChangshaChina
- Clinical Transfusion Research Centre, Xiangya HospitalCentral South UniversityChangshaChina
| | - Zhimin Zhang
- Department of Blood Transfusion, Xiangya HospitalCentral South UniversityChangshaChina
- Clinical Transfusion Research Centre, Xiangya HospitalCentral South UniversityChangshaChina
| | - Wenyu Yin
- Department of Blood Transfusion, Xiangya HospitalCentral South UniversityChangshaChina
- Clinical Transfusion Research Centre, Xiangya HospitalCentral South UniversityChangshaChina
| | - Ning Li
- Department of Blood Transfusion, Xiangya HospitalCentral South UniversityChangshaChina
- Clinical Transfusion Research Centre, Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
28
|
Bhange M, Telange D. Convergence of nanotechnology and artificial intelligence in the fight against liver cancer: a comprehensive review. Discov Oncol 2025; 16:77. [PMID: 39841330 PMCID: PMC11754566 DOI: 10.1007/s12672-025-01821-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025] Open
Abstract
Liver cancer is one of the most challenging malignancies, often associated with poor prognosis and limited treatment options. Recent advancements in nanotechnology and artificial intelligence (AI) have opened new frontiers in the fight against this disease. Nanotechnology enables precise, targeted drug delivery, enhancing the efficacy of therapeutics while minimizing off-target effects. Simultaneously, AI contributes to improved diagnostic accuracy, predictive modeling, and the development of personalized treatment strategies. This review explores the convergence of nanotechnology and AI in liver cancer treatment, evaluating current progress, identifying existing research gaps, and discussing future directions. We highlight how AI-powered algorithms can optimize nanocarrier design, facilitate real-time monitoring of treatment efficacy, and enhance clinical decision-making. By integrating AI with nanotechnology, clinicians can achieve more accurate patient stratification and treatment personalization, ultimately improving patient outcomes. This convergence holds significant promise for transforming liver cancer therapy into a more precise, individualized, and efficient process. However, data privacy, regulatory hurdles, and the need for large-scale clinical validation remain. Addressing these issues will be essential to fully realizing the potential of these technologies in oncology.
Collapse
Affiliation(s)
- Manjusha Bhange
- Department of Pharmaceutics, Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (DU), Sawangi Meghe, Wardha, Maharashtra, 442001, India.
| | - Darshan Telange
- Department of Pharmaceutics, Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (DU), Sawangi Meghe, Wardha, Maharashtra, 442001, India
| |
Collapse
|
29
|
Kung CP, Terzich ND, Ilagen MXG, Prinsen MJ, Kaushal M, Kladney RD, Weber JH, Mabry AR, Torres LS, Bramel ER, Freeman EC, Sabloak T, Cottrell KA, Ryu S, Weber WM, Maggi L, Shriver LP, Patti GJ, Weber JD. ADAR1 Regulates Lipid Remodeling through MDM2 to Dictate Ferroptosis Sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633410. [PMID: 39896528 PMCID: PMC11785053 DOI: 10.1101/2025.01.16.633410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Triple-negative breast cancer (TNBC), lacking expression of estrogen, progesterone, and HER2 receptors, is aggressive and lacks targeted treatment options. An RNA editing enzyme, adenosine deaminase acting on RNA 1 (ADAR1), has been shown to play important roles in TNBC tumorigenesis. We posit that ADAR1 functions as a homeostatic factor protecting TNBC from internal and external pressure, including metabolic stress. We tested the hypothesis that the iron- dependent cell death pathway, ferroptosis, is a ADAR1-protected metabolic vulnerability in TNBC by showing that ADAR1 knockdown sensitizes TNBC cells to GPX4 inhibitors. By performing single-reaction monitoring-based liquid chromatography coupled to mass spectrometry (LC-MS) to measure intracellular lipid contents, we showed that ADAR1 loss increased the abundance of polyunsaturated fatty acid phospholipids (PUFA-PL), of which peroxidation is the primary driver of ferroptosis. Transcriptomic analyses led to the discovery of the proto-oncogene MDM2 contributing to the lipid remodeling in TNBC upon ADAR1 loss. A phenotypic drug screen using a ferroptosis-focused library was performed to identify FDA- approved cobimetinib as a drug-repurposing candidate to synergize with ADAR1 loss to suppress TNBC tumorigenesis. By demonstrating that ADAR1 regulates the metabolic fitness of TNBC through desensitizing ferroptosis, we aim to leverage this metabolic vulnerability to inform basic, pre-clinical, and clinical studies to develop novel therapeutic strategies for TNBC.
Collapse
|
30
|
Wang W, Hashimi B, Wang P. Targeting ferroptosis: the role of non-coding RNAs in hepatocellular carcinoma progression and therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03791-y. [PMID: 39820644 DOI: 10.1007/s00210-025-03791-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
One of the most common tumors is hepatocellular carcinoma (HCC), and the prognosis for late-stage HCC is still not good. It is anticipated that improved outcomes would result from a deeper comprehension of the pathophysiology of HCC. Ferroptosis as a new discovered cell death type is linked to the progression of HCC and may be crucial for its detection, prevention, therapy, and prognosis. Numerous studies suggest that epigenetic alterations mediated by non-coding RNAs (ncRNA) might influence cancer cell susceptibility to ferroptosis. This study elucidates the processes of ferroptosis and delineates the paths by which ncRNAs influence HCC by modulating ferroptosis. Furthermore, it offers significant insights into ferroptosis-associated ncRNAs, intending to discover novel therapeutic approaches for HCC. It also explores innovative concepts for the future use of ncRNA-based ferroptosis-targeted therapeutics.
Collapse
Affiliation(s)
- Weijia Wang
- Department of Oncology, Qilu Hospital of Shandong University Dezhou Hospital (Dezhou People's Hospital), Shandong Province, China
| | - Behishta Hashimi
- Department of Midwifery, Jahan Institute of Health Sciences, Kabul, Afghanistan
| | - Ping Wang
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong Province, China.
| |
Collapse
|
31
|
Xie Z, Hou Q, He Y, Xie Y, Mo Q, Wang Z, Zhao Z, Chen X, Peng T, Li L, Xie W. Ferritin Hinders Ferroptosis in Non-Tumorous Diseases: Regulatory Mechanisms and Potential Consequences. Curr Protein Pept Sci 2025; 26:89-104. [PMID: 39225224 DOI: 10.2174/0113892037315874240826112422] [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: 03/30/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Ferritin, as an iron storage protein, has the potential to inhibit ferroptosis by reducing excess intracellular free iron concentrations and lipid reactive oxygen species (ROS). An insufficient amount of ferritin is one of the conditions that can lead to ferroptosis through the Fenton reaction mediated by ferrous iron. Consequently, upregulation of ferritin at the transcriptional or posttranscriptional level may inhibit ferroptosis. In this review, we have discussed the essential role of ferritin in ferroptosis and the regulatory mechanism of ferroptosis in ferritin-deficient individuals. The description of the regulatory factors governing ferritin and its properties in regulating ferroptosis as underlying mechanisms for the pathologies of diseases will allow potential therapeutic approaches to be developed.
Collapse
Affiliation(s)
- Zhongcheng Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Qin Hou
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yinling He
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Yushu Xie
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Qinger Mo
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Ziyi Wang
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Ziye Zhao
- Class of Clinical Medicine, University of South China, Hengyang 421001, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Liang Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| |
Collapse
|
32
|
Sun Y, Wang D, Yuan C, Lang X, Fu S. Lapatinib: A Potential Therapeutic Agent for Colon Cancer Targeting Ferroptosis. Anticancer Agents Med Chem 2025; 25:114-123. [PMID: 39238394 DOI: 10.2174/0118715206327756240830062531] [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: 05/21/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Colon cancer poses a significant threat to the lives of several patients, impacting their quality of life, thus necessitating its urgent treatment. Lapatinib, a new generation of targeted anti-tumor drugs for clinical application, has yet to be studied for its molecular mechanisms in treating colon cancer. OBJECTIVES This study aimed to uncover the underlying molecular mechanisms through which lapatinib exerts its therapeutic effects in colon cancer treatment. METHODS We accessed pertinent data on patients with colon cancer from the Cancer Genome Atlas (TCGA) database and performed bioinformatics analysis to derive valuable insights. The cell counting kit-8 (CCK8) assay was employed to assess whether lapatinib has a potential inhibitory effect on the growth and proliferation of HT- 29 cells. Additionally, we employed western blot and real-time quantitative polymerase chain reaction methods to investigate whether lapatinib regulates the expression of the ferroptosis-associated protein GPX4 in HT-29 cells. Furthermore, we utilized specific assay kits to measure the levels of reactive oxygen species (ROS) and malondialdehyde in HT-29 cells treated with lapatinib, aiming to elucidate the precise pattern of cell damage induced by this compound. RESULTS GPX4 exhibited high expression levels in tissues from patients with colon cancer and was significantly associated with patient prognosis and diagnosis. Lapatinib inhibited the growth and proliferation of the colon cancer cell line HT-29. Additionally, lapatinib suppressed the expression of GPX4 in HT-29 cells, while the ferroptosis inhibitor ferrostatin-1 (Fer-1) partially restored its expression. Lapatinib induced an increase in intracellular ROS levels and malondialdehyde content in HT-29 cells, with Fer-1 partially restoring these levels. CONCLUSION Our findings demonstrated that lapatinib could effectively suppress the mRNA and protein expression of GPX4 in colon cancer cells, which elevates intracellular levels of ROS and malondialdehyde, ultimately inducing ferroptosis in these cells. This mechanism underscores the potential of lapatinib as a therapeutic strategy for targeting tumors.
Collapse
Affiliation(s)
- Yue Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin, Heilongjiang, 150081, China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, China
| | - Dan Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin, Heilongjiang, 150081, China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, China
| | - Chen Yuan
- Basic Medical College, Harbin Medical University, Harbin, Heilongjiang, 150086, China
| | - Xiujuan Lang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang, 150081, China
| | - Songbo Fu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin, Heilongjiang, 150081, China
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, China
| |
Collapse
|
33
|
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
|
34
|
Peng C, Chen Y, Jiang M. Targeting ferroptosis: a promising strategy to overcome drug resistance in breast cancer. Front Oncol 2024; 14:1499125. [PMID: 39759144 PMCID: PMC11695291 DOI: 10.3389/fonc.2024.1499125] [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: 09/20/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Breast cancer is one of the most prevalent malignancies affecting women worldwide, with its incidence increasingly observed in younger populations. In recent years, drug resistance has emerged as a significant challenge in the treatment of breast cancer, making it a central focus of contemporary research aimed at identifying strategies to overcome this issue. Growing evidence indicates that inducing ferroptosis through various mechanisms, particularly by inhibiting System Xc-, depleting glutathione (GSH), and inactivating glutathione peroxidase 4 (GPX4), holds great potential in overcoming drug resistance in breast cancer. It is anticipated that therapies targeting ferroptosis will emerge as a promising strategy to reverse tumor resistance, offering new hope for breast cancer patients. This review will explore the latest advancements in understanding ferroptosis in the context of breast cancer drug resistance, with a particular emphasis on the roles of ferroptosis inducers and inhibitors, and the impact of ferroptotic pathways on overcoming drug resistance in breast cancer.
Collapse
|
35
|
Zou P, He Q, Xia H, Zhong W. Ferroptosis and its impact on common diseases. PeerJ 2024; 12:e18708. [PMID: 39713140 PMCID: PMC11663406 DOI: 10.7717/peerj.18708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/23/2024] [Indexed: 12/24/2024] Open
Abstract
Ferroptosis is a novel form of programmed cell death characterized by iron accumulation, lipid peroxidation, and a decline in antioxidant capacity, all of which are regulated by gene expression. The onset of numerous diseases is closely associated with ferroptosis. Common diseases affect a large population, reduce the quality of life, and impose an increased burden on the healthcare system. The role of ferroptosis in common diseases, its therapeutic potential, and even its translation into clinical drug treatments are currently significant research topics worldwide. This study preliminarily explores the theoretical basis of ferroptosis, its mechanism and treatment prospect in common diseases including ischaemia-reperfusion injury, inflammatory bowel diseases, liver fibrosis, acute kidney injury, diabetic kidney disease, stroke, Alzheimer's disease, cardiovascular disease, immune and cancer. This review provides a theoretical foundation for the further study and development of ferroptosis, as well as for the prevention and treatment of common diseases.
Collapse
Affiliation(s)
- Pengjian Zou
- Department of Pediatric Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qiuming He
- Department of Pediatric Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Zhong
- Department of Pediatric Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
36
|
Moon JH, Munna AN, Hong JM, Seol JW, Park SY. HIF-1α stabilization inhibits Japanese encephalitis virus propagation and neurotoxicity via autophagy pathways. Biochem Biophys Res Commun 2024; 736:150853. [PMID: 39454305 DOI: 10.1016/j.bbrc.2024.150853] [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: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Japanese encephalitis (JE) is a widespread flavivirus that induces brain inflammation and affects the central nervous system (CNS). Deferoxamine, an iron chelator, has shown promising results in stabilizing HIF-1α, a protein that improves hypoxic conditions, offers protective effects against neurological, and neurodegenerative diseases. This study aimed to assess the impact of HIF-1α stabilization during JEV infection using SH-SY5Y neuroblastoma cell lines as a model. Our findings demonstrated that deferoxamine treatment increased HIF-1α protein levels, leading to a reduction in JEV propagation. Moreover, RT-PCR analysis revealed that deferoxamine ameliorated JEV-induced neuroinflammation and neurotoxicity. We proved that inducing HIF-1α is essential for having an impact of deferoxamine against JEV-mediated neurotoxicity. Thus, our findings offer a potential therapeutic approach to mitigate the detrimental effects of JEV infection on neuronal cells. Further investigations also demonstrated that deferoxamine could reverse JEV-induced autophagy inhibition by stabilizing HIF-1α, which plays a crucial role in mitigating neuronal cell damage and neuroinflammation. Based on our data, HIF-1α stabilization emerges as a vital factor against JEV infection in the neurons, highlighting deferoxamine as a promising and innovative target for developing anti-JEV agents.
Collapse
Affiliation(s)
- Ji-Hong Moon
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Ali Newaz Munna
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Jeong-Min Hong
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Jae-Won Seol
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea.
| |
Collapse
|
37
|
Gao C, Zhang H, Wang X. Current advances on the role of ferroptosis in tumor immune evasion. Discov Oncol 2024; 15:736. [PMID: 39621177 PMCID: PMC11612115 DOI: 10.1007/s12672-024-01573-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/11/2024] [Indexed: 12/06/2024] Open
Abstract
Ferroptosis is a non-apoptotic form of regulated cell death characterized by iron accumulation and uncontrolled lipid peroxidation, leading to plasma membrane rupture and intracellular content release. Cancer immunotherapy, especially immune checkpoint inhibitors (ICIs) targeting PD-1 and PD-L1, has been considered a breakthrough in cancer treatment, achieving encouraging clinical anti-tumor effects in a variety of cancers. However, tumor immune evasion is indispensable to immunotherapy failure. The mechanisms of tumor immune evasion are quite complex, and its occurrence is inseparable from the ferroptosis in tumor microenvironment (TME). Thus, a comprehensive understanding of the role of ferroptosis in tumor immune evasion is crucial to enhance the efficacy of immunotherapy. In this review, we provide an overview of the recent advancements in understanding ferroptosis in cancer, covering molecular mechanisms and interactions with the TME. We also summarize the potential applications of ferroptosis induction in immunotherapy, as well as ferroptosis inhibition for cancer treatment in various conditions. We finally discuss ferroptosis as a double-edged sword, including the current challenges and future directions regarding its potential for cancer treatment.
Collapse
Affiliation(s)
- Changlin Gao
- Graduate School of Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Haoran Zhang
- Central Hospital Affiliated to Dalian University of Technology, Dalian, 116000, Liaoning, China
- Graduate School of Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Xianwei Wang
- Central Hospital Affiliated to Dalian University of Technology, Dalian, 116000, Liaoning, China.
| |
Collapse
|
38
|
Wang K, Zhang X, Fan Y, Zhou L, Duan Y, Li S, Sun Z, Zhang C, Yang H, Yuan W, Peng L, Ma X, Xiang S, Wang T, Yang M, Zhang Z, Wang J, Wang Z, Qian M. Reactivation of MAPK-SOX2 pathway confers ferroptosis sensitivity in KRAS G12C inhibitor resistant tumors. Redox Biol 2024; 78:103419. [PMID: 39527862 PMCID: PMC11585794 DOI: 10.1016/j.redox.2024.103419] [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: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
The clinical success of KRASG12C inhibitors (G12Ci) including AMG510 and MRTX849 is limited by the eventual development of acquired resistance. A novel and effective treatment to revert or target this resistance is urgent. To this end, we established G12Ci (AMG510 and MRTX849) resistant KRASG12C mutant cancer cell lines and screened with an FDA-approved drug library. We found the ferroptosis inducers including sorafenib and lapatinib stood out with an obvious growth inhibition in the G12Ci resistant cells. Mechanistically, the G12Ci resistant cells exhibited reactivation of MAPK signaling, which repressed SOX2-mediated expression of cystine transporter SLC7A11 and iron exporter SLC40A1. Consequently, the low intracellular GSH level but high iron content engendered hypersensitivity of these resistant tumors to ferroptosis inducers. Ectopic overexpression of SOX2 or SLC7A11 and SLC40A1 conferred resistance to ferroptosis in the G12Ci resistant cells. Ferroptosis induced by sulfasalazine (SAS) achieved obvious inhibition on the tumor growth of xenografts derived from AMG510-resistant KRASG12C-mutant cells. Collectively, our results suggest a novel therapeutic strategy to treat patients bearing G12Ci resistant cancers with ferroptosis inducers.
Collapse
Affiliation(s)
- Kai Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yufei Fan
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Liang Zhou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yajun Duan
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Su Li
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhongkan Sun
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Chunqian Zhang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Haoyu Yang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Wenxiu Yuan
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Linyuan Peng
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoyu Ma
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Siliang Xiang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Tianzhi Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Mei Yang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhenyuan Zhang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiaxuan Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhongyuan Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| | - Minxian Qian
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
39
|
Wu S, Wu X, Wang Q, Chen Z, Li L, Chen H, Qi H. Bufalin induces ferroptosis by modulating the 2,4-dienoyl-CoA reductase (DECR1)-SLC7A11 axis in breast cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156130. [PMID: 39427521 DOI: 10.1016/j.phymed.2024.156130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/19/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024]
Abstract
Breast cancer (BC) is a leading cause of cancer-related mortality worldwide. 2,4-dienoyl-CoA reductase (DECR1), an auxiliary component of beta-oxidation, has been recognized for its role in enhancing lipid peroxidation and inducing ferroptosis in prostate cancer. However, its involvement in breast cancer remains largely unexplored. Our study revealed a notably elevated expression of DECR1 in breast cancer tissues, which correlated with increased malignant characteristics. Importantly, the overexpression of DECR1 significantly enhanced proliferation and migration capabilities in MDA-MB-231 cells. Through a comprehensive high-content screening approach, we identified bufalin and its derivative as potent inhibitors of DECR1 expression. Notably, bufalin demonstrated the highest binding energy during molecular docking studies and was found to promote the degradation of DECR1 via autophagy and ubiquitination. Furthermore, bufalin induced ferroptosis in MDA-MB-231 cells by modulating levels of malondialdehyde (MDA), triglycerides (TG), reactive oxygen species (ROS) and Fe2+ while downregulating the expression of hormone-sensitive lipase (HSL), ferritin heavy chain protein 1 (FPN), solute carrier family 7 member 11 (SLC7A11), and glutathione peroxidase 4 (GPX4). These effects were counteracted by DECR1 overexpression. In vivo experiments demonstrated that bufalin inhibited the tumor growth, while decreasing the expression levels of HSL, FPN, SLC7A11, and GPX4, alongside increasing levels of 4-hydroxynonenal (4-HNE). Crucially, the ferroptosis effects induced by bufalin in vivo were also reversed by DECR1 overexpression. Subsequently, we discovered that SLC7A11 interacts with DECR1, inhibition of SLC7A11 led to decreased expression levels of DECR1 along with an accumulation of MDA and Fe2+, effects that were similarly reversed by DECR1 overexpression. Collectively, our findings suggest that targeted therapy against DECR1 combined with further inhibition of its downstream pathway involving SLC7A11/GPX4 may represent a promising strategy for treating breast cancer.
Collapse
Affiliation(s)
- Shiqi Wu
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, PR China
| | - Xuemin Wu
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, PR China
| | - Qin Wang
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, PR China
| | - Zhigang Chen
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, PR China
| | - Li Li
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, PR China
| | - Hongdan Chen
- Chongqing General Hospital, Chongqing University, Department of Breast and Thyroid Surgery, Chongqing, 401147, PR China.
| | - Hongyi Qi
- College of Pharmaceutical Sciences & College of Chinese Medicine, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
40
|
Ge A, Xiang W, Li Y, Zhao D, Chen J, Daga P, Dai CC, Yang K, Yan Y, Hao M, Zhang B, Xiao W. Broadening horizons: the multifaceted role of ferroptosis in breast cancer. Front Immunol 2024; 15:1455741. [PMID: 39664391 PMCID: PMC11631881 DOI: 10.3389/fimmu.2024.1455741] [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: 06/27/2024] [Accepted: 10/21/2024] [Indexed: 12/13/2024] Open
Abstract
Breast cancer poses a serious threat to women's health globally. Current radiotherapy and chemotherapy regimens can induce drug-resistance effects in cancer tissues, such as anti-apoptosis, anti-pyroptosis, and anti-necroptosis, leading to poor clinical outcomes in the treatment of breast cancer. Ferroptosis is a novel programmed cell death modality characterized by iron overload, excessive generation of reactive oxygen species, and membrane lipid peroxidation. The occurrence of ferroptosis results from the imbalance between intracellular peroxidation mechanisms (executive system) and antioxidant mechanisms (defensive system), specifically involving iron metabolism pathways, amino acid metabolism pathways, and lipid metabolism pathways. In recent years, it has been found that ferroptosis is associated with the progression of various diseases, including tumors, hypertension, diabetes, and Alzheimer's disease. Studies have confirmed that triggering ferroptosis in breast cancer cells can significantly inhibit cancer cell proliferation and invasion, and improve cancer cell sensitivity to radiotherapy and chemotherapy, making induction of ferroptosis a potential strategy for the treatment of breast cancer. This paper reviews the development of the concept of ferroptosis, the mechanisms of ferroptosis (including signaling pathways such as GSH-GPX4, FSP1-CoQ1, DHODH-CoQ10, and GCH1-BH4) in breast cancer disease, the latest research progress, and summarizes the research on ferroptosis in breast cancer disease within the framework of metabolism, reactive oxygen biology, and iron biology. The key regulatory factors and mechanisms of ferroptosis in breast cancer disease, as well as important concepts and significant open questions in the field of ferroptosis and related natural compounds, are introduced. It is hoped that future research will make further breakthroughs in the regulatory mechanisms of ferroptosis and the use of ferroptosis in treating breast cancer cells. Meanwhile, natural compounds may also become a new direction for potential drug development targeting ferroptosis in breast cancer treatment. This provides a theoretical basis and opens up a new pathway for research and the development of drugs for the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wang Xiang
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| | - Yan Li
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Da Zhao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junpeng Chen
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, United States
- Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China
| | - Pawan Daga
- Department of Internal Medicine, University of Louisville, Louisville, KY, United States
| | - Charles C. Dai
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States
| | - Kailin Yang
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yexing Yan
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | | | - Wei Xiao
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| |
Collapse
|
41
|
Liu J, Tang R, Zheng J, Luo K. Targeting ferroptosis reveals a new strategy for breast cancer treatment: a bibliometric study. Discov Oncol 2024; 15:679. [PMID: 39560863 DOI: 10.1007/s12672-024-01569-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Studies exploring the role of ferroptosis in the pathogenesis of breast cancer have proliferated over the past decade, especially in 2023, with a staggering 217 publications in related studies. However, there are still significant gaps in comprehensive scientometric analysis and mapping of scientific studies, especially in terms of temporal and study area tracking, principal investigators, and the emergence of new hotspots. OBJECTIVE This study aims to summarize the role of ferroptosis in the development of breast cancer and the latest research results on the ferroptosis-targeted treatment of breast cancer and to use bibliometric methods to draw a visual map to explore future research trends. METHODS On May 11, 2024, this study updated the research progress related to ferroptosis and breast cancer over the past 11 years by retrieving data from January 1, 2014, to May 1, 2024, from the Web of Science database. In this research, many scientific analysis software including VOSviewer, chorddiag R Language Pack, Scimago Graphica, Citespace 6.3.R1, Cluster Profiler, enrichplot, ggplot2 R Language Pack, Cytoscape, and STRING online platform are used to make in-depth scientific analysis and visualization of the measurement results. RESULTS Statistical analysis of these data showed that China accounted for 74.43% of the total publications, highlighting China's dominant role in research on the relationship between ferroptosis and breast cancer. Several research institutions, including Sun Yat-sen University, Zhejiang University, and Shanghai Jiao Tong University, have achieved impressive results. Efferth, Thomas is the most prominent author in this field and has the highest number of publications in the subfield of oncology. This study clearly shows that ferroptosis plays a crucial role in the development of triple-negative breast cancer, hepatocellular carcinoma, glioma, leukemia, mitochondrial disease, lymphoma, bladder tumors, lung adenocarcinoma, and esophageal tumors. CONCLUSION This study provides a comprehensive bibliometric evaluation that deepens our understanding of the role of ferroptosis in the pathogenesis of breast cancer and the current status of targeting ferroptosis for treating breast cancer. Thus, it helps researchers in related fields explore new research directions by comprehensively extracting important information and research hotspots.
Collapse
Affiliation(s)
- Junlin Liu
- Pathology Department of Jingmen Central Hospital, Jingmen, Hubei, China
- Pathology Department of Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| | - Rong Tang
- Pathology Department of Jingmen Central Hospital, Jingmen, Hubei, China
- Pathology Department of Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| | - Jie Zheng
- Pathology Department of Jingmen Central Hospital, Jingmen, Hubei, China
- Pathology Department of Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, Hubei, China
| | - Ke Luo
- Pathology Department of Jingmen Central Hospital, Jingmen, Hubei, China.
- Pathology Department of Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, Hubei, China.
| |
Collapse
|
42
|
Ge T, Wang Y, Han Y, Bao X, Lu C. Exploring the Updated Roles of Ferroptosis in Liver Diseases: Mechanisms, Regulators, and Therapeutic Implications. Cell Biochem Biophys 2024:10.1007/s12013-024-01611-3. [PMID: 39543068 DOI: 10.1007/s12013-024-01611-3] [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] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
Ferroptosis, a newly discovered mode of cell death, is a type of iron-dependent regulated cell death characterized by intracellular excessive lipid peroxidation and imbalanced redox. As the liver is susceptible to oxidative damage and the abnormal iron accumulation is a major feature of most liver diseases, studies on ferroptosis in the field of liver diseases are of great interest. Studies show that targeting the key regulators of ferroptosis can effectively alleviate or even reverse the deterioration process of liver diseases. System Xc- and glutathione peroxidase 4 are the main defense regulators of ferroptosis, while acyl-CoA synthetase long chain family member 4 is a key enzyme causing peroxidation in ferroptosis. Generally speaking, ferroptosis should be suppressed in alcoholic liver disease, non-alcoholic fatty liver disease, and drug-induced liver injury, while it should be induced in liver fibrosis and hepatocellular carcinoma. In this review, we summarize the main regulators involved in ferroptosis and then the mechanisms of ferroptosis in different liver diseases. Treatment options of drugs targeting ferroptosis are further concluded. Determining different triggers of ferroptosis can clarify the mechanism of ferroptosis occurs at both physiological and pathological levels.
Collapse
Affiliation(s)
- Ting Ge
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yang Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yiwen Han
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
43
|
Bai X, Li J, Guo X, Huang Y, Xu X, Tan A, Jia Y, Sun Q, Guo X, Chen J, Kang J. LncRNA MALAT1 promotes Erastin-induced ferroptosis in the HBV-infected diffuse large B-cell lymphoma. Cell Death Dis 2024; 15:819. [PMID: 39532842 PMCID: PMC11557927 DOI: 10.1038/s41419-024-07209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
In a retrospective analysis of clinical data from 587 DLBCL (diffuse large B-cell lymphoma) patients in China, 13.8% of cases were associated with HBV (hepatitis B virus) infection, leading to distinct clinical features and poorer prognosis. Moreover, HBV infection has a more pronounced impact on the survival of the GCB (germinal center B-cell-like) type DLBCL patients compared to the ABC (activated B-cell-like) type. In this study, we found that the expression of LncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) was downregulated in the HBV-infected GCB-type DLBCL patients, and the HBV core protein (HBX) directly inhibited the MALAT1 expression in DLBCL cells. Notably, the overexpression of HBX could attenuate the Erastin-induced ferroptosis in the GCB-type DLBCLs, while MALAT1 re-expression restored sensitivity in the HBX-overexpressing DLBCLs in vitro and in vivo. Mechanistically, MALAT1 competitively hindered SFPQ (splicing factor proline and glutamine-rich) from effectively splicing the pre-mRNA of SLC7A11 (solute carrier family 7 member 11), due to a shared TTGGTCT motif, which impeded the SLC7A11 pre-mRNA maturation and hence diminished its negative regulation on ferroptosis. Together, our study identified HBX's role in inhibiting MALAT1 expression, promoting SFPQ-mediated splicing of SLC7A11 pre-mRNA, and reducing the GCB-type DLBCL sensitivity to Erastin-induced ferroptosis. Combined with the recent studies that ferroptosis may be involved in the occurrence and development of DLBCL, these findings explain our clinical data analysis that DLBCL patients with low expression of MALAT1 have poorer prognosis and shorter overall survival, and provide a valuable therapeutic target for the HBV-infected GCB-type DLBCL patients.
Collapse
MESH Headings
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/virology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Ferroptosis/genetics
- Ferroptosis/drug effects
- Animals
- Hepatitis B virus/genetics
- Cell Line, Tumor
- Male
- Mice
- Hepatitis B/complications
- Hepatitis B/genetics
- Female
- Viral Regulatory and Accessory Proteins
- Gene Expression Regulation, Neoplastic/drug effects
- Middle Aged
- Trans-Activators/metabolism
- Trans-Activators/genetics
- Mice, Nude
Collapse
Affiliation(s)
- Xiaofei Bai
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jianguo Li
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xuecong Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yinghui Huang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xu Xu
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ailing Tan
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yisha Jia
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiaoyi Sun
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xudong Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Jie Chen
- Department of Hematology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Jiuhong Kang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
44
|
Yao H, Tian J, Cheng S, Dou H, Zhu Y. The mechanism of hypoxia-inducible factor-1α enhancing the transcriptional activity of transferrin ferroportin 1 and regulating the Nrf2/HO-1 pathway in ferroptosis after cerebral ischemic injury. Neuroscience 2024; 559:26-38. [PMID: 39168172 DOI: 10.1016/j.neuroscience.2024.08.025] [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: 01/06/2024] [Revised: 07/05/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024]
Abstract
Cerebral ischemic/reperfusion (I/R) injury has high disability and morbidity. Hypoxia-inducible factor-1α (HIF-1α) may enhance the transcriptional activity of transferrin ferroportin 1 (FPN1) in regulating ferroptosis after cerebral ischemia injury (CII). In this study, cerebral I/R injury rat models were established and treated with pcDNA3.1-HIF-1α, pcDNA3.1-NC lentiviral plasmid, or ML385 (a specific Nrf2 inhibitor). Additionally, oxygen-glucose deprivation/reoxygenation (OGD/R) exposed PC12 cells were used as an in vitro model of cerebral ischemia and treated with pcDNA3.1-HIF-1α, si-FPN1, or ML385. The results elicited that cerebral I/R injury rats exhibited increased Longa scores, TUNEL and NeuN co-positive cells, Fe2+ concentration, ROS and HIF-1α levels, and MDA content, while reduced cell density and number, GSH content, and GPX4 protein level. Morphologically abnormal and disordered hippocampal neurons were also observed in CII rats. HIF-1α inhibited brain neuron ferroptosis and ameliorated I/R injury. HIF-1α alleviated OGD-induced PC12 cell ferroptosis. OGD/R decreased FPN1 protein level in PC12 cells, and HIF-1α enhanced FPN1 transcriptional activity. FPN1 knockdown reversed HIF-1α-mediated alleviation of OGD/R-induced ferroptosis. HIF-1α activated the Nrf2/HO-1 pathway by enhancing FPN1 expression and alleviating OGD/R-induced ferroptosis. Conjointly, HIF-1α enhanced the transcriptional activity of FPN1, activated the Nrf2/HO-1 pathway, and inhibited ferroptosis of brain neurons, thereby improving I/R injury in CII rats.
Collapse
Affiliation(s)
- Haiqian Yao
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Jianan Tian
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Shi Cheng
- Department of Orthopaedics, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Haitong Dou
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China
| | - Yulan Zhu
- Department of Neurology, The second Affiliated of Harbin Medical University, Xuefu Road 246, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
45
|
Tian L, Liu Q, Guo H, Zang H, Li Y. Fighting ischemia-reperfusion injury: Focusing on mitochondria-derived ferroptosis. Mitochondrion 2024; 79:101974. [PMID: 39461581 DOI: 10.1016/j.mito.2024.101974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of mortality and morbidity. Current treatments for IRI have limited efficacy and novel therapeutic strategies are needed. Mitochondrial dysfunction not only initiates IRI but also plays a significant role in ferroptosis pathogenesis. Recent studies have highlighted that targeting mitochondrial pathways is a promising therapeutic approach for ferroptosis-induced IRI. The association between ferroptosis and IRI has been reviewed many times, but our review provides the first comprehensive overview with a focus on recent mitochondrial research. First, we present the role of mitochondria in ferroptosis. Then, we summarize the evidence on mitochondrial manipulation of ferroptosis in IRI and review recent therapeutic strategies aimed at targeting mitochondria-related ferroptosis to mitigate IRI. We hope our review will provide new ideas for the treatment of IRI and accelerate the transition from bench to bedside.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, China
| | - Hong Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Honggang Zang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
46
|
Zheng Y, Yan F, He S, Luo L. Targeting ferroptosis in autoimmune diseases: Mechanisms and therapeutic prospects. Autoimmun Rev 2024; 23:103640. [PMID: 39278299 DOI: 10.1016/j.autrev.2024.103640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Ferroptosis is a form of regulated cell death that relies on iron and exhibits unique characteristics, including disrupted iron balance, reduced antioxidant defenses, and abnormal lipid peroxidation. Recent research suggests that ferroptosis is associated with the onset and progression of autoimmune disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and multiple sclerosis (MS). However, the precise effects and molecular mechanisms remain incompletely understood. This article presents an overview of how ferroptosis mechanisms contribute to the development and advancement of autoimmune diseases, as well as the involvement of various immune cells in linking ferroptosis to autoimmune conditions. It also explores potential drug targets within the ferroptosis pathway and recent advancements in therapeutic approaches aimed at preventing and treating autoimmune diseases by targeting ferroptosis. Lastly, the article discusses the challenges and opportunities in utilizing ferroptosis as a potential therapeutic avenue for autoimmune disorders.
Collapse
Affiliation(s)
- Yingzi Zheng
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Fangfang Yan
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China.
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
47
|
Zhang C, Wu Q, Yang H, Zhang H, Liu C, Yang B, Hu Q. Ferroptosis-related gene signature for predicting prognosis and identifying potential therapeutic drug in EGFR wild-type lung adenocarcinoma. Commun Biol 2024; 7:1416. [PMID: 39478024 PMCID: PMC11525656 DOI: 10.1038/s42003-024-07117-1] [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: 02/16/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Epidermal growth factor receptor wild type lung adenocarcinoma (EGFRWT LUAD) still has limited treatment options and unsatisfactory clinical outcomes. Ferroptosis, as a form of cell death, has been reported to play a dual role in regulating tumor cell survival. In this study, we constructed a 3-ferroptosis-gene signature, FeSig, and verified its accuracy and efficacy in predicting EGFRWT LUAD prognosis at both the RNA and protein levels. Patients with higher FeSig scores were found to have worse clinical outcomes. Additionally, we explored the relationship between FeSig and tumor microenvironment, revealing that enhanced interactions between fibroblasts and tumor cells in FeSighigh patients causing tumor resistance to ferroptosis. To address this challenge, we screened potential drugs from NCI-60 (The US National Cancer Institute 60 human tumour cell line anticancer drug screen) and Connectivity map database, ultimately identifying 6-mercatopurine (6-MP) as a promising candidate. Both in vitro and in vivo experiments demonstrated its efficacy in treating FeSighigh EGFRWT LUAD tumor models. In summary, we develop a novel FeSig for predicting prognosis and guiding drug application.
Collapse
Affiliation(s)
- Chuankai Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qi Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hongwei Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hui Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Changqing Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bo Yang
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qingsong Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
| |
Collapse
|
48
|
Tang D, Kang R. NFE2L2 and ferroptosis resistance in cancer therapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:41. [PMID: 39534872 PMCID: PMC11555182 DOI: 10.20517/cdr.2024.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
NFE2-like basic leucine zipper transcription factor 2 (NFE2L2, also known as NRF2), is a key transcription factor in the cellular defense against oxidative stress, playing a crucial role in cancer cell survival and resistance to therapies. This review outlines the current knowledge on the link between NFE2L2 and ferroptosis - a form of regulated cell death characterized by iron-dependent lipid peroxidation - within cancer cells. While NFE2L2 activation can protect normal cells from oxidative damage, its overexpression in cancer cells contributes to drug resistance by upregulating antioxidant defenses and inhibiting ferroptosis. We delve into the molecular pathways of ferroptosis, highlighting the involvement of NFE2L2 and its target genes, such as NQO1, HMOX1, FTH1, FTL, HERC2, SLC40A1, ABCB6, FECH, PIR, MT1G, SLC7A11, GCL, GSS, GSR, GPX4, AIFM2, MGST1, ALDH1A1, ALDH3A1, and G6PD, in ferroptosis resistance. Understanding the delicate balance between NFE2L2's protective and deleterious roles could pave the way for novel therapeutic strategies targeting NFE2L2 to enhance the efficacy of ferroptosis inducers in cancer therapy.
Collapse
Affiliation(s)
- Daolin Tang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TA 75390, USA
| | - Rui Kang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TA 75390, USA
| |
Collapse
|
49
|
Ensoy M, Cansaran-Duman D. Targeting Ferroptosis with Small Molecule Atranorin (ATR) as a Novel Therapeutic Strategy and Providing New Insight into the Treatment of Breast Cancer. Pharmaceuticals (Basel) 2024; 17:1380. [PMID: 39459017 PMCID: PMC11509994 DOI: 10.3390/ph17101380] [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: 08/06/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Ferroptosis results from the accumulation of iron-dependent lipid peroxides and reactive oxygen species (ROS). Previous research has determined the effect of atranorin (ATR) on other cell death mechanisms, but its potential for a ferroptotic effect depending on ROS levels is unclear. This study details the therapeutic role of small-molecule ATR through ferroptosis by suppressing MDA-MB-231, MCF-7, BT-474, and SK-BR-3 breast cancer cells. Methods: The anti-proliferative effect of ATR on cells was evaluated by xCELLigence analysis, and ferroptotic activity was evaluated by enzymatic assay kits. The changes in gene and protein expression levels of ATR were investigated by the qRT-PCR and western blot. In addition, mitochondrial changes were examined by transmission electron microscopy. Results: ATR was found to reduce cell viability in cancer cells in a dose- and time-dependent manner without showing cytotoxic effects on normal breast cells. In BT-474 and MDA-MB-231 cells, ATR, which had a higher anti-proliferative effect, increased iron, lipid peroxidation, and ROS levels in cells and decreased the T-GSH/GSSG ratio. The results revealed for the first time that small-molecule ATR exhibited anti-cancer activity by inducing the glutathione pathway and ferroptosis. Conclusions: This study highlights the potential of ATR as a drug candidate molecule that can be used in the development of new therapeutic strategies for the treatment of triple-negative and luminal-B breast cancer subtypes.
Collapse
|
50
|
Yin J, Xu X, Guo Y, Sun C, Yang Y, Liu H, Yu P, Wu T, Song X. Repair and regeneration: ferroptosis in the process of remodeling and fibrosis in impaired organs. Cell Death Discov 2024; 10:424. [PMID: 39358326 PMCID: PMC11447141 DOI: 10.1038/s41420-024-02181-2] [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: 04/02/2024] [Revised: 09/01/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
As common clinical-pathological processes, wound healing and tissue remodelling following injury or stimulation are essential topics in medical research. Promoting the effective healing of prolonged wounds, improving tissue repair and regeneration, and preventing fibrosis are important and challenging issues in clinical practice. Ferroptosis, which is characterized by iron overload and lipid peroxidation, is a nontraditional form of regulated cell death. Emerging evidence indicates that dysregulated metabolic pathways and impaired iron homeostasis play important roles in various healing and regeneration processes via ferroptosis. Thus, we review the intrinsic mechanisms of tissue repair and remodeling via ferroptosis in different organs and systems under various conditions, including the inflammatory response in skin wounds, remodeling of joints and cartilage, and fibrosis in multiple organs. Additionally, we summarize the common underlying mechanisms, key molecules, and targeted drugs for ferroptosis in repair and regeneration. Finally, we discuss the potential of therapeutic agents, small molecules, and novel materials emerging for targeting ferroptosis to promote wound healing and tissue repair and attenuate fibrosis.
Collapse
Affiliation(s)
- Jiali Yin
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xinjun Xu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Ying Guo
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Caiyu Sun
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yujuan Yang
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Huifang Liu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Second Clinical Medicine College, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Pengyi Yu
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Tong Wu
- Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China.
| | - Xicheng Song
- Department of Otolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China.
| |
Collapse
|