1
|
Chen H, Liu B, Xu P, Wang H, Guo X, Liu G, Yuan J. Mechanistic role of environmental toxicants in inducing cellular ferroptosis and its associated diseases. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118269. [PMID: 40344778 DOI: 10.1016/j.ecoenv.2025.118269] [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: 08/26/2024] [Revised: 03/04/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025]
Abstract
Due to exposure factors such as industrial exhaust, sewage discharge, pesticide runoff, automobile exhaust, and fuel combustion, environmental toxicants are widely present in daily life. Organisms are exposed to these environmental toxicants through contaminated air, food, and drinking water, and these environmental toxicants enter the human body and cause cytotoxicity and diseases through various pathways. As a new cell death mode that is different from cell necrosis, apoptosis, and autophagy, ferroptosis are mainly dysregulation of intracellular iron metabolism, lipid metabolism disorders, and the dysregulation of the antioxidant defense system, leading to lipid peroxidation and ultimately to the rupture of the cell membrane, damage, and cell death. Studies have shown that environmental toxicants induce a series of diseases, such as digestive diseases, urinary diseases, respiratory diseases, neurological disorders, and reproductive diseases, through the above mechanisms. We elaborate the mechanism of common environmental toxicants in inducing ferroptosis and the related systemic diseases mediated through the ferroptosis to provide the theoretical basis for preventing and treating environmental toxicant-related diseases. Nonetheless, our understanding of ferroptosis remains incomplete. For example, mechanisms and methods for the selective control of ferroptosis remain elusive, elucidating these mechanisms and strategies may be critical for leveraging knowledge of ferroptosis to treat related diseases.
Collapse
Affiliation(s)
- Hong Chen
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Bingchun Liu
- Stem Cell Laboratory; Central Laboratory of Organ Transplantation;Inner Mongolia Autonomous Region Engineering Laboratory for Genetic Test and Research of Tumor Cells, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Peixin Xu
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Huizeng Wang
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Xin Guo
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Gang Liu
- Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China
| | - Jianlong Yuan
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, Inner Mongolia 010050, China.
| |
Collapse
|
2
|
Li GZ, Liu JY, Zhou H. Ferroptosis: A novel therapeutic target for diabetic cardiomyopathy. World J Diabetes 2025; 16:104665. [DOI: 10.4239/wjd.v16.i6.104665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/24/2025] [Accepted: 05/07/2025] [Indexed: 06/13/2025] Open
Abstract
Ferroptosis is a new type of programmed cell death caused by the accumulation of iron-dependent lipid peroxides, and it plays a role in the occurrence and progression of diverse diseases. Diabetic cardiomyopathy (DCM), a serious cardiovascular complication in patients with diabetes, eventually progresses to refractory heart failure (HF), which increases the risk of hospitalization for HF and cardiovascular death in patients with diabetes. Despite glycemic control, effective strategies to prevent DCM onset are currently lacking. Accumulating evidence suggests that ferroptosis is involved in oxidative stress, inflammation, and abnormal autophagy in diabetic myocardium, which plays an important role in myocardial apoptosis, hypertrophy, and cardiac fibrosis. The inhibition of ferroptosis can relieve DCM. Presently, ferroptosis inhibitors have been broadly suggested for the treatment of iron overload-related cardiomyopathy. This article reviewed relevant studies to offer a new therapeutic target for DCM.
Collapse
Affiliation(s)
- Gui-Zhi Li
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Jia-Yin Liu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Hong Zhou
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
3
|
Wang Y, Lu J, Lin B, Chen J, Lin F, Zheng Q, Xue X, Wei Y, Chen S, Xu N. Integrated analysis of MIOX gene in prognosis of clear-cell renal cell carcinoma. Cell Death Dis 2025; 16:368. [PMID: 40341358 PMCID: PMC12062366 DOI: 10.1038/s41419-025-07698-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/10/2025]
Abstract
Clear-cell renal cell carcinoma (ccRCC) is a highly aggressive malignancy that originates in the kidney. It often exhibits a limited response or can be refractory to a wide range of anti-cancer therapies, including tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors. Ferroptosis is a form of oxidative, iron-dependent cell death characterized by lipid peroxidation. Targeting ferroptosis may offer a promising alternative therapeutic strategy for cancer cells that are resistant to existing treatments. The impact of ferroptosis-related genes on the prognosis of ccRCC patients is still not fully understood. In this study, we identified 30 differentially expressed ferroptosis-related genes in ccRCC samples compared to normal tissues using data from The Cancer Genome Atlas (TCGA). Lasso regression analyses, along with Kaplan-Meier analysis, were conducted to identify genes associated with prognosis. Based on scRNA-seq and spatial transcriptome analysis, we identified specificity of MIOX in ccRCC. Furthermore, MIOX demonstrated the highest significance, highlighting its independent prognostic value as a single gene in ccRCC. Our findings suggest that MIOX could serve as potential targets for therapeutic interventions in ccRCC.
Collapse
Affiliation(s)
- Yiqiu Wang
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiayi Lu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bohan Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiayin Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Fei Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qingshui Zheng
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xueyi Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shaohao Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
4
|
Mu F, Luo P, Zhu Y, Nie P, Li B, Bai X. Iron Metabolism and Ferroptosis in Diabetic Kidney Disease. Cell Biochem Funct 2025; 43:e70067. [PMID: 40166850 DOI: 10.1002/cbf.70067] [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: 09/15/2024] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
Diabetic kidney disease (DKD) is a major diabetic microvascular complication that still lacks effective therapeutic drugs. Ferroptosis is a recently identified form of programmed cell death that is triggered by iron overload. It is characterized by unrestricted lipid peroxidation and subsequent membrane damage and is found in various diseases. Accumulating evidence has highlighted the crucial roles of iron overload and ferroptosis in DKD. Here, we review iron metabolism and the biology of ferroptosis. The role of aberrant ferroptosis in inducing diverse renal intrinsic cell death, oxidative stress, and renal fibrosis in DKD is summarized, and we elaborate on critical regulatory factors related to ferroptosis in DKD. Finally, we focused on the significance of ferroptosis in the treatment of DKD and highlight recent data regarding the novel activities of some drugs as ferroptosis inhibitors in DKD, aiming to provide new research targets and treatment strategies on DKD.
Collapse
Affiliation(s)
- Fangxin Mu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Ping Nie
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Xue Bai
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Wang MF, Guo J, Yuan SJ, Li K, Zhang Q, Lei HM, Wu JL, Zhao L, Xu YH, Chen X. Targeted sonodynamic therapy induces tumor cell quasi-immunogenic ferroptosis and macrophage immunostimulatory autophagy in glioblastoma. Biomaterials 2025; 315:122913. [PMID: 39471712 DOI: 10.1016/j.biomaterials.2024.122913] [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: 06/19/2024] [Revised: 09/13/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
In this study, we demonstrated the mechanism of a glioblastoma (GBM)-targeted sonodynamic therapy (SDT) strategy employing platelets loaded with a sonosensitizer based on functionalized boron nitride nanoparticles carrying chlorin e6 (BNPD-Ce6). In the in vitro study, we first found that the BNPD-Ce6-mediated sonodynamic action (SDA) induced remarkable viability loss, DNA damage, and cell death in the GBM cells (GBCs) but not macrophages. Surprisingly, the SDA-exposed GBCs displayed a ferroptotic phenotype while the SDA-exposed macrophages underwent immuno-stimulatory autophagy and potently potentiated the SDA's toxicity to the GBCs. The ferroptotic GBCs induced by the SDA were found to be quasi-immunogenic, characterized by the emission of some alarmins such as ATP, HSP90, and CRT, but absent HMGB1, a potent endogenous adjuvant. As such, the SDA-stressed GBCs were unable to stimulate the BMDMs. This defect, interestingly, could be rescued by platelets as a donor of HMGB1 which markedly enhanced the BNPD-Ce6's sonotoxicity to the GBCs. In the in vivo study, we first employed BNPD-Ce6-loaded platelets to achieve ultrasound-triggered, targeted delivery of BNPD-Ce6 in grafted intra-cranial GBMs and subsequent sonodynamic tumor damage. An SDT regimen designed based on these results slowed the growth of grafted intra-cranial GBMs and significantly increased the survival of the host animals. Pathological examination of the SDT-treated GBMs revealed tissue necrosis and destruction and validated the in vitro observations. Finally, the depletion of macrophages was found to abrogate the efficacy of the SDT in subcutaneous GBC grafts. In conclusion, the BNPD-Ce6@Plt-mediated SDT is a practicable and efficacious anti-GBM therapy. Its therapeutic mechanism critically involves a synergy of tumor cell ferroptosis, macrophage stimulation, and platelet activation induced by the SDA.
Collapse
Affiliation(s)
- Meng-Fei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Jie Guo
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Shen-Jun Yuan
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Ke Li
- Center for Lab Teaching, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Quan Zhang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Hui-Mei Lei
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Jia-Lin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China
| | - Li Zhao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RADX), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yong-Hong Xu
- Institute of Ophthalmological Research, Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiao Chen
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Donghu Avenue No.185, Wuhan, 430072, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430072, China.
| |
Collapse
|
6
|
Mao N, Zhang M, Shen M, Yuan J, Lin Z. Research progress on ferroptosis in cerebral hemorrhage. Biomed Pharmacother 2025; 185:117932. [PMID: 40015051 DOI: 10.1016/j.biopha.2025.117932] [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: 10/04/2024] [Revised: 02/06/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
The pathophysiology of intracerebral hemorrhage (ICH) is complex and can cause variable degrees of cell death. Recently, ferroptosis, an emerging cell death mechanism, has garnered significant attention in cerebral hemorrhage disorder. This study aimed to examine iron mortality after cerebral hemorrhage and current targets for potential therapeutic interventions. We specifically focused on iron metabolism abnormalities, lipid peroxidation, and related neuroinflammation and introduced molecular mechanisms, including transcription factors, to gain a better understanding of the underlying mechanisms of ferroptosis and investigate possible therapeutic options for ICH.
Collapse
Affiliation(s)
- Niping Mao
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Zhang
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming Shen
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junhui Yuan
- Department of Neonatology, Wenling maternal and child health care hospital, Wenling, Zhejiang, China.
| | - Zhenlang Lin
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
7
|
Guo D, Cai S, Deng L, Xu W, Fu S, Lin Y, Jiang T, Li Q, Shen Z, Zhang J, Luo P, Tang B, Wang L. Ferroptosis in Pulmonary Disease and Lung Cancer: Molecular Mechanisms, Crosstalk Regulation, and Therapeutic Strategies. MedComm (Beijing) 2025; 6:e70116. [PMID: 39991627 PMCID: PMC11847630 DOI: 10.1002/mco2.70116] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 02/25/2025] Open
Abstract
Ferroptosis is a distinct form of iron-dependent programmed cell death characterized primarily by intracellular iron accumulation and lipid peroxidation. Multiple cellular processes, including amino acid metabolism, iron metabolism, lipid metabolism, various signaling pathways, and autophagy, have been demonstrated to influence the induction and progression of ferroptosis. Recent investigations have elucidated that ferroptosis plays a crucial role in the pathogenesis of various pulmonary disorders, including lung injury, chronic obstructive pulmonary disease, pulmonary fibrosis, and asthma. Ferroptosis is increasingly recognized as a promising novel strategy for cancer treatment. Various immune cells within the tumor microenvironment, including CD8+ T cells, macrophages, regulatory T cells, natural killer cells, and dendritic cells, have been shown to induce ferroptosis in tumor cells and modulate the process through the regulation of iron and lipid metabolism pathways. Conversely, ferroptosis can reciprocally alter the metabolic environment, leading to the activation or inhibition of immune cell functions, thereby modulating immune responses. This paper reviews the molecular mechanism of ferroptosis and describes the tumor immune microenvironment, discusses the connection between ferroptosis and the tumor microenvironment in lung cancer and pulmonary diseases, and discusses the development prospect of their interaction in the treatment of lung cancer and pulmonary diseases.
Collapse
Affiliation(s)
- Dandan Guo
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Songhua Cai
- Department of Thoracic SurgeryNational Cancer CenterNational Clinical Research Center for CancerCancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeShenzhenGuangdongChina
| | - Lvdan Deng
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Wangting Xu
- Department of RespiratoryFirst Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Sentao Fu
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Yaling Lin
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Tong Jiang
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Qing Li
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Zhijun Shen
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Jian Zhang
- The Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Peng Luo
- The Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Bufu Tang
- Department of Radiation OncologyZhongshan HospitalFudan UniversityShanghaiShanghaiChina
| | - Ling Wang
- The Department of OncologyFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| |
Collapse
|
8
|
Chu J. Study of an N6-methyladenosine- and ferroptosis-related prognostic model and the mechanisms underlying the molecular network in neuroblastoma based on multiple datasets. Discov Oncol 2025; 16:200. [PMID: 39964621 PMCID: PMC11836251 DOI: 10.1007/s12672-025-01975-9] [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: 09/06/2024] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
Recent research highlights the pivotal role of N6-methyladenosine (m6A) modification and ferroptosis in the evolution of various cancers. This study aimed to establish a prognostic framework centered on genes associated with m6A and ferroptosis to enhance the accuracy of prognosis predictions for neuroblastoma (NB) patients, thereby improving targeted therapeutic strategies. Patient data, including expression profiles and clinical information from NB cases, were acquired from The Cancer Genome Atlas. Genes related to m6A modification and ferroptosis were identified, and those significant for prognosis were pinpointed using a combination of Cox regression analysis and the least absolute shrinkage and selection operator (LASSO) regression. For further validation, the study utilized external datasets GSE62564 and GSE85047. A prognostic index was computed for each NB patient, followed by analyses of immune cell infiltration and potential drug responsiveness based on the prognostic model. Additionally, enrichment analysis was conducted on the prognostic scores. These scores showed a strong association with the tumor immune environment and the efficacy of prevalent cancer therapies. Moreover, the model's prognostic score emerged as an independent predictive marker for NB. This research succeeded in creating and confirming a prognostic model rooted in m6A and ferroptosis-linked genes, promising to enrich the prognostic understanding and treatment approaches for NB.
Collapse
Affiliation(s)
- Jing Chu
- Department of Pathology, Anhui Provincial Children's Hospital, 39 Wangjiang East Road, Hefei, 230051, Anhui, China.
| |
Collapse
|
9
|
Hu J, Zhang Y, Zhang Y, Shi N, Miu Y, Huang J, Miao M, Ci X. Bergenin inhibits ferritinophagy and ferroptosis in cisplatin-induced acute kidney injury by activating the p-GSK3β/Nrf2/PPARγ pathway. Int Immunopharmacol 2025; 147:114004. [PMID: 39793228 DOI: 10.1016/j.intimp.2024.114004] [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: 10/28/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
Ferroptosis plays a key role in cisplatin-induced acute kidney injury (AKI). Bergenin, which is extracted from Ardisiae Japonicae Herba and has long been used in folk tea and herbal tea drinks, is known to activate Nrf2 and has anti-inflammatory and antioxidant properties, however, its protective influence on CI-AKI has not been elucidated. We used models of cisplatin-induced nephrotoxicity in vitro and CI-AKI models in vivo. In vitro, we found that ferroptosis and ferritinophagy biomarkers were strongly regulated by bergenin treatment. Mechanistic experiments demonstrated that bergenin bound to and phosphorylated GSK3β, which inhibited its activity, to promote the nuclear translocation of Nrf2 and its subsequent binding to the PPARγ promoter sequence to activate PPARγ. However, the protective effects of bergenin on ferroptosis and ferritinophagy in cisplatin-exposed HK-2 cells were diminished when Nrf2 or PPARγ was inhibited. In vivo, bergenin effectively inhibited renal damage induced by cisplatin. Furthermore, bergenin attenuated ferritinophagy-mediated ferroptosis caused by cisplatin; these effects were abolished in Nrf2 knockout mice. Our findings revealed that bergenin effectively protected against ferritinophagy and ferroptosis in CI-AKI, which was largely dependent on the activation of the p-GSK3β/Nrf2/PPARγ pathway.
Collapse
Affiliation(s)
- Jianqiang Hu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Yan Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Yanmin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Ningmohan Shi
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Yufan Miu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Jing Huang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Mochi Miao
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130001, China.
| |
Collapse
|
10
|
Peterson JM, Leclair V, Oyebode OE, Herzallah DM, Nestor-Kalinoski AL, Morais J, Zahedi RP, Alamr M, Di Battista JA, Hudson M. A window into intracellular events in myositis through subcellular proteomics. Inflamm Res 2025; 74:31. [PMID: 39890639 PMCID: PMC11785624 DOI: 10.1007/s00011-025-01996-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/15/2024] [Accepted: 01/06/2025] [Indexed: 02/03/2025] Open
Abstract
OBJECTIVE AND DESIGN Idiopathic inflammatory myopathies (IIM) are a heterogeneous group of inflammatory muscle disorders of unknown etiology. It is postulated that mitochondrial dysfunction and protein aggregation in skeletal muscle contribute to myofiber degeneration. However, molecular pathways that lead to protein aggregation in skeletal muscle are not well defined. SUBJECTS Here we have isolated membrane-bound organelles (e.g., nuclei, mitochondria, sarcoplasmic/endoplasmic reticulum, Golgi apparatus, and plasma membrane) from muscle biopsies of normal (n = 3) and muscle disease patients (n = 11). Of the myopathy group, 10 patients displayed mitochondrial abnormalities (IIM (n = 9); mitochondrial myopathy (n = 1)), and one IIM patient did not show mitochondrial abnormalities (polymyositis). METHODS Global proteomic analysis was performed using an Orbitrap Fusion mass spectrometer. Upon unsupervised clustering, normal and mitochondrial myopathy muscle samples clustered separately from IIM samples. RESULTS We have confirmed previously known protein alterations in IIM and identified several new ones. For example, we found differential expression of (i) nuclear proteins that control cell division, transcription, RNA regulation, and stability, (ii) ER and Golgi proteins involved in protein folding, degradation, and protein trafficking in the cytosol, and (iii) mitochondrial proteins involved in energy production/metabolism and alterations in cytoskeletal and contractile machinery of the muscle. CONCLUSIONS Our data demonstrates that molecular alterations are not limited to protein aggregations in the cytosol (inclusions) and occur in nuclear, mitochondrial, and membrane compartments of IIM skeletal muscle.
Collapse
Affiliation(s)
- Jennifer M Peterson
- Department of Exercise and Rehabilitative Sciences, The University of Toledo, 2801 W. Bancroft St., MS 119, Toledo, OH, 43606, USA.
| | - Valérie Leclair
- Division of Rheumatology, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Olumide E Oyebode
- Department of Exercise and Rehabilitative Sciences, The University of Toledo, 2801 W. Bancroft St., MS 119, Toledo, OH, 43606, USA
| | - Dema M Herzallah
- Department of Exercise and Rehabilitative Sciences, The University of Toledo, 2801 W. Bancroft St., MS 119, Toledo, OH, 43606, USA
| | - Andrea L Nestor-Kalinoski
- Department of Surgery, Advanced Microscopy and Imaging Center, University of Toledo, Toledo, OH, USA
| | - Jose Morais
- Division of Geriatric Medicine and Research Institute, McGill University Health Centre, Montreal, QC, Canada
| | - René P Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Montreal, QC, Canada
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, MB, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
| | - Mazen Alamr
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - John A Di Battista
- Department of Medicine and Experimental Medicine, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Marie Hudson
- Division of Rheumatology, Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
11
|
Gao G, Zhang X. Broadening horizons: research on ferroptosis in lung cancer and its potential therapeutic targets. Front Immunol 2025; 16:1542844. [PMID: 39917300 PMCID: PMC11799241 DOI: 10.3389/fimmu.2025.1542844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/03/2025] [Indexed: 02/09/2025] Open
Abstract
Ferroptosis is a novel form of cell death distinct from traditional mechanisms, characterized by the accumulation of iron ions and the production of lipid peroxides. It not only affects the survival of tumor cells but is also closely linked to changes in the tumor microenvironment. Lung cancer is one of the leading malignancies worldwide in terms of incidence and mortality, and its complex biological mechanisms and resistance make treatment challenging. Recent studies have shown that ferroptosis plays a key role in the onset and progression of lung cancer, with its intricate regulatory mechanisms influencing tumor development and response to therapy. As research into ferroptosis deepens, related molecular pathways, such as glutamate metabolism, iron metabolism, and antioxidant defense, have been gradually revealed. However, in clinical practice, ferroptosis-based therapeutic strategies for lung cancer are still in their early stages. Challenges remain, including the incomplete understanding of the specific mechanisms of ferroptosis, insufficient research on related regulatory factors, and limited insight into the interactions within the tumor microenvironment. Therefore, effective modulation of ferroptosis to enhance lung cancer treatment remains an urgent issue. This review summarizes the biological mechanisms of ferroptosis, analyzes the regulatory factors of ferroptosis in lung cancer cells and their interaction with the tumor microenvironment, and further explores potential therapeutic strategies targeting ferroptosis. By synthesizing the latest research, this paper aims to provide new perspectives and directions for lung cancer treatment, with the goal of advancing clinical applications.
Collapse
Affiliation(s)
| | - Xindi Zhang
- Department of Pulmonary Disease (Department of Respiratory and Critical Care Medicine), Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
12
|
Yan L, Luo G, Han C, Meng J, Liang C. Exploring the oncogenic role of RGS19 in bladder cancer progression and prognosis. Acta Histochem 2024; 126:152212. [PMID: 39481225 DOI: 10.1016/j.acthis.2024.152212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024]
Abstract
This study investigates the role of autophagy-related genes (ARGs) in bladder cancer (BLCA), focusing on the regulator of G protein signaling 19 (RGS19). Using data from The Cancer Genome Atlas (TCGA) and the Human Autophagy Database (HADb), we identified RGS19 as significantly upregulated and linked to poor prognosis in BLCA. Kaplan-Meier survival analysis confirmed its association with increased mortality and. In vitro, RGS19 knockdown in BLCA cell lines inhibited proliferation, migration, and invasion, while inducing apoptosis and autophagy. Transmission electron microscopy showed autophagic structures in RGS19-silenced cells. In vivo, a xenograft mouse model demonstrated reduced tumor growth with RGS19 knockdown. Immunohistochemical (IHC) analysis revealed decreased Ki67 and increased autophagy markers in tumors with reduced RGS19. Pathway analysis suggested RGS19 acts through the cGMP-PKG signaling pathway, validated by altered expression of soluble guanylate cyclase (sGC), protein kinase G (PKG1), phosphodiesterase 5 A (PDE5A), vasodilator-stimulated phosphoprotein (VASP), and phosphorylated VASP (p-VASP) upon RGS19 knockdown. These results highlight RGS19 as a potential biomarker and therapeutic target in BLCA.
Collapse
Affiliation(s)
- Lei Yan
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Institute of Urology, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Guangyue Luo
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Institute of Urology, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Chengxiang Han
- Department of Urology, People's Hospital of Hanshan County, Anhui, China
| | - Jialin Meng
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Institute of Urology, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Chaozhao Liang
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Institute of Urology, Anhui Medical University, Hefei, Anhui, China; Anhui Provincial Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China.
| |
Collapse
|
13
|
Wang M, Chen X, Li S, Wang L, Tang H, Pu Y, Zhang D, Fang B, Bai X. A crosstalk between autophagy and apoptosis in intracerebral hemorrhage. Front Cell Neurosci 2024; 18:1445919. [PMID: 39650799 PMCID: PMC11622039 DOI: 10.3389/fncel.2024.1445919] [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: 06/08/2024] [Accepted: 10/31/2024] [Indexed: 12/11/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is a severe condition that devastatingly harms human health and poses a financial burden on families and society. Bcl-2 Associated X-protein (Bax) and B-cell lymphoma 2 (Bcl-2) are two classic apoptotic markers post-ICH. Beclin 1 offers a competitive architecture with that of Bax, both playing a vital role in autophagy. However, the interaction between Beclin 1 and Bcl-2/Bax has not been conjunctively analyzed. This review aims to examine the crosstalk between autophagy and apoptosis in ICH by focusing on the interaction and balance of Beclin 1, Bax, and Bcl-2. We also explored the therapeutic potential of Western conventional medicine and traditional Chinese medicine (TCM) in ICH via controlling the crosstalk between autophagy and apoptosis.
Collapse
Affiliation(s)
- Moyan Wang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xin Chen
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Shuangyang Li
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Lingxue Wang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hongmei Tang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yuting Pu
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Dechou Zhang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Bangjiang Fang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Department of Emergency, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue Bai
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
14
|
Zhang C, Zhan S, He Y, Pan Z, You Z, Zhu X, Lin Q. Inhibition of CISD2 enhances sensitivity to doxorubicin in diffuse large B-cell lymphoma by regulating ferroptosis and ferritinophagy. Front Pharmacol 2024; 15:1482354. [PMID: 39605902 PMCID: PMC11598492 DOI: 10.3389/fphar.2024.1482354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Background CDGSH iron-sulfur domain 2 (CISD2), an iron-sulfur protein with a [2Fe-2S] cluster, plays a pivotal role in the progression of various cancers, including Diffuse Large B-cell Lymphoma (DLBCL). However, the mechanisms by which CISD2 regulates the occurrence and development of DLBCL remain to be fully elucidated. Methods The potential role of CISD2 as a predictive marker in DLBCL patients treated with the R-CHOP regimen was investigated through bioinformatics analysis and clinical cohort studies. DLBCL cell lines (SUDHL-4 and HBL-1) were employed in this research. Adenoviral (AV) plasmids were used to either silence or overexpress CISD2 in these DLBCL cell lines. Additionally, the induction of ferroptosis in DLBCL cell lines was assessed. Various parameters, including cell proliferation, intracellular free iron levels, lipid peroxides, reactive oxygen species (ROS), and mitochondrial membrane potential (MMP), were measured. Furthermore, the expression of proteins associated with ferroptosis and ferritinophagy was analyzed. Drug-resistant DLBCL cell lines were developed by gradually increasing doxorubicin (DOX) concentration over 6 months. The biological role of CISD2 in these drug-resistant DLBCL cell lines was subsequently assessed. Results Elevated CISD2 levels were found to be associated with decreased sensitivity of DLBCL patients to the R-CHOP regimen, as indicated by bioinformatics and clinical cohort analysis. Silencing CISD2 significantly reduced cell proliferation, increased iron accumulation, depleted glutathione (GSH), and elevated malondialdehyde (MDA) levels, alongside the accumulation of ROS and increased MMP. Additionally, BECN1 and NCOA4 expressions were upregulated, while p62, FTH1, and GPX4 expressions were downregulated. Conversely, overexpression of CISD2 reversed these effects. Treatment of DLBCL cell lines with Erastin led to decreased CISD2 levels. Notably, in drug-resistant DLBCL cell lines, CISD2 knockdown promoted ferroptosis and ferritinophagy, restoring sensitivity to DOX and enhancing the efficacy of Erastin treatment. Conclusion Our findings suggest that CISD2 may play a role in the drug resistance observed in DLBCL patients. Inhibition of CISD2 could enhance ferroptosis and ferritinophagy, potentially improving the sensitivity of DLBCL cells to DOX treatment.
Collapse
Affiliation(s)
- Chaofeng Zhang
- Department of Hematology and Rheumatology, the Affiliated Hospital of Putian University, Putian, Fujian Province, China
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
- School of Basic Medical Science, Putian University, Putian, Fujian Province, China
| | - Siting Zhan
- School of Basic Medical Science, Putian University, Putian, Fujian Province, China
| | - Yanjun He
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Zhiqun Pan
- Department of Hematology and Rheumatology, the Affiliated Hospital of Putian University, Putian, Fujian Province, China
| | - Zhongyi You
- School of Basic Medical Science, Putian University, Putian, Fujian Province, China
| | - Xiongpeng Zhu
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Qi Lin
- Department of Pharmacy, The Affiliated Hospital of Putian University, Putian, Fujian Province, China
- Key Laboratory of Translational Tumor Medicine in Fujian Province, Putian University, Putian, Fujian Province, China
| |
Collapse
|
15
|
Gao D, Wu Y, Zhan Y, Peng L, Zhao L, Cao S, Xue Z, Wang W. Chronic hypoxia drives the occurrence of ferroptosis in liver of fat greening (Hexagrammos otakii) by activating HIF-1α and promoting iron production. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117135. [PMID: 39353379 DOI: 10.1016/j.ecoenv.2024.117135] [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: 06/04/2024] [Revised: 09/08/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Hypoxia caused by global climate change and human activities has become a growing concern eliciting serious effect and damages to aquatic animals. Hexagrammos otakii is usually a victim of hypoxia which caused by high density aquaculture and high nutrient input. The mechanism underlying ferroptosis regulation after hypoxia-stress in liver of H. otakii, however, remains elusive. METHODS For a duration of 15 days, expose the H. otakii to low concentrations of dissolved oxygen (3.4 ± 0.2 mg/L). Detecting alterations in the H. otakii liver tissue by chemical staining, immunohistochemistry, and electron microscopy. The expression variations of relevant genes in the liver of the H. otakii were simultaneously detected using Western blot and qPCR. A correlation analysis was performed between HIF-1α and iron ion expression in the liver of H. otakii following hypoxic stress. RESULTS In this study, we conducted the whole ferroptosis integrated analysis of H. otakii under chronic hypoxic condition. Reactive oxygen species (ROS) are highly accumulated under the hypoxia treatment (Superoxide Dismutase, SOD; Catalase, CAT), and which results in a significantly enhanced of lipid peroxidation (Lipid Peroxidation, LPO; Malondialdehyde, MDA; Aminotransferase, AST; Alanine aminotransferase, ALT) in liver tissue. The HIF-1α signaling is activated to cope with the hypoxia stress through strategies including changing iron ion concentration (Fe3+ and TFR1) to breaking the oxidation balance (GSH and GSH-Px), and enhancing ferroptosis gene expression (GPX4). The expression of genes related to ferroptosis pathway (DMT1, FTH1, STEAP3, ACSL4, γ-GCS, SLC7A11) is significantly upregulated and associated to the expression of iron and HIF-1α. CONCLUSIONS It is demonstrated that the HIF-1α/Fe3+/ROS/GPX4 axis is involved in promoting ferroptosis in fat greening hepatocytes following hypoxia-stress. Ultimately, our findings unveil a process by which hypoxic stress strongly encourages ferroptosis by triggering HIF-1α and boosting iron synthesis.
Collapse
Affiliation(s)
- Dongxu Gao
- Key Laboratory of Applied Biology and Aquaculture of Northern Fishes in Liaoning Province, Dalian Ocean University, Dalian 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Yiting Wu
- Key Laboratory of Applied Biology and Aquaculture of Northern Fishes in Liaoning Province, Dalian Ocean University, Dalian 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Yu Zhan
- Key Laboratory of Applied Biology and Aquaculture of Northern Fishes in Liaoning Province, Dalian Ocean University, Dalian 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Lei Peng
- Key Laboratory of Applied Biology and Aquaculture of Northern Fishes in Liaoning Province, Dalian Ocean University, Dalian 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Ling Zhao
- Key Laboratory of Applied Biology and Aquaculture of Northern Fishes in Liaoning Province, Dalian Ocean University, Dalian 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Shengnan Cao
- Key Laboratory of Applied Biology and Aquaculture of Northern Fishes in Liaoning Province, Dalian Ocean University, Dalian 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Zhuang Xue
- Key Laboratory of Applied Biology and Aquaculture of Northern Fishes in Liaoning Province, Dalian Ocean University, Dalian 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China.
| | - Wei Wang
- Key Laboratory of Applied Biology and Aquaculture of Northern Fishes in Liaoning Province, Dalian Ocean University, Dalian 116023, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
16
|
Li YZ, Deng J, Zhang XD, Li DY, Su LX, Li S, Pan JM, Lu L, Ya JQ, Yang N, Zhou J, Yang LH. Naringenin enhances the efficacy of ferroptosis inducers by attenuating aerobic glycolysis by activating the AMPK-PGC1α signalling axis in liver cancer. Heliyon 2024; 10:e32288. [PMID: 38912485 PMCID: PMC11190665 DOI: 10.1016/j.heliyon.2024.e32288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/25/2024] Open
Abstract
Liver cancer is a heterogeneous disease characterized by poor responses to standard therapies and therefore unfavourable clinical outcomes. Understanding the characteristics of liver cancer and developing novel therapeutic strategies are imperative. Ferroptosis, a type of programmed cell death induced by lipid peroxidation, has emerged as a potential target for treatment. Naringenin, a natural compound that modulates lipid metabolism by targeting AMPK, shows promise in enhancing the efficacy of ferroptosis inducers. In this study, we utilized liver cancer cell lines and xenograft mice to explore the synergistic effects of naringenin in combination with ferroptosis inducers, examining both phenotypic outcomes and molecular mechanisms. Our study results indicate that the use of naringenin at non-toxic doses to hepatocytes can significantly enhance the anticancer effects of ferroptosis inducers (erastin, RSL3, and sorafenib). The combination index method confirmed a synergistic effect between naringenin and ferroptosis inducers. In comparison to naringenin or ferroptosis inducers alone, the combined therapy caused more robust lipid peroxidation and hence more severe ferroptotic damage to cancer cells. The inhibition of aerobic glycolysis mediated by the AMPK-PGC1α signalling axis is the key to naringenin's effect on reducing ferroptosis resistance in liver cancer, and the synergistic cytotoxic effect of naringenin and ferroptosis inducers on cancer cells was reversed after pretreatment with an AMPK inhibitor or a PGC1α inhibitor. Taken together, these findings suggest that naringenin could boost cancer cell sensitivity to ferroptosis inducers, which has potential clinical translational value.
Collapse
Affiliation(s)
- Yong-Zhuo Li
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Deng
- Academic Affairs Office, School of Nursing, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiao-Dong Zhang
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Dong-Yang Li
- Academic Affairs Office, School of Nursing, Guangxi Medical University, Nanning, Guangxi, China
| | - Li-Xi Su
- Academic Affairs Office, School of Nursing, Guangxi Medical University, Nanning, Guangxi, China
| | - Shan Li
- Academic Affairs Office, School of Nursing, Guangxi Medical University, Nanning, Guangxi, China
| | - Jian-Min Pan
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Lan Lu
- Academic Affairs Office, School of Nursing, Guangxi Medical University, Nanning, Guangxi, China
| | - Jia-Qi Ya
- Academic Affairs Office, School of Nursing, Guangxi Medical University, Nanning, Guangxi, China
| | - Nuo Yang
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Zhou
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Li-Hui Yang
- Academic Affairs Office, School of Nursing, Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Biological Molecular Medicine Research(Guangxi Medical University), Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
17
|
Liu K, Chen H, Li Y, Wang B, Li Q, Zhang L, Liu X, Wang C, Ertas YN, Shi H. Autophagy flux in bladder cancer: Cell death crosstalk, drug and nanotherapeutics. Cancer Lett 2024; 591:216867. [PMID: 38593919 DOI: 10.1016/j.canlet.2024.216867] [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: 01/21/2024] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
Autophagy, a self-digestion mechanism, has emerged as a promising target in the realm of cancer therapy, particularly in bladder cancer (BCa), a urological malignancy characterized by dysregulated biological processes contributing to its progression. This highly conserved catabolic mechanism exhibits aberrant activation in pathological events, prominently featured in human cancers. The nuanced role of autophagy in cancer has been unveiled as a double-edged sword, capable of functioning as both a pro-survival and pro-death mechanism in a context-dependent manner. In BCa, dysregulation of autophagy intertwines with cell death mechanisms, wherein pro-survival autophagy impedes apoptosis and ferroptosis, while pro-death autophagy diminishes tumor cell survival. The impact of autophagy on BCa progression is multifaceted, influencing metastasis rates and engaging with the epithelial-mesenchymal transition (EMT) mechanism. Pharmacological modulation of autophagy emerges as a viable strategy to impede BCa progression and augment cell death. Notably, the introduction of nanoparticles for targeted autophagy regulation holds promise as an innovative approach in BCa suppression. This review underscores the intricate interplay of autophagy with cell death pathways and its therapeutic implications in the nuanced landscape of bladder cancer.
Collapse
Affiliation(s)
- Kuan Liu
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Huijing Chen
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Yanhong Li
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Bei Wang
- Department of Gynecology, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Qian Li
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Lu Zhang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China
| | - Xiaohui Liu
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China.
| | - Ce Wang
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China.
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, 38039, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Turkey; UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, 06800, Turkey.
| | - Hongyun Shi
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding, Hebei, 071000, PR China.
| |
Collapse
|