1
|
Chakrabarti A, Verma S. Identifying potential genes driving ferroptosis in the substantia nigra and dopaminergic neurons in Parkinson's disease. Mol Cell Neurosci 2025; 132:103993. [PMID: 39848553 DOI: 10.1016/j.mcn.2025.103993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/07/2025] [Accepted: 01/16/2025] [Indexed: 01/25/2025] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder marked by dopaminergic (DA) neuron degeneration in the substantia nigra (SN). Conventional dopamine replacement therapies provide limited long-term efficacy and significant side effects. Emerging evidence suggests ferroptosis-a form of cell death driven by iron-dependent lipid peroxidation-contributes to PD pathology, though direct evidence linking dysregulation of ferroptosis-related genes in DA neuron loss in PD remains limited. This study explores the expression of ferroptosis-associated genes in the SN and DA neurons of PD patients, identifying potential therapeutic targets. We analyzed two independent RNA-seq datasets, GSE7621 and GSE8397 (GPL-96), from the GEO database to identify common differentially expressed ferroptosis-related genes in the SN of PD patients. We also conducted Gene Ontology and pathway enrichment analyses of these genes to explore the underlying mechanisms and constructed a protein-protein interaction network. The findings were further validated using an additional dataset, GSE49036. We further explored the dysregulation of these ferroptosis-related genes in DA neurons using RNA-seq data GSE169755, derived from DA neurons isolated from the SN of PD patients and controls. Lastly, the proposed hypothesis was experimentally validated in an in vitro PD model. This comprehensive multi-dataset analysis uncovers novel insights into the expression of ferroptosis-related genes in PD, suggesting potential biomarkers and therapeutic targets for mitigating DA neuron loss and PD progression.
Collapse
Affiliation(s)
- Ardra Chakrabarti
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sonia Verma
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, UP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
2
|
Feng W, Chen T. Epigenetic modification mediated by PHF20/METTL14/HOXA13 signaling axis modulates osteogenic differentiation of mesenchymal stem cells. Funct Integr Genomics 2025; 25:7. [PMID: 39757292 DOI: 10.1007/s10142-024-01516-7] [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: 09/14/2024] [Revised: 11/18/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025]
Abstract
This study investigates the mechanism of PHF20 in osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). BMSCs from Balb/c mouse were cultured and identified through osteogenesis, adipogenesis, and flow cytometry. After osteogenic induction, the levels of OPN and OCN in BMSCs were detected by RT-qPCR. Alizarin red staining and alkaline phosphatase (ALP) staining were used to evaluate the osteogenic differentiation ability of BMSCs. PHF20, METTL14, and HOXA13 expressions were detected by RT-qPCR and Western blot. After quantitative analysis of m6A level, RNA immunoprecipitation (RIP) was performed to measure the enrichment of IGF2BP3 or m6A on HOXA13 mRNA. HOXA13 mRNA stability was assessed after actinomycin D treatment. PHF20, METT14, and HOXA13 expressions gradually increased during osteogenic differentiation of BMSCs. Suppression of PHF20 expression repressed the osteogenic differentiation of BMSCs, mainly resulted in a decrease in OPN and OCN levels, reduced mineralization, and weakened ALP activity. Mechanistically, PHF20 elevated METTL14 expression by enhancing the enrichment of H3K4me3 on its promoter, and METTL14 strengthened HOXA13 m6A methylation to maintain HOXA13 mRNA stability through IGF2BP3. In conclusion, PHF20 elevates METTL14 expression by enhancing H3K4me3 enrichment on its promoter and enhances HOXA13 mRNA stability via IGF2BP3-mediated m6A modification, thus facilitating HOXA13 expression and eventually inducing osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Weijia Feng
- Department of Pediatric Orthopaedic, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Ting Chen
- Department of Pediatric Orthopaedic, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
3
|
Luo M, Wang YM, Zhao FK, Luo Y. Recent Advances in Nanomaterial-Mediated Cell Death for Cancer Therapy. Adv Healthc Mater 2025; 14:e2402697. [PMID: 39498722 DOI: 10.1002/adhm.202402697] [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: 07/22/2024] [Revised: 10/01/2024] [Indexed: 11/07/2024]
Abstract
Nanomedicine has shown great anticancer potential by disrupting redox homeostasis and increasing the levels of oxidative stress, but the therapeutic effect is limited by factors including the intrinsic self-protection mechanism of tumors. Cancer cell death can be induced by the exploration of different cell death mechanisms, such as apoptosis, pyroptosis, necroptosis, cuproptosis, and ferroptosis. The merging of nanotechnology with biomedicine has provided tremendous opportunities to construct cell death-based nanomedicine for innovative cancer therapy. Nanocarriers are not only used for the targeted delivery of cell death inducers, but also as therapeutic components to induce cell death to achieve efficient tumor treatment. This review focuses on seven cell death modalities mediated by nanomaterials, such as apoptosis, pyroptosis, necroptosis, ferroptosis, cuprotosis, immunogenic cell death, and autophagy. The mechanisms of these seven cell death modalities are described in detail, as well as the preparation of nanomaterials that induce them and the mechanisms, they used to exert their effects. Finally, this work describes the potential future development based on the current knowledge related to cell death induced by nanomaterials.
Collapse
Affiliation(s)
- Min Luo
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Yuan-Min Wang
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Fu-Kun Zhao
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Yong Luo
- Department of Neurology, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| |
Collapse
|
4
|
Zheng Y, Sun R, Yang H, Gu T, Han M, Yu C, Chen P, Zhang J, Jiang T, Ding Y, Liang L, Quan R, Yao S, Zhao X. Aucubin Promotes BMSCs Proliferation and Differentiation of Postmenopausal Osteoporosis Patients by Regulating Ferroptosis and BMP2 Signalling. J Cell Mol Med 2025; 29:e70288. [PMID: 39823248 PMCID: PMC11740986 DOI: 10.1111/jcmm.70288] [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: 12/13/2023] [Revised: 09/02/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a chronic systemic bone metabolism disorder. Promotion in the patterns of human bone marrow mesenchymal stem cells (hBMSCs) differentiation towards osteoblasts contributes to alleviating osteoporosis. Aucubin, a natural compound isolated from the well-known herbal medicine Eucommia, was previously shown to possess various pharmacological effects. However, its effects on hBMSCs of PMOP patients are unknown. The aim of this present research was to investigate the impact and underlying process of aucubin on cell proliferation and osteogenic differentiation in hBMSCs isolated from PMOP patients. The ability of aucubin to inhibit the ferroptosis induced by erastin in hBMSCs was detected; ROS production, ferrous ion levels, SOD, MDA, and GPX activities were tested by using commercial kits. Next, ALP staining, ARS staining, RT-qPCR, RNA-sequencing, and Western blot were applied for determining the mRNA and protein expression levels associated with the osteogenesis of hBMSCs. The study also explored the involvement of BMP2/Smads signalling in aucubin promoting the osteogenesis of hBMSCs and evaluated the effects of aucubin intervention on osteoporosis using an ovariectomised rat model. The results indicated that aucubin significantly inhibited ROS generation and oxidative stress induced by erastin and protected against ferroptosis in hBMSCs. Additionally, aucubin facilitated osteogenic differentiation of hBMSCs by activating the BMP2/SMADs pathway and attenuated the progression of osteoporosis in OVX rats, suggesting a potential therapeutic benefit for postmenopausal osteoporosis (PMOP).
Collapse
Affiliation(s)
- Yang Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Research Institute of OrthopedicsThe Affiliated Jiangnan Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Rongtai Sun
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Huan Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
| | - Tianyuan Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Meichun Han
- Third Clinical Medical SchoolZhejiang Chinese Medical UniversityHangzhouChina
| | - Congcong Yu
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Pengyu Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Jianhua Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Ting Jiang
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Yangyang Ding
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Long Liang
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Renfu Quan
- Research Institute of OrthopedicsThe Affiliated Jiangnan Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- Third Clinical Medical SchoolZhejiang Chinese Medical UniversityHangzhouChina
| | - Shasha Yao
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Xing Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
5
|
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
|
6
|
Wu SF, Ga Y, Ma DY, Hou SL, Hui QY, Hao ZH. The role of ferroptosis in environmental pollution-induced male reproductive system toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125118. [PMID: 39414070 DOI: 10.1016/j.envpol.2024.125118] [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: 06/30/2024] [Revised: 09/28/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
This article provides a comprehensive review of the toxic effects of environmental pollution on the male reproductive system, with a particular emphasis on ferroptosis, a form of programmed cell death. Research has shown that environmental pollutants, such as heavy metals, pesticide residues, and plastic additives, can disrupt oxidative stress, increasing the production of reactive oxygen species (ROS) in germ cells. This disruption damages cellular lipids, proteins, and DNA, culminating in cell dysfunction or death. Ferroptosis, a cell death pathway closely linked to oxidative stress, is characterized by the accumulation of intracellular iron ions and elevated levels of lipid ROS. This review also explores the role of ferroptosis in male reproductive disorders, including its contributions to reduced sperm count, decreased motility, and abnormal morphology. Environmental pollutants, particularly heavy metals, can induce ferroptosis by interfering with intracellular antioxidant systems, notably the NRF2, GSH, and GPX4 pathways, accumulating toxic lipid peroxides. Furthermore, the article examines the potential interplay between ferroptosis and other forms of cell death, such as apoptosis, autophagy, pyroptosis, and necrosis, in the context of male reproductive health. The review underscores the critical need for further research into the link between environmental pollutants and male fertility, particularly focusing on ferroptosis. It advocates for targeted research efforts to mitigate the adverse effects of ferroptosis and protect reproductive health, emphasizing that a deeper understanding of these mechanisms could lead to innovative preventive strategies against environmental threats to fertility.
Collapse
Affiliation(s)
- Shao-Feng Wu
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Yu Ga
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Dan-Yang Ma
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Si-Lu Hou
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Qiao-Yue Hui
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China.
| | - Zhi-Hui Hao
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya, 572025, China; Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China; National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing, 100193, China.
| |
Collapse
|
7
|
Xie Y, Xie J, Li L. The Role of Methylation in Ferroptosis. J Cardiovasc Transl Res 2024; 17:1219-1228. [PMID: 39075241 DOI: 10.1007/s12265-024-10539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/21/2024] [Indexed: 07/31/2024]
Abstract
Methylation modification is a crucial epigenetic alteration encompassing RNA methylation, DNA methylation, and histone methylation. Ferroptosis represents a newly discovered form of programmed cell death (PCD) in 2012, which is characterized by iron-dependent lipid peroxidation. The comprehensive investigation of ferroptosis is therefore imperative for a more profound comprehension of the pathological and pathophysiological mechanisms implicated in a wide array of diseases. Researches show that methylation modifications can exert either promotive or inhibitory effects on cell ferroptosis. Consequently, this review offers a comprehensive overview of the pivotal role played by methylation in ferroptosis, elucidating its associated factors and underlying mechanisms.
Collapse
Affiliation(s)
- Yushu Xie
- Class of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jie Xie
- Class of Excellent Doctor, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Liang Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
8
|
Yu B, Zeng A, Liu H, Yang Z, Gu C, Luo X, Fu M. LncRNA HOXA11-AS intercepts the POU2F2-mediated downregulation of SLC3A2 in osteoarthritis to suppress ferroptosis. Cell Signal 2024; 124:111399. [PMID: 39251054 DOI: 10.1016/j.cellsig.2024.111399] [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: 05/14/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent ailment characterized by the gradual degradation of joints, resulting in discomfort and restricted movement. The recently proposed mechanism of ferroptosis is intricately associated with the initiation and progression of OA. Our study found that the long non-coding RNA HOXA11-AS reduces ferroptosis by increasing the expression of SLC3A2 through the transcription factor POU2F2. MATERIALS AND METHODS HOXA11-AS was identified through lncRNA microarray analysis, and its impact on chondrocytes and extracellular matrix was assessed using real-time quantitative PCR, western blotting, and CCK8 assays. Subsequently, overexpression of HOXA11-AS in the knee joints of mice confirmed its protective efficacy on chondrocyte phenotype in the OA model. The involvement of HOXA11-AS in regulating ferroptosis via SLC3A2 was further validated through RNA sequencing analysis of mouse cartilage and the assessment of malondialdehyde levels and glutathione peroxidase activity. Finally, a combination of RNA sequencing, pull-down assays, mass spectrometry (MS), and chromatin immunoprecipitation (ChIP) techniques was employed to identify POU2F2 as the crucial transcription factor responsible for repressing the expression of SLC3A2, which can be effectively inhibited by HOXA11-AS. RESULTS Our study demonstrated that HOXA11-AS effectively enhanced the metabolic homeostasis of chondrocytes, and alleviated the progression of OA in vitro and in vivo experiments. Furthermore, HOXA11-AS was found to enhance SLC3A2 expression, a key regulator of ferroptosis, by interacting with the transcriptional repressor POU2F2. CONCLUSIONS HOXA11-AS promotes SLC3A2 expression and inhibits chondrocyte ferroptosis, by binding to the transcriptional repressor POU2F2, offering a promising and innovative therapeutic approach for OA.
Collapse
Affiliation(s)
- Baoxi Yu
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China.
| | - Anyu Zeng
- Department of Bone and Soft Tissue Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China.
| | - Hailong Liu
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China; Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, PR China.
| | - Zhijian Yang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China.
| | - Cheng Gu
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China.
| | - Xuming Luo
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China.
| | - Ming Fu
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, PR China.
| |
Collapse
|
9
|
Li F, Wang F, Wang L, Wang J, Wei S, Meng J, Li Y, Feng L, Jiang P. m6A reader YTHDC2 mediates NCOA4 mRNA stability affecting ferritinophagy to alleviate secondary injury after intracerebral haemorrhage. Epigenetics 2024; 19:2326868. [PMID: 38465865 PMCID: PMC10936596 DOI: 10.1080/15592294.2024.2326868] [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: 07/27/2023] [Accepted: 03/01/2024] [Indexed: 03/12/2024] Open
Abstract
Oxidative stress and neuronal dysfunction caused by intracerebral haemorrhage (ICH) can lead to secondary injury. The m6A modification has been implicated in the progression of ICH. This study aimed to investigate the role of the m6A reader YTHDC2 in ICH-induced secondary injury. ICH models were established in rats using autologous blood injection, and neuronal cell models were induced with Hemin. Experiments were conducted to overexpress YTH domain containing 2 (YTHDC2) and examine its effects on neuronal dysfunction, brain injury, and neuronal ferritinophagy. RIP-qPCR and METTL3 silencing were performed to investigate the regulation of YTHDC2 on nuclear receptor coactivator 4 (NCOA4). Finally, NCOA4 overexpression was used to validate the regulatory mechanism of YTHDC2 in ICH. The study found that YTHDC2 expression was significantly downregulated in the brain tissues of ICH rats. However, YTHDC2 overexpression improved neuronal dysfunction and reduced brain water content and neuronal death after ICH. Additionally, it reduced levels of ROS, NCOA4, PTGS2, and ATG5 in the brain tissues of ICH rats, while increasing levels of FTH and FTL. YTHDC2 overexpression also decreased levels of MDA and Fe2+ in the serum, while promoting GSH synthesis. In neuronal cells, YTHDC2 overexpression alleviated Hemin-induced injury, which was reversed by Erastin. Mechanistically, YTHDC2-mediated m6A modification destabilized NCOA4 mRNA, thereby reducing ferritinophagy and alleviating secondary injury after ICH. However, the effects of YTHDC2 were counteracted by NCOA4 overexpression. Overall, YTHDC2 plays a protective role in ICH-induced secondary injury by regulating NCOA4-mediated ferritinophagy.
Collapse
Affiliation(s)
- Fengfeng Li
- Department of Neurosurgery, Tengzhou Central People’s Hospital, Jining Medical University, Tengzhou, China
| | - Fang Wang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| | - Lei Wang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| | - Jianhua Wang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| | - Shanshan Wei
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Junjun Meng
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| | - Yanan Li
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| | - Lei Feng
- Department of Neurosurgery, Jining First People’s Hospital, Shandong First Medical University, Jining, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
| |
Collapse
|
10
|
Zheng Z, Song Y. Integrated analysis of disulfidptosis-related genes SLC7A11, SLC3A2, RPN1 and NCKAP1 across cancers. Discov Oncol 2024; 15:724. [PMID: 39612126 DOI: 10.1007/s12672-024-01612-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: 03/20/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
Disulfidptosis, a newly identified form of regulated cell death associated with disruption of disulfide bond formation in the endoplasmic reticulum, involves the dysregulation of disulfidptosis-related genes (DRGs) that may contribute to cancer development and progression. However, the molecular mechanisms and clinical implications of DRGs in different cancer types remain poorly characterized. Therefore, in this comprehensive study, we investigated the expression, prognostic value, and functional roles of four recently identified DRGs (SLC7A11, SLC3A2, RPN1, and NCKAP1) across various cancers. Especially, in clinical samples of glioblastoma, we found that RPN1 was significantly correlated with patient survival. Through mutation landscape analysis, we identified diverse missense mutations in these DRGs, with NCKAP1 exhibiting the highest mutation frequency (5.9% in skin cutaneous melanoma). Additionally, we observed positive correlations between these DRGs and tumor stemness (DNAss DNA stemness score and RNAss RNA stemness score) as well as RNA modifications, particularly m6A modification, in several cancer types. Furthermore, high expression of SLC7A11, RPN1, and NCKAP1 was positively associated with infiltration of T-helper type 2 (Th2) cells in various cancers, while high expression of SLC7A11, SLC3A2, and RPN1 correlated with tumor mutational burden (TMB) in 10, 4, and 8 tumor types, respectively. Utilizing a protein-protein interaction network, we identified the RHO GTPases Activate WASPs and WAVEs pathway as significantly enriched, suggesting the involvement of these DRGs in cancer-related signaling pathways. Collectively, our findings provide novel insights into the molecular mechanisms and clinical implications of DRGs in pan-cancer, highlighting their potential as biomarkers and therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Zequn Zheng
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, No. 378 Dongqing Road, Yinzhou District, Ningbo, 315000, Zhejiang, China.
- Shantou University Medical College, No.22, Xinling Road, Jinping District, Shantou, 515000, Guangdong, China.
| | - Yongfei Song
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, No. 378 Dongqing Road, Yinzhou District, Ningbo, 315000, Zhejiang, China.
| |
Collapse
|
11
|
Tao X, Kang N, Zheng Z, Zhu Z, Ma J, He W. The regulatory mechanisms of N6-methyladenosine modification in ferroptosis and its implications in disease pathogenesis. Life Sci 2024; 355:123011. [PMID: 39181316 DOI: 10.1016/j.lfs.2024.123011] [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: 05/06/2024] [Revised: 07/30/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
HEADING AIMS Based on the current knowledge of the molecular mechanisms by which m6A influences ferroptosis, our objective is to underscore the intricate and interdependent relationships between m6A and the principal regulatory pathways of ferroptosis, as well as other molecules, emphasizing its relevance to diseases associated with this cell death mode. MATERIALS AND METHODS We conducted a literature search using the keywords "m6A and ferroptosis" across PubMed, Web of Science, and Medline. The search was limited to English-language publications from 2017 to 2024. Retrieved articles were managed using Endnote software. Two authors independently screened the search results and reviewed the full texts of selected articles. KEY FINDINGS Abnormal m6A levels are often identified as critical regulators of ferroptosis. Specifically, "writers", "readers" and "erasers" that dynamically modulate m6A function regulate various pathways in ferroptosis including iron metabolism, lipid metabolism and antioxidant system. Additionally, we provide an overview of the role of m6A-mediated ferroptosis in multiple diseases and summarize the potential applications of m6A-mediated ferroptosis, including its use as a therapeutic target for diseases and as diagnostic as well as prognostic biomarkers. SIGNIFICANCE N6-methyladenosine (m6A) modification, a prevalent RNA modification in eukaryotic cells, is crucial in regulating various aspects of RNA metabolism. Notably, accumulating evidence has implicated m6A modification in ferroptosis, a form of iron-dependent cell death characterized by elevated iron levels and lipid peroxide accumulation. Overall, this review sheds light on the potential diagnostic and therapeutic applications of m6A regulators in addressing conditions associated with ferroptosis.
Collapse
Affiliation(s)
- Xiao Tao
- Department of Clinical Medicine, The First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Ningning Kang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, PR China
| | - Zongqin Zheng
- Department of Anesthesiology, The Second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Ziyi Zhu
- Department of Clinical Medicine, The First School of Clinical Medicine, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Junting Ma
- Department of Immunology and Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China.
| | - Wei He
- Department of Immunology and Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, PR China.
| |
Collapse
|
12
|
Yin Z, Ma L, Tian X, Sun Q, Zhang C, Wang Y, Miao Y, Xue X, Wang Y, Wang J, Zhang X, Hou X. Downregulation of the m 6A reader YTHDC2 upregulates exosome content in lung adenocarcinoma via inhibiting IFIT and OAS family members. J Biol Chem 2024; 300:107783. [PMID: 39303913 PMCID: PMC11736008 DOI: 10.1016/j.jbc.2024.107783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/01/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024] Open
Abstract
N6-Methyladenosine (m6A) is the most prevalent mRNA modification. Its biological function primarily relies on its "Reader" protein, such as YTHDC2. Previous studies have shown that YTHDC2 downregulation is a procarcinogenic phenomenon in lung adenocarcinoma (LUAD). However, further investigation is needed to understand the molecular mechanisms of downstream genes and the associated biological phenomena following YTHDC2 downregulation. Here, we found that YTHDC2 knockout upregulated exosome content in LUAD. Following YTHDC2 knockout, the mRNA levels of OAS family members (OASs) and IFIT family members (IFITs) also decreased; and inhibition of OASs and IFITs could promote exosome content. Several m6A modification sites on the NT domain of OASs and the TPR12 domain of IFITs were found to increase the stability of OASs and IFITs in a YTHDC2-dependent manner. OASs and IFITs affected exosome content through target genes including RAB5A, RAB7, and RAB11A, and three arginine (R) amino acids on IFITs were critical for combination IFITs with targeted RAB mRNAs and subsequent degradation. Simultaneously, OASs degraded targeted RABs through RNAseL. Additionally, mutual bindings between OASs and IFITs were critical for their target gene degradation. Collectively, the above findings might provide a theoretical basis for the treatment of LUAD patients with low YTHDC2 expression.
Collapse
Affiliation(s)
- Zhixin Yin
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifang Ma
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoting Tian
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Sun
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Congcong Zhang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yikun Wang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yayou Miao
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangfei Xue
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongjie Wang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiayi Wang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiao Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xumin Hou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Ma B, Hu X, Ai X, Zhang Y. Research progress of ferroptosis and inflammatory bowel disease. Biometals 2024; 37:1039-1062. [PMID: 38713412 DOI: 10.1007/s10534-024-00604-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/09/2024] [Indexed: 05/08/2024]
Abstract
Inflammatory bowel disease (IBD) is a non-specific chronic inflammatory disorder of the gastrointestinal tract, imposing significant burdens on both society and individuals. As a new type of regulated cell death (RCD), ferroptosis is different from classic RCDs such as apoptosis and necrosis in cell morphology, biochemistry and genetics. The main molecular mechanisms of ferroptosis include dysregulation of iron metabolism, impaired antioxidant capacity, mitochondrial dysfunction, accumulation of lipid-associated super-oxides, and membrane disruption. In recent years, increasing evidence has shown that ferroptosis is involved in the pathophysiology of inflammatory bowel disease. However, the exact roles and underlying molecular mechanisms have not been fully elucidated. This article reviews the mechanism of ferroptosis in the occurrence and development of inflammatory bowel disease, in order to provide new ideas for the pathophysiological research of inflammatory bowel disease. Additionally, we discuss potential strategies for the prevention and treatment of inflammatory bowel disease by targeting ferroptosis.
Collapse
Affiliation(s)
- Baolian Ma
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Xiaoxue Hu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Xiaowen Ai
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Yonglan Zhang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China.
| |
Collapse
|
14
|
Shi JX, Zhang ZC, Yin HZ, Piao XJ, Liu CH, Liu QJ, Zhang JC, Zhou WX, Liu FC, Yang F, Wang YF, Liu H. RNA m6A modification in ferroptosis: implications for advancing tumor immunotherapy. Mol Cancer 2024; 23:213. [PMID: 39342168 PMCID: PMC11437708 DOI: 10.1186/s12943-024-02132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
The pursuit of innovative therapeutic strategies in oncology remains imperative, given the persistent global impact of cancer as a leading cause of mortality. Immunotherapy is regarded as one of the most promising techniques for systemic cancer therapies among the several therapeutic options available. Nevertheless, limited immune response rates and immune resistance urge us on an augmentation for therapeutic efficacy rather than sticking to conventional approaches. Ferroptosis, a novel reprogrammed cell death, is tightly correlated with the tumor immune environment and interferes with cancer progression. Highly mutant or metastasis-prone tumor cells are more susceptible to iron-dependent nonapoptotic cell death. Consequently, ferroptosis-induction therapies hold the promise of overcoming resistance to conventional treatments. The most prevalent post-transcriptional modification, RNA m6A modification, regulates the metabolic processes of targeted RNAs and is involved in numerous physiological and pathological processes. Aberrant m6A modification influences cell susceptibility to ferroptosis, as well as the expression of immune checkpoints. Clarifying the regulation of m6A modification on ferroptosis and its significance in tumor cell response will provide a distinct method for finding potential targets to enhance the effectiveness of immunotherapy. In this review, we comprehensively summarized regulatory characteristics of RNA m6A modification on ferroptosis and discussed the role of RNA m6A-mediated ferroptosis on immunotherapy, aiming to enhance the effectiveness of ferroptosis-sensitive immunotherapy as a treatment for immune-resistant malignancies.
Collapse
Affiliation(s)
- Jun-Xiao Shi
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Zhi-Chao Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Hao-Zan Yin
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Xian-Jie Piao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Cheng-Hu Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Qian-Jia Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Jia-Cheng Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Wen-Xuan Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu-Chen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu Yang
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China.
- Key Laboratory of Biosafety Defense, Ministry of Education, Shanghai, 200433, China.
- Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China.
| | - Yue-Fan Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| |
Collapse
|
15
|
Sun LL, He HY, Li W, Jin WL, Wei YJ. The solute carrier transporters (SLCs) family in nutrient metabolism and ferroptosis. Biomark Res 2024; 12:94. [PMID: 39218897 PMCID: PMC11367818 DOI: 10.1186/s40364-024-00645-2] [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: 07/12/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Ferroptosis is a novel form of programmed cell death caused by damage to lipid membranes due to the accumulation of lipid peroxides in response to various stimuli, such as high levels of iron, oxidative stress, metabolic disturbance, etc. Sugar, lipid, amino acid, and iron metabolism are crucial in regulating ferroptosis. The solute carrier transporters (SLCs) family, known as the "metabolic gating" of cells, is responsible for transporting intracellular nutrients and metabolites. Recent studies have highlighted the significant role of SLCs family members in ferroptosis by controlling the transport of various nutrients. Here, we summarized the function and mechanism of SLCs in ferroptosis regulated by ion, metabolic control of nutrients, and multiple signaling pathways, with a focus on SLC-related transporters that primarily transport five significant components: glucose, amino acid, lipid, trace metal ion, and other ion. Furthermore, the potential clinical applications of targeting SLCs with ferroptosis inducers for various diseases, including tumors, are discussed. Overall, this paper delves into the novel roles of the SLCs family in ferroptosis, aiming to enhance our understanding of the regulatory mechanisms of ferroptosis and identify new therapeutic targets for clinical applications.
Collapse
Affiliation(s)
- Li-Li Sun
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Hai-Yan He
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, P. R. China.
| | - Yi-Ju Wei
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
16
|
Chen J, Li X, Tao J, Luo L. Identification of Marine-Derived SLC7A11 Inhibitors: Molecular Docking, Structure-Based Virtual Screening, Cytotoxicity Prediction, and Molecular Dynamics Simulation. Mar Drugs 2024; 22:375. [PMID: 39195490 PMCID: PMC11355350 DOI: 10.3390/md22080375] [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: 07/18/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
The search for anticancer drugs that target ferroptosis is a promising avenue of research. SLC7A11, a key protein involved in ferroptosis, has been identified as a potential target for drug development. Through screening efforts, novel inhibitors of SLC7A11 have been designed with the aim of promoting ferroptosis and ultimately eliminating cancer cells. We initially screened 563 small molecules using pharmacophore and 2D-QSAR models. Molecular docking and ADMET toxicity predictions, with Erastin as a positive control, identified the small molecules 42711 and 27363 as lead compounds with strong inhibitory activity against SLC7A11. Further optimization resulted in the development of a new inhibitor structure (42711_11). Molecular docking and ADMET re-screening demonstrated successful fragment substitution for this small molecule. Final molecular dynamics simulations also confirmed its stable interaction with the protein. These findings represent a significant step towards the development of new therapeutic strategies for ferroptosis-related diseases.
Collapse
Affiliation(s)
- Jiaqi Chen
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (J.C.); (X.L.); (J.T.)
| | - Xuan Li
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (J.C.); (X.L.); (J.T.)
| | - Jiahua Tao
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (J.C.); (X.L.); (J.T.)
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang 524023, China
| |
Collapse
|
17
|
Li MY, Shi YC, Xu WX, Zhao L, Zhang AZ. Exploring Cr(VI)-induced blood-brain barrier injury and neurotoxicity in zebrafish and snakehead fish, and inhibiting toxic effects of astaxanthin. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 355:124280. [PMID: 38815890 DOI: 10.1016/j.envpol.2024.124280] [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: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
Cr(VI) is a common hazardous heavy metal contaminant that seriously endangers human and aquatic animal health. GPX4 was the key enzyme that reduces heavy metal toxicity through inhibiting ferroptosis pathway. Astaxanthin was GPX4 activator that can weaken biological toxicity induced by Cr(VI) exposure. The present study was conducted to evaluate the major role of GPX4 in astaxanthin protects Cr(VI)-induced oxidative damage, blood-brain barrier injury and neurotoxicity in brain-liver axis through inhibiting ferroptosis pathway. In the current study, astaxanthin intervention can effectively alleviate Cr(VI)-induced oxidative stress, blood-brain barrier damage, and neurotoxicity. GPX4 plays a major role in mediating astaxanthin nutritional intervention to reduce ROS and liver non-heme iron accumulation, which would contribute to the reduction of ferroptosis. Meanwhile, astaxanthin maintains the stability of transport receptors and protein macromolecules such as TMEM163, SLC7A11, SLC3A2, FPN1 and GLUT1 in the brain liver axis, promoting substance exchange and energy supply. Moreover, astaxanthin alleviates Cr(VI)-induced neurotoxicity by promoting tight protein expression and reducing blood-brain barrier permeability.
Collapse
Affiliation(s)
- Mu-Yang Li
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Yan-Chao Shi
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Wan-Xin Xu
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Lei Zhao
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China.
| | - Ai-Zhong Zhang
- Key Laboratory of Efficient Utilization of Feed Resources and Nutrition Manipulation in Cold Region of Heilongjiang Province, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China.
| |
Collapse
|
18
|
Zhou J, Tang J, Zhang C, Li G, Lin X, Liao S, Luo J, Yu G, Zheng F, Guo Z, Shao W, Hu H, Xu L, Wu S, Li H. ALKBH5 targets ACSL4 mRNA stability to modulate ferroptosis in hyperbilirubinemia-induced brain damage. Free Radic Biol Med 2024; 220:271-287. [PMID: 38734267 DOI: 10.1016/j.freeradbiomed.2024.05.014] [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: 03/14/2024] [Revised: 04/18/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
Bilirubin-induced brain damage is a serious clinical consequence of hyperbilirubinemia, yet the underlying molecular mechanisms remain largely unknown. Ferroptosis, an iron-dependent cell death, is characterized by iron overload and lipid peroxidation. Here, we report a novel regulatory mechanism of demethylase AlkB homolog 5 (ALKBH5) in acyl-coenzyme A synthetase long-chain family member 4 (ACSL4)-mediated ferroptosis in hyperbilirubinemia. Hyperdifferential PC12 cells and newborn Sprague-Dawley rats were used to establish in vitro and in vivo hyperbilirubinemia models, respectively. Proteomics, coupled with bioinformatics analysis, first suggested the important role of ferroptosis in hyperbilirubinemia-induced brain damage. In vitro experiments showed that ferroptosis is activated in hyperbilirubinemia, and ferroptosis inhibitors (desferrioxamine and ferrostatin-1) treatment effectively alleviates hyperbilirubinemia-induced oxidative damage. Notably, we observed that the ferroptosis in hyperbilirubinemia was regulated by m6A modification through the downregulation of ALKBH5 expression. MeRIP-seq and RIP-seq showed that ALKBH5 may trigger hyperbilirubinemia ferroptosis by stabilizing ACSL4 mRNA via m6A modification. Further, hyperbilirubinemia-induced oxidative damage was alleviated through ACSL4 genetic knockdown or rosiglitazone-mediated chemical repression but was exacerbated by ACSL4 overexpression. Mechanistically, ALKBH5 promotes ACSL4 mRNA stability and ferroptosis by combining the 669 and 2015 m6A modified sites within 3' UTR of ACSL4 mRNA. Our findings unveil a novel molecular mechanism of ferroptosis and suggest that m6A-dependent ferroptosis could be an underlying clinical target for the therapy of hyperbilirubinemia.
Collapse
Affiliation(s)
- Jinfu Zhou
- Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian Province, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Jianping Tang
- Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Chenran Zhang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Guilin Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Xinpei Lin
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Sining Liao
- Center for Disease Control and Prevention of Shantou, Shantou, Guangdong, 515000, China
| | - Jinying Luo
- Obstetrics and Gynecology Department, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian Province, China
| | - Guangxia Yu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Fuli Zheng
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Zhenkun Guo
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Wenya Shao
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Hong Hu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Liangpu Xu
- Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, 350001, Fujian Province, China.
| | - Siying Wu
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China.
| | - Huangyuan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China.
| |
Collapse
|
19
|
Wang X, Ren X, Lin X, Li Q, Zhang Y, Deng J, Chen B, Ru G, Luo Y, Lin N. Recent progress of ferroptosis in cancers and drug discovery. Asian J Pharm Sci 2024; 19:100939. [PMID: 39246507 PMCID: PMC11378902 DOI: 10.1016/j.ajps.2024.100939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 03/08/2024] [Accepted: 04/30/2024] [Indexed: 09/10/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron dependence and lipid peroxidation. Ferroptosis is involved in a range of pathological processes, such as cancer. Many studies have confirmed that ferroptosis plays an essential role in inhibiting cancer cell proliferation. In addition, a series of small-molecule compounds have been developed, including erastin, RSL3, and FIN56, which can be used as ferroptosis inducers. The combination of ferroptosis inducers with anticancer drugs can produce a significant synergistic effect in cancer treatment, and patients treated with these combinations exhibit a better prognosis than patients receiving traditional therapy. Therefore, a thorough understanding of the roles of ferroptosis in cancer is of great significance for the treatment of cancer. This review mainly elaborates the molecular biological characteristics and mechanism of ferroptosis, summarizes the function of ferroptosis in cancer development and treatment,illustrates the application of ferroptosis in patient's prognosis prediction and drug discovery, and discusses the prospects of targeting ferroptosis.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Xinxin Ren
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China
- Clinical Research Center for Cancer of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China
| | - Xu Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qi Li
- Department of Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Yingqiong Zhang
- Department of Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Jun Deng
- Department of Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Binxin Chen
- Department of Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Guoqing Ru
- Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China
| | - Ying Luo
- Department of Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Nengming Lin
- Department of Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou 310024, China
| |
Collapse
|
20
|
Nalla LV, Khairnar A. Empagliflozin drives ferroptosis in anoikis-resistant cells by activating miR-128-3p dependent pathway and inhibiting CD98hc in breast cancer. Free Radic Biol Med 2024; 220:288-300. [PMID: 38734268 DOI: 10.1016/j.freeradbiomed.2024.05.018] [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: 11/15/2023] [Revised: 02/18/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024]
Abstract
A tumour suppressor miRNA, miR-128-3p, is widely involved in various biological processes and has been found to get downregulated in breast cancer patients. We previously published that ectopically expressed miR-128-3p suppressed migration, invasion, cell cycle arrest, and breast cancer stem cells. In the present study, we explored the role of Empagliflozin (EMPA) as a miR-128-3p functionality-mimicking drug in inducing ferroptosis by inhibiting CD98hc. Given that CD98hc is one of the proteins critical in triggering ferroptosis, we confirmed that miR-128-3p and EMPA inhibited SP1, leading to inhibition of CD98hc expression. Further, transfection with siCD98hc, miR-128-3p mimics, and inhibitors was performed to assess their involvement in the ferroptosis of anoikis-resistant cells. We proved that anoikis-resistant cells possess high ROS and iron levels. Further, miR-128-3p and EMPA treatments induced ferroptosis by inhibiting GSH and enzymatic activity of GPX4 and also induced lipid peroxidation. Moreover, EMPA suppressed bioluminescence of 4T1-Red-FLuc induced thoracic cavity, peritoneal tumour burden and lung nodules in an in-vivo metastatic model of breast cancer. Collectively, we revealed that EMPA sensitized the ECM detached cells to ferroptosis by synergically activating miR-128-3p and lowering the levels of SP1 and CD98hc, making it a potential adjunct drug for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Lakshmi Vineela Nalla
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat, India; Department of Pharmacology, GITAM School of Pharmacy, GITAM (Deemed to be University), Rushikonda, Visakhapatnam, Andhra Pradesh, 530045, India
| | - Amit Khairnar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat, India; International Clinical Research Center, St. Anne's University Hospital Brno, Brno, 602 00, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 62500, Czech Republic; International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 6250, Czech Republic.
| |
Collapse
|
21
|
Han X, Zhu Y, Ke J, Zhai Y, Huang M, Zhang X, He H, Zhang X, Zhao X, Guo K, Li X, Han Z, Zhang Y. Progression of m 6A in the tumor microenvironment: hypoxia, immune and metabolic reprogramming. Cell Death Discov 2024; 10:331. [PMID: 39033180 PMCID: PMC11271487 DOI: 10.1038/s41420-024-02092-2] [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: 12/24/2023] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024] Open
Abstract
Recently, N6-methyladenosine (m6A) has aroused widespread discussion in the scientific community as a mode of RNA modification. m6A comprises writers, erasers, and readers, which regulates RNA production, nuclear export, and translation and is very important for human health. A large number of studies have found that the regulation of m6A is closely related to the occurrence and invasion of tumors, while the homeostasis and function of the tumor microenvironment (TME) determine the occurrence and development of tumors to some extent. TME is composed of a variety of immune cells (T cells, B cells, etc.) and nonimmune cells (tumor-associated mesenchymal stem cells (TA-MSCs), cancer-associated fibroblasts (CAFs), etc.). Current studies suggest that m6A is involved in regulating the function of various cells in the TME, thereby affecting tumor progression. In this manuscript, we present the composition of m6A and TME, the relationship between m6A methylation and characteristic changes in TME, the role of m6A methylation in TME, and potential therapeutic strategies to provide new perspectives for better treatment of tumors in clinical work.
Collapse
Affiliation(s)
- Xuan Han
- First Clinical College of Changzhi Medical College, Changzhi, China
| | - Yu Zhu
- Linfen Central Hospital, Linfen, China
| | - Juan Ke
- Linfen Central Hospital, Linfen, China
| | | | - Min Huang
- Linfen Central Hospital, Linfen, China
| | - Xin Zhang
- Linfen Central Hospital, Linfen, China
| | | | | | | | | | | | - Zhongyu Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | |
Collapse
|
22
|
Zhao Z, Miao Z, Hou Y, Zhong Y, Zhang X, Fang X. A novel signature constructed by cuproptosis-related RNA methylation regulators suggesting downregulation of YTHDC2 may induce cuproptosis resistance in colorectal cancer. Int Immunopharmacol 2024; 139:112691. [PMID: 39029230 DOI: 10.1016/j.intimp.2024.112691] [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: 03/22/2024] [Revised: 06/28/2024] [Accepted: 07/12/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND A newly identified type of cell death due to intracellular copper accumulation is known as cuproptosis and RNA methylation is a post-transcriptional modification mechanism, both of which perform vital roles in the immune microenvironment of colorectal cancer (CRC), but the link between the two needs more research. METHODS TCGA database provided RNA-seq data and details clinically of CRC samples. Cuproptosis-related RNA methylation regulators (CRRMRs) were identified by correlation analysis. We screened 6 CRRMRs for prognostic model construction by employing LASSO-Cox regression analysis and calculated risk scores by CRRMRs (CuMS). GSE39582 and GSE38832 cohort were used as external validation sets. This research concentrated on the connection between the prognostic model and somatic mutation, anti-cancer drug sensitivity, immune infiltration, immune checkpoint expression. In addition, we investigated the differential expression of YTHDC2 in epithelial cell subpopulations by single-cell analysis with GSE166555, calculated cuproptosis scores and performed pathway enrichment. In vitro experiments were performed to explore the consequences of knockdown of YTHDC2 on CRC cell proliferation and migration, as well as changes in CRC cell viability in response to elesclomol after knockdown of YTHDC2. In vivo experiments, we constructed the cell line-derived xenograft model to further validate the results of the in vitro experiments. RESULTS The prognosis of CRC can be predicted by CuMS, which GSE39582 and GSE38832 confirmed. Two CuMS groups showed different tumor mutation burden (TMB) and immune infiltration. CuMS was connected to emerging immune checkpoints CD47 and PVR, therefore, it can be clinically complementary to TMB and microsatellite instability (MSI) status. In single-cell analysis, a subpopulation of epithelial cells with high YTHDC2 expression had a high cuproptosis score. In vitro experiments, knocking down YTHDC2 promoted cell proliferation and migration in CRC, and weaken the inhibitory effect of elesclomol and elesclomol-Cu on cell viability, which in vivo experiments validated. CONCLUSION We developed a prognostic model constructed by 6 CRRMRs to assess overall survival and immune microenvironment of CRC patients. YTHDC2 might regulate cuproptosis in multiple ways.
Collapse
Affiliation(s)
- Zhongkai Zhao
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, No. 126 Sendai Street, Changchun, Jilin, China.
| | - Zeyu Miao
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, Jilin, China.
| | - Yuyang Hou
- Department of Immunology, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, Jilin, China.
| | - Yifan Zhong
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, Jilin, China.
| | - Xiaorong Zhang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun, Jilin, China.
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, No. 126 Sendai Street, Changchun, Jilin, China.
| |
Collapse
|
23
|
Yang Z, Verghese M, Yang S, Shah P, He YY. The m 6A reader YTHDC2 regulates UVB-induced DNA damage repair and histone modification. Photochem Photobiol 2024; 100:1031-1040. [PMID: 38190286 PMCID: PMC11228125 DOI: 10.1111/php.13904] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024]
Abstract
Ultraviolet B (UVB) radiation represents a major carcinogen for the development of all skin cancer types. Mechanistically, UVB induces damage to DNA in the form of lesions, including cyclobutane pyrimidine dimers (CPDs). Disruption of the functional repair processes, such as nucleotide excision repair (NER), allows persistence of DNA damage and contributes to skin carcinogenesis. Recent work has implicated m6A RNA methylation and its regulatory proteins as having critical roles in facilitating UVB-induced DNA damage repair. However, the biological functions of the m6A reader YTHDC2 are unknown in this context. Here, we show that YTHDC2 inhibition enhances the repair of UVB-induced DNA damage. We discovered that YTHDC2 inhibition increased the expression of PTEN while it decreased the expression of the PRC2 component SUZ12 and the levels of the histone modification H3K27me3. However, none of these functions were causally linked to the improvements in DNA repair, suggesting that the mechanism utilized by YTHDC2 may be unconventional. Moreover, inhibition of the m6A writer METTL14 reversed the effect of YTHDC2 inhibition on DNA repair while inhibition of the m6A eraser FTO mimicked the effect of YTHDC2 inhibition, indicating that YTHDC2 may regulate DNA repair through the m6A pathway. Finally, compared to normal human skin, YTHDC2 expression was upregulated in human cutaneous squamous cell carcinomas (cSCC), suggesting that it may function as a tumor-promoting factor in skin cancer. Taken together, our findings demonstrate that the m6A reader YTHDC2 plays a role in regulating UVB-induced DNA damage repair and may serve as a potential biomarker in cSCC.
Collapse
Affiliation(s)
- Zizhao Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
- These authors contributed equally to this work
| | - Michelle Verghese
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
- These authors contributed equally to this work
| | - Seungwon Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
| | - Palak Shah
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, Illinois, USA
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
24
|
Huang Z, Lin G, Hong Y, Weng L, Zhu K, Zhuang W. High expression of AlkB homolog 5 suppresses the progression of non-small cell lung cancer by facilitating ferroptosis through m6A demethylation of SLC7A11. ENVIRONMENTAL TOXICOLOGY 2024; 39:4035-4046. [PMID: 38642004 DOI: 10.1002/tox.24272] [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: 11/27/2023] [Revised: 03/17/2024] [Accepted: 03/23/2024] [Indexed: 04/22/2024]
Abstract
OBJECTIVE Non-small cell lung cancer (NSCLC) is a prevailing LC characterized by poor outcomes. AlkB homolog 5 (ALKBH5) functions as a tumor suppressor in several cancers. This study delved into the role of ALKBH5 in NSCLC development. METHODS TCGA database predicted ALKBH5 expression in NSCLC patients. ALKBH5 levels in NSCLC and human bronchial epithelial cells were determined. pcDNA3.1-ALKBH5/NC, pcDNA3.1-SLC7A11/NC, and ferrostatin-1 were used to explore the interactions among ALKBH5, SLC7A11, and ferroptosis. SLC7A11 mRNA and its protein levels were measured by RT-qPCR and Western blot. Cell viability, apoptosis, migration, and invasion were assessed by CCK-8, flow cytometry, and Transwell. Total N6-methyladenosine (m6A) quantification and its enrichment on SLC7A11 mRNA were determined, followed by the observation of Ki67, ALKBH5 and SLC7A11-positive cell numbers. Glutathione (GSH), lipid reactive oxygen species (lipid-ROS), malondialdehyde (MDA), and iron ion contents were determined. Animal experiments further analyzed the role of ALKBH5 in tumor development and glutathione peroxidase 4 (GPX4) expression. RESULTS Bioinformatics analysis revealed the lowly-expressed ALKBH5 in LC patients. ALKBH5 was downregulated in NSCLC cells and its upregulation repressed proliferation activity, invasion, and migration, and facilitated apoptosis. ALKBH5 upregulation decreased GSH, increased lipid-ROS, MDA, and iron ion contents, and downregulated SLC7A11 by reducing m6A modification. SLC7A11 upregulation partly annulled the effect of ALKBH5 overexpression on cell ferroptosis and malignant behaviors. In vivo assays elucidated the suppression of ALKBH5 upregulation on tumor development and GPX4 levels. CONCLUSION ALKBH5 upregulation downregulates SLC7A11 transcription by decreasing m6A modification, thus promoting NSCLC cell ferroptosis and ultimately repressing NSCLC progression.
Collapse
Affiliation(s)
- Zhangzhou Huang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Gen Lin
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yaping Hong
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Lihong Weng
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Kai Zhu
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Wu Zhuang
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
25
|
Hushmandi K, Einollahi B, Saadat SH, Lee EHC, Farani MR, Okina E, Huh YS, Nabavi N, Salimimoghadam S, Kumar AP. Amino acid transporters within the solute carrier superfamily: Underappreciated proteins and novel opportunities for cancer therapy. Mol Metab 2024; 84:101952. [PMID: 38705513 PMCID: PMC11112377 DOI: 10.1016/j.molmet.2024.101952] [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: 03/21/2024] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND Solute carrier (SLC) transporters, a diverse family of membrane proteins, are instrumental in orchestrating the intake and efflux of nutrients including amino acids, vitamins, ions, nutrients, etc, across cell membranes. This dynamic process is critical for sustaining the metabolic demands of cancer cells, promoting their survival, proliferation, and adaptation to the tumor microenvironment (TME). Amino acids are fundamental building blocks of cells and play essential roles in protein synthesis, nutrient sensing, and oncogenic signaling pathways. As key transporters of amino acids, SLCs have emerged as crucial players in maintaining cellular amino acid homeostasis, and their dysregulation is implicated in various cancer types. Thus, understanding the intricate connections between amino acids, SLCs, and cancer is pivotal for unraveling novel therapeutic targets and strategies. SCOPE OF REVIEW In this review, we delve into the significant impact of amino acid carriers of the SLCs family on the growth and progression of cancer and explore the current state of knowledge in this field, shedding light on the molecular mechanisms that underlie these relationships and highlighting potential avenues for future research and clinical interventions. MAJOR CONCLUSIONS Amino acids transportation by SLCs plays a critical role in tumor progression. However, some studies revealed the tumor suppressor function of SLCs. Although several studies evaluated the function of SLC7A11 and SLC1A5, the role of some SLC proteins in cancer is not studied well. To exert their functions, SLCs mediate metabolic rewiring, regulate the maintenance of redox balance, affect main oncogenic pathways, regulate amino acids bioavailability within the TME, and alter the sensitivity of cancer cells to therapeutics. However, different therapeutic methods that prevent the function of SLCs were able to inhibit tumor progression. This comprehensive review provides insights into a rapidly evolving area of cancer biology by focusing on amino acids and their transporters within the SLC superfamily.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
26
|
Wei Q, Xue C, Li M, Wei J, Zheng L, Chen S, Duan Y, Deng H, Tang F, Xiong W, Zhou M. Ferroptosis: a critical mechanism of N 6-methyladenosine modification involved in carcinogenesis and tumor progression. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1119-1132. [PMID: 38811442 DOI: 10.1007/s11427-023-2474-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/23/2023] [Indexed: 05/31/2024]
Abstract
Ferroptosis is an iron-dependent regulatory cell necrosis induced by iron overload and lipid peroxidation. It occurs when multiple redox-active enzymes are ectopically expressed or show abnormal function. Hence, the precise regulation of ferroptosis-related molecules is mediated across multiple levels, including transcriptional, posttranscriptional, translational, and epigenetic levels. N6-methyladenosine (m6A) is a highly evolutionarily conserved epigenetic modification in mammals. The m6A modification is commonly linked to tumor proliferation, progression, and therapy resistance because it is involved in RNA metabolic processes. Intriguingly, accumulating evidence suggests that dysregulated ferroptosis caused by the m6A modification drives tumor development. In this review, we summarized the roles of m6A regulators in ferroptosis-mediated malignant tumor progression and outlined the m6A regulatory mechanism involved in ferroptosis pathways. We also analyzed the potential value and application strategies of targeting m6A/ferroptosis pathway in the clinical diagnosis and therapy of tumors.
Collapse
Affiliation(s)
- Qingqing Wei
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, China
| | - Changning Xue
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, China
| | - Mengna Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, China
| | - Jianxia Wei
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, China
| | - Lemei Zheng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, China
- Hunan Key Laboratory of Oncotarget Gene, Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Shipeng Chen
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, China
| | - Yumei Duan
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, China
| | - Hongyu Deng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- Hunan Key Laboratory of Oncotarget Gene, Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Faqing Tang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- Hunan Key Laboratory of Oncotarget Gene, Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China.
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, China.
- Hunan Key Laboratory of Oncotarget Gene, Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China.
| |
Collapse
|
27
|
Wu X, Chen H, Li K, Zhang H, Li K, Tan H. The biological function of the N6-Methyladenosine reader YTHDC2 and its role in diseases. J Transl Med 2024; 22:490. [PMID: 38790013 PMCID: PMC11119022 DOI: 10.1186/s12967-024-05293-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
N6-methyladenosine (m6A) stands as the most prevalent modified form of RNA in eukaryotes, pivotal in various biological processes such as regulating RNA stability, translation, and transcription. All members within the YT521-B homology (YTH) gene family are categorized as m6A reading proteins, capable of identifying and binding m6A modifications on RNA, thereby regulating RNA metabolism and functioning across diverse physiological processes. YTH domain-containing 2 (YTHDC2), identified as the latest member of the YTH family, has only recently started to emerge for its biological function. Numerous studies have underscored the significance of YTHDC2 in human physiology, highlighting its involvement in both tumor progression and non-tumor diseases. Consequently, this review aims to further elucidate the pathological mechanisms of YTHDC2 by summarizing its functions and roles in tumors and other diseases, with a particular focus on its downstream molecular targets and signaling pathways.
Collapse
Affiliation(s)
- Xudong Wu
- Department of Thoracic Surgery, The Third Hospital of Changsha, Changsha, 410015, Hunan, People's Republic of China
| | - Hui Chen
- Department of Thoracic Surgery, The Third Hospital of Changsha, Changsha, 410015, Hunan, People's Republic of China
| | - Kai Li
- Department of Thoracic Surgery, The Third Hospital of Changsha, Changsha, 410015, Hunan, People's Republic of China
| | - Hong Zhang
- Department of Thoracic Surgery, The Third Hospital of Changsha, Changsha, 410015, Hunan, People's Republic of China
| | - Kai Li
- Department of Thoracic Surgery, Xiangxi Autonomous Prefecture People's Hospital, Jishou, 410015, Hunan, People's Republic of China
| | - Haoyu Tan
- Department of Cardio-vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
28
|
Han J, Wang C, Yang H, Luo J, Zhang X, Zhang XA. Novel Insights into the Links between N6-Methyladenosine and Regulated Cell Death in Musculoskeletal Diseases. Biomolecules 2024; 14:514. [PMID: 38785921 PMCID: PMC11117795 DOI: 10.3390/biom14050514] [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: 03/29/2024] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/25/2024] Open
Abstract
Musculoskeletal diseases (MSDs), including osteoarthritis (OA), osteosarcoma (OS), multiple myeloma (MM), intervertebral disc degeneration (IDD), osteoporosis (OP), and rheumatoid arthritis (RA), present noteworthy obstacles associated with pain, disability, and impaired quality of life on a global scale. In recent years, it has become increasingly apparent that N6-methyladenosine (m6A) is a key regulator in the expression of genes in a multitude of biological processes. m6A is composed of 0.1-0.4% adenylate residues, especially at the beginning of 3'-UTR near the translation stop codon. The m6A regulator can be classified into three types, namely the "writer", "reader", and "eraser". Studies have shown that the epigenetic modulation of m6A influences mRNA processing, nuclear export, translation, and splicing. Regulated cell death (RCD) is the autonomous and orderly death of cells under genetic control to maintain the stability of the internal environment. Moreover, distorted RCDs are widely used to influence the course of various diseases and receiving increasing attention from researchers. In the past few years, increasing evidence has indicated that m6A can regulate gene expression and thus influence different RCD processes, which has a central role in the etiology and evolution of MSDs. The RCDs currently confirmed to be associated with m6A are autophagy-dependent cell death, apoptosis, necroptosis, pyroptosis, ferroptosis, immunogenic cell death, NETotic cell death and oxeiptosis. The m6A-RCD axis can regulate the inflammatory response in chondrocytes and the invasive and migratory of MM cells to bone remodeling capacity, thereby influencing the development of MSDs. This review gives a complete overview of the regulatory functions on the m6A-RCD axis across muscle, bone, and cartilage. In addition, we also discuss recent advances in the control of RCD by m6A-targeted factors and explore the clinical application prospects of therapies targeting the m6A-RCD in MSD prevention and treatment. These may provide new ideas and directions for understanding the pathophysiological mechanism of MSDs and the clinical prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang 110100, China; (J.H.); (C.W.)
| | - Cuijing Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang 110100, China; (J.H.); (C.W.)
| | - Haolin Yang
- College of Pharmacy, Jilin University, Changchun 132000, China;
| | - Jiayi Luo
- College of Exercise and Health, Shenyang Sport University, Shenyang 110100, China; (J.H.); (C.W.)
| | - Xiaoyi Zhang
- College of Second Clinical Medical, China Medical University, Shenyang 110100, China;
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang 110100, China; (J.H.); (C.W.)
| |
Collapse
|
29
|
Qin L, Zhong Y, Li Y, Yang Y. TCM targets ferroptosis: potential treatments for cancer. Front Pharmacol 2024; 15:1360030. [PMID: 38738174 PMCID: PMC11082647 DOI: 10.3389/fphar.2024.1360030] [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: 12/22/2023] [Accepted: 04/08/2024] [Indexed: 05/14/2024] Open
Abstract
Ferroptosis is caused by the accumulation of cellular reactive oxygen species that exceed the antioxidant load that glutathione (GSH) and phospholipid hydroperoxidases with GSH-based substrates can carry When the antioxidant capacity of cells is reduced, lipid reactive oxygen species accumulate, which can cause oxidative death. Ferroptosis, an iron-dependent regulatory necrosis pathway, has emerged as a new modality of cell death that is strongly associated with cancer. Surgery, chemotherapy and radiotherapy are the main methods of cancer treatment. However, resistance to these mainstream anticancer drugs and strong toxic side effects have forced the development of alternative treatments with high efficiency and low toxicity. In recent years, an increasing number of studies have shown that traditional Chinese medicines (TCMs), especially herbs or herbal extracts, can inhibit tumor cell growth and metastasis by inducing ferroptosis, suggesting that they could be promising agents for cancer treatment. This article reviews the current research progress on the antitumor effects of TCMs through the induction of ferroptosis. The aim of these studies was to elucidate the potential mechanisms of targeting ferroptosis in cancer, and the findings could lead to new directions and reference values for developing better cancer treatment strategies.
Collapse
Affiliation(s)
- Liwen Qin
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Yuhan Zhong
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Li
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Center of Precision Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yongfeng Yang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Center of Precision Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Mete M, Ojha A, Dhar P, Das D. Deciphering Ferroptosis: From Molecular Pathways to Machine Learning-Guided Therapeutic Innovation. Mol Biotechnol 2024:10.1007/s12033-024-01139-0. [PMID: 38613722 DOI: 10.1007/s12033-024-01139-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/11/2024] [Indexed: 04/15/2024]
Abstract
Ferroptosis is a unique form of cell death reliant on iron and lipid peroxidation. It disrupts redox balance, causing cell death by damaging the plasma membrane, with inducers acting through enzymatic pathways or transport systems. In cancer treatment, suppressing ferroptosis or circumventing it holds significant promise. Beyond cancer, ferroptosis affects aging, organs, metabolism, and nervous system. Understanding ferroptosis mechanisms holds promise for uncovering novel therapeutic strategies across a spectrum of diseases. However, detection and regulation of this regulated cell death are still mired with challenges. The dearth of cell, tissue, or organ-specific biomarkers muted the pharmacological use of ferroptosis. This review covers recent studies on ferroptosis, detailing its properties, key genes, metabolic pathways, and regulatory networks, emphasizing the interaction between cellular signaling and ferroptotic cell death. It also summarizes recent findings on ferroptosis inducers, inhibitors, and regulators, highlighting their potential therapeutic applications across diseases. The review addresses challenges in utilizing ferroptosis therapeutically and explores the use of machine learning to uncover complex patterns in ferroptosis-related data, aiding in the discovery of biomarkers, predictive models, and therapeutic targets. Finally, it discusses emerging research areas and the importance of continued investigation to harness the full therapeutic potential of targeting ferroptosis.
Collapse
Affiliation(s)
- Megha Mete
- Department of Bioengineering, National Institute of Technology Agartala, Agartala, Tripura, 799046, India
| | - Amiya Ojha
- Department of Bioengineering, National Institute of Technology Agartala, Agartala, Tripura, 799046, India
| | - Priyanka Dhar
- CSIR-Indian Institute of Chemical Biology, Kolkata, 700032, India
| | - Deeplina Das
- Department of Bioengineering, National Institute of Technology Agartala, Agartala, Tripura, 799046, India.
| |
Collapse
|
31
|
Li Y, Li X, Li J. Ferroptosis in lung cancer: dual role, multi-level regulation, and new therapeutic strategies. Front Oncol 2024; 14:1360638. [PMID: 38515565 PMCID: PMC10955378 DOI: 10.3389/fonc.2024.1360638] [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: 12/23/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Lung cancer is a highly prevalent malignant tumor worldwide, with high incidence and death rates. Recently, there has been increasing recognition of the role of ferroptosis, a unique cell death mechanism, in lung cancer. This review aims to summarize the current research progress on the relationship between ferroptosis and lung cancer. It also provides a comprehensive analysis of the regulatory processes of ferroptosis in various stages, including epigenetics, transcription, post-transcription, translation, and post-translation. Additionally, the review explores the dual nature of ferroptosis in lung cancer progression, which presents interesting therapeutic possibilities. On one hand, ferroptosis can promote the escape of immune surveillance and reduce the efficacy of treatment in the early stages of tumors. On the other hand, it can counter drug resistance, enhance radiosensitivity, and promote immunotherapy. The article also discusses various combination treatment strategies based on the mechanism of ferroptosis. Overall, this review offers a holistic perspective on the role of ferroptosis in the onset, progression, and treatment of lung cancer. It aims to contribute to future research and clinical interventions in this field.
Collapse
Affiliation(s)
| | | | - Jian Li
- Department of Thoracic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
32
|
Arabpour J, Rezaei K, Khojini JY, Razi S, Hayati MJ, Gheibihayat SM. The potential role and mechanism of circRNAs in Ferroptosis: A comprehensive review. Pathol Res Pract 2024; 255:155203. [PMID: 38368664 DOI: 10.1016/j.prp.2024.155203] [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: 12/22/2023] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/20/2024]
Abstract
Cell death encompasses various mechanisms, including necrosis and apoptosis. Ferroptosis, a unique form of regulated cell death, emerged as a non-apoptotic process reliant on iron and reactive oxygen species (ROS). Distinguishing itself from other forms of cell death, ferroptosis exhibits distinct morphological, biochemical, and genetic features. Circular RNAs (circRNAs), a novel class of RNA molecules, play crucial regulatory roles in ferroptosis-mediated pathways and cellular processes. With their circular structure and stability, circRNAs function as microRNA sponges and participate in protein regulation, offering diverse mechanisms for cellular control. Accumulating evidence indicates that circRNAs are key players in diseases associated with ferroptosis, presenting opportunities for diagnostic and therapeutic applications. This study explores the regulatory roles of circRNAs in ferroptosis and their potential in diseases such as cancer, neurological disorders, and cardiovascular diseases. By investigating the relationship between circRNAs and ferroptosis, this research provides new insights into the diagnosis, treatment, and prognosis of ferroptosis-related diseases. Furthermore, the therapeutic implications of targeting circRNAs in cancer treatment and the modulation of ferroptosis pathways demonstrate the potential of circRNAs as diagnostic markers and therapeutic targets. Overall, understanding the involvement of circRNAs in regulating ferroptosis opens up new avenues for advancements in disease management.
Collapse
Affiliation(s)
- Javad Arabpour
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Kimia Rezaei
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Shokufeh Razi
- Department of Genetics, Faculty of Basic Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Javad Hayati
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Seyed Mohammad Gheibihayat
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
33
|
Xia R, Peng HF, Zhang X, Zhang HS. Comprehensive review of amino acid transporters as therapeutic targets. Int J Biol Macromol 2024; 260:129646. [PMID: 38272411 DOI: 10.1016/j.ijbiomac.2024.129646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
The solute carrier (SLC) family, with more than 400 membrane-bound proteins, facilitates the transport of a wide array of substrates such as nutrients, ions, metabolites, and drugs across biological membranes. Amino acid transporters (AATs) are membrane transport proteins that mediate transfer of amino acids into and out of cells or cellular organelles. AATs participate in many important physiological functions including nutrient supply, metabolic transformation, energy homeostasis, redox regulation, and neurological regulation. Several AATs have been found to significantly impact the progression of human malignancies, and dysregulation of AATs results in metabolic reprogramming affecting tumor growth and progression. However, current clinical therapies that directly target AATs have not been developed. The purpose of this review is to highlight the structural and functional diversity of AATs, the molecular mechanisms in human diseases such as tumors, kidney diseases, and emerging therapeutic strategies for targeting AATs.
Collapse
Affiliation(s)
- Ran Xia
- College of Chemistry and Life Science, Beijing University of Technology, Pingleyuan 100(#), District of Chaoyang, Beijing 100124, China
| | - Hai-Feng Peng
- College of Chemistry and Life Science, Beijing University of Technology, Pingleyuan 100(#), District of Chaoyang, Beijing 100124, China
| | - Xing Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Pingleyuan 100(#), District of Chaoyang, Beijing 100124, China
| | - Hong-Sheng Zhang
- College of Chemistry and Life Science, Beijing University of Technology, Pingleyuan 100(#), District of Chaoyang, Beijing 100124, China.
| |
Collapse
|
34
|
Kong J, Lyu H, Ouyang Q, Shi H, Zhang R, Xiao S, Guo D, Zhang Q, Chen XZ, Zhou C, Tang J. Insights into the Roles of Epigenetic Modifications in Ferroptosis. BIOLOGY 2024; 13:122. [PMID: 38392340 PMCID: PMC10886775 DOI: 10.3390/biology13020122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
Ferroptosis is a non-apoptotic mode of cell death driven by membrane lipid peroxidation and is characterized by elevated intracellular levels of Fe2+, ROS, and lipid peroxidation. Studies have shown that ferroptosis is related to the development of multiple diseases, such as cancer, neurodegenerative diseases, and acute myeloid leukemia. Ferroptosis plays a dual role in the occurrence and development of these diseases. Ferroptosis mainly involves iron metabolism, ROS, and lipid metabolism. Various mechanisms, including epigenetic regulation, have been reported to be deeply involved in ferroptosis. Abnormal epigenetic modifications have been reported to promote tumor onset or other diseases and resistance to chemotherapy drugs. In recent years, diversified studies have shown that epigenetic modification is involved in ferroptosis. In this review, we reviewed the current resistance system of ferroptosis and the research progress of epigenetic modification, such as DNA methylation, RNA methylation, non-coding RNAs, and histone modification in cancer and other diseases by regulating ferroptosis.
Collapse
Affiliation(s)
- Jinghua Kong
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Qian Ouyang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Hao Shi
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Rui Zhang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Shuai Xiao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Dong Guo
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Qi Zhang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
35
|
Shi X, Yang J, Wang M, Xia L, Zhang L, Qiao S. Hsa_circ_0050900 affects ferroptosis in intrahepatic cholangiocarcinoma cells by targeting hsa‑miR-605‑3p to regulate SLC3A2. Oncol Lett 2024; 27:2. [PMID: 38028176 PMCID: PMC10665981 DOI: 10.3892/ol.2023.14135] [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: 10/11/2022] [Accepted: 09/20/2023] [Indexed: 12/01/2023] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a highly lethal hepatobiliary tumor with high aggressiveness. The role of circular RNA (circRNA) in ICC remains to be explored. The present study aimed to investigate whether hsa_circ_0050900 affected ferroptosis in ICC cells by regulating hsa-microRNA (miR)-605-3p/solute carrier family 3 member 2 (SLC3A2). Human ICC cells were cultured and hsa_circ_0050900 expression was evaluated by reverse transcription-quantitative PCR. hsa_circ_0050900 was knocked down and ferroptosis inhibitor ferrostatin-1 was added to HuCCT-1 cells. Following knockdown or overexpression of hsa-miR-605-3p, Fe2+, reactive oxygen species (ROS), glutathione peroxidase 4 and SLC3A2 levels were assessed using iron and ROS assay kit or RT-qPCR and western blotting, respectively. Cell function experiments were performed to examine proliferation and migration abilities. Dual-luciferase reporter gene and argonaute2-RNA immunoprecipitation assay verified the relationship among hsa_circ_0050900, hsa-miR-605-3p, and SLC3A2. hsa_circ_0050900 was derived from actinin alpha 4 gene and was elevated in ICC cells. Among HuCCT-1, QBC-939, HCCC-9810, and RBE cell lines, the highest expression was in HuCCT-1 cells. Inhibition of hsa_circ_0050900 inhibited proliferation and migration by facilitating ICC cell ferroptosis. hsa-miR-605-3p expression was elevated after knocking down hsa_circ_0050900 and hsa-miR-605-3p was negatively regulated by hsa_circ_0050900. In addition, hsa-miR-605-3p targeted SLC3A2. Overexpression of hsa-miR-605-3p regulated SLC3A2 to promote ICC cell ferroptosis and inhibit proliferation and migration. Taken together, knockdown of hsa_circ_0050900 inhibited SLC3A2 expression via sponging hsa-miR-605-3p to promote ICC cell ferroptosis, and finally suppressed proliferation and migration. The present study suggested that hsa_circ_0050900 was a potential therapeutic target for ICC.
Collapse
Affiliation(s)
- Xiangtian Shi
- Department of Hepatobiliary Surgery, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Jiarui Yang
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong 523058, P.R. China
- Department of Pancreatic Hepato-Biliary-Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Meng Wang
- Department of Hepatobiliary Surgery, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Long Xia
- Department of Hepatobiliary-Pancreatic-Splenic Surgery, Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia Autonomous Region 010017, P.R. China
| | - Lei Zhang
- Department of Pancreatic Hepato-Biliary-Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Shan Qiao
- Department of Hepatobiliary Surgery, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| |
Collapse
|
36
|
Liu W, Zeng H. IGF2BP2 attenuates intestinal epithelial cell ferroptosis in colitis by stabilizing m 6A-modified GPX4 mRNA. Cytokine 2024; 173:156388. [PMID: 38039694 DOI: 10.1016/j.cyto.2023.156388] [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: 08/14/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 12/03/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic and uncontrolled inflammatory bowel disease. N6-methyladenine (m6A) is a reversible mRNA modification method. IGF2BP2 is an RNA-binding protein regulated by m6A methylation. However, understanding of m6A-related proteins in UC is limited. This study was to analyze the function and related mechanism of IGF2BP2 in UC. METHODS The UC models were established by dextran sulfate sodium (DSS) in NCM460 cells and mice. The expression of IGF2BP2 and GPX4 in UC were detected by qPCR and western blot. The effects of IGF2BP2 on inflammation, ferroptosis and colon injury were measured by gain- and loss-of-function experiments. This study conducted a clinical evaluation of mice using the Disease Activity Index score. The molecular mechanism of IGF2BP2 in ferroptosis were analyzed by m6A RNA methylation quantification kit, RNA immunoprecipitation-qPCR analysis, and RNA stability assay. RESULTS IGF2BP2 and GPX4 were under-expressed in DSS-treated UC. IGF2BP2 enhanced the stability of GPX4 mRNA modified by m6A. IGF2BP2 overexpression repressed the ROS, MDA, and iron levels but enhanced the GSH and GPX4 levels in DSS-triggered NCM460 cells, which were partially reversed by GPX4 silencing. In UC mice, IGF2BP2 high-expression ameliorated symptoms, Disease Activity Index score, pathological changes, inflammatory reaction, and ferroptosis, which were also partly neutralized by GPX4 inhibition. CONCLUSIONS IGF2BP2 augmented the GPX4 expression by the m6A modification to weaken UC progression via suppressing ferroptosis.
Collapse
Affiliation(s)
- Wei Liu
- Department of Pediatric Gastroenterology, Children's Hospital of The First People's Hospital of Chenzhou City, Chenzhou 423000, Hunan Province, PR China
| | - Hui Zeng
- Department of Pediatric Gastroenterology, Children's Hospital of The First People's Hospital of Chenzhou City, Chenzhou 423000, Hunan Province, PR China.
| |
Collapse
|
37
|
Zhang G, Mi W, Wang C, Li J, Zhang Y, Liu N, Jiang M, Jia G, Wang F, Yang G, Zhang L, Wang J, Fu Y, Zhang Y. Targeting AKT induced Ferroptosis through FTO/YTHDF2-dependent GPX4 m6A methylation up-regulating and degradating in colorectal cancer. Cell Death Discov 2023; 9:457. [PMID: 38102129 PMCID: PMC10724184 DOI: 10.1038/s41420-023-01746-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023] Open
Abstract
Ferroptosis is a new type of iron-dependent programmed cell death induced by lipid peroxidation. However, the underlying mechanisms and function in tumor therapy still remain undisclosed especially in post-transcription regulation. Here, we found that targeting AKT significantly induced GPX4 dependent ferroptosis and suppressed colorectal cancer growth both in vitro and in vivo. During this process, demethylase FTO was downregulated, which increased the m6A methylation level of GPX4, subsequently recognized by YTHDF2 and degraded. Prediction results showed that there are three potential methylated sites (193/647/766), and 193 site was identified as the right one, which was demethylated by FTO and read by YTHDF2. In parallel, AKT inhibition caused the accumulation of ROS which had a negative feedback on GPX4 expression. In addition, protective autophagy was initiated by MK2206 stimulation, while blocking autophagy further increased ferroptosis and markedly enhanced the anti-tumor activity of MK2206. In a word, inhibiting AKT activated ferroptosis through FTO/YTHDF2/GPX4 axis to suppress colon cancer progression, which raised FTO/GPX4 as potential biomarkers and targets in colorectal cancer therapy.
Collapse
Affiliation(s)
- Ge Zhang
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Wunan Mi
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
- Department of Surgery, Erasmus MC Transplant Institute, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Chuyue Wang
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Jiehan Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yizheng Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Nannan Liu
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Meimei Jiang
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Guiyun Jia
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Feng Wang
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, 200072, China
| | - Ge Yang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Lingling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jiangang Wang
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Yingjie Zhang
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, China.
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China.
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China.
| |
Collapse
|
38
|
Wang Y, Hu J, Wu S, Fleishman JS, Li Y, Xu Y, Zou W, Wang J, Feng Y, Chen J, Wang H. Targeting epigenetic and posttranslational modifications regulating ferroptosis for the treatment of diseases. Signal Transduct Target Ther 2023; 8:449. [PMID: 38072908 PMCID: PMC10711040 DOI: 10.1038/s41392-023-01720-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/16/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Ferroptosis, a unique modality of cell death with mechanistic and morphological differences from other cell death modes, plays a pivotal role in regulating tumorigenesis and offers a new opportunity for modulating anticancer drug resistance. Aberrant epigenetic modifications and posttranslational modifications (PTMs) promote anticancer drug resistance, cancer progression, and metastasis. Accumulating studies indicate that epigenetic modifications can transcriptionally and translationally determine cancer cell vulnerability to ferroptosis and that ferroptosis functions as a driver in nervous system diseases (NSDs), cardiovascular diseases (CVDs), liver diseases, lung diseases, and kidney diseases. In this review, we first summarize the core molecular mechanisms of ferroptosis. Then, the roles of epigenetic processes, including histone PTMs, DNA methylation, and noncoding RNA regulation and PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, methylation, and ADP-ribosylation, are concisely discussed. The roles of epigenetic modifications and PTMs in ferroptosis regulation in the genesis of diseases, including cancers, NSD, CVDs, liver diseases, lung diseases, and kidney diseases, as well as the application of epigenetic and PTM modulators in the therapy of these diseases, are then discussed in detail. Elucidating the mechanisms of ferroptosis regulation mediated by epigenetic modifications and PTMs in cancer and other diseases will facilitate the development of promising combination therapeutic regimens containing epigenetic or PTM-targeting agents and ferroptosis inducers that can be used to overcome chemotherapeutic resistance in cancer and could be used to prevent other diseases. In addition, these mechanisms highlight potential therapeutic approaches to overcome chemoresistance in cancer or halt the genesis of other diseases.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jing Hu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300060, PR China
| | - Shuang Wu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, PR China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yulin Li
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Yinshi Xu
- Department of Outpatient, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Wailong Zou
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Yukuan Feng
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China.
| | - Hongquan Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| |
Collapse
|
39
|
Zhang H, Wang SQ, Zhu JB, Wang LN, Lin H, Li LF, Cheng YD, Duan CJ, Zhang CF. LncRNA CALML3-AS1 modulated by m 6A modification induces BTNL9 methylation to drive non-small-cell lung cancer progression. Cancer Gene Ther 2023; 30:1649-1662. [PMID: 37884580 DOI: 10.1038/s41417-023-00670-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/31/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023]
Abstract
Non-small cell lung cancer (NSCLC) is a common and lethal malignancy. The carcinogenic roles of lncRNA CALML3 antisense RNA 1 (CALML3-AS1) have been documented. However, the function and potential mechanisms of CALML3-AS1 in the progression of NSCLC need to be further explored. The molecule expression was assessed by qRT-PCR and Western blot. The subcellular localization of CALML3-AS1 was observed by fluorescence in situ hybridization (FISH). The malignant behaviors of NSCLC cells were evaluated by CCK-8, colony formation, EdU, wound healing and transwell assays. In vivo xenograft tumor and liver metastatic models were established. The molecular mechanisms were investigated by RIP, RNA pull-down and ChIP assays. The methylation level was detected by MSP. Herein, we found that CALML3-AS1 was upregulated, while butyrophilin-like 9 (BTNL9) was downregulated in NSCLC. Functionally, CALML3-AS1 depletion repressed NSCLC cell malignant phenotypes, in vivo tumor growth, and liver metastasis. Mechanistically, AlkB homolog 5 (ALKBH5) enhanced CALML3-AS1 stability via N6-methyladenosine (m6A) demethylation, whereas m6A reader YTH domain-containing 2 (YTHDC2) destabilized CALML3-AS1. Moreover, CALML3-AS1 inhibited BTNL9 transcription and expression through the recruitment of Zeste homolog 2 (EZH2). Rescue experiments demonstrated that BTNL9 downregulation counteracted sh-CALML3-AS1-mediated antitumor effects on NSCLC. Taken together, CALML3-AS1 modulated by ALKBH5 and YTHDC2 in an m6A modification dependent manner drives NSCLC progression via epigenetically repressing BTNL9.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P. R. China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P. R. China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan Province, P. R. China.
| | - Shao-Qiang Wang
- Department of Thoracic Surgery, Weifang People's Hospital, Weifang Medical University, Weifang, 261041, Shandong Province, P.R. China
- Department of Scientific Research Management, Weifang People's Hospital, Weifang Medical University, Weifang, 261041, Shandong Province, P.R. China
| | - Jie-Bo Zhu
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P. R. China
| | - Li-Na Wang
- Department of Thoracic Surgery, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, P. R. China
| | - Hang Lin
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P. R. China
| | - Lin-Feng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P. R. China
| | - Yuan-Da Cheng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P. R. China
| | - Chao-Jun Duan
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P. R. China
| | - Chun-Fang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P. R. China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, P. R. China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan Province, P. R. China.
- Hunan Engineering Research Center for Pulmonary Nodules Precise Diagosis & Treatment, Changsha, 410008, Hunan Province, P. R. China.
| |
Collapse
|
40
|
Wang Z, Li H, Cai H, Liang J, Jiang Y, Song F, Hou C, Hou J. FTO Sensitizes Oral Squamous Cell Carcinoma to Ferroptosis via Suppressing ACSL3 and GPX4. Int J Mol Sci 2023; 24:16339. [PMID: 38003537 PMCID: PMC10671523 DOI: 10.3390/ijms242216339] [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: 09/22/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Ferroptosis is a newly established form of regulated cell death characterized by intracellular lipid peroxidation and iron accumulation that may be a promising cancer treatment strategy. However, the function and therapeutic value of ferroptosis in oral squamous cell carcinoma (OSCC) remain inadequately understood. In the present study, we investigated the biological role of the fat mass and obesity-associated gene (FTO) in ferroptosis in the context of OSCC. We found that OSCC had greater potential for ferroptosis, and FTO is associated with ferroptosis. Furthermore, higher FTO expression sensitized OSCC cells to ferroptosis in vitro and in vivo. Mechanistically, FTO suppressed the expression of anti-ferroptotic factors, acyl-CoA synthetase long-chain family member 3 (ACSL3) and glutathione peroxidase 4 (GPX4), by demethylating the m6A modification on the mRNA of ACSL3 and GPX4 and decreasing their stability. Taken together, our findings revealed that FTO promotes ferroptosis through ACSL3 and GPX4 regulation. Thus, ferroptosis activation in OSCC with high FTO levels may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan Road West, Guangzhou 510055, China; (Z.W.); (H.L.); (H.C.); (J.L.); (Y.J.); (F.S.); (C.H.)
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Hongyu Li
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan Road West, Guangzhou 510055, China; (Z.W.); (H.L.); (H.C.); (J.L.); (Y.J.); (F.S.); (C.H.)
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Hongshi Cai
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan Road West, Guangzhou 510055, China; (Z.W.); (H.L.); (H.C.); (J.L.); (Y.J.); (F.S.); (C.H.)
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Jianfeng Liang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan Road West, Guangzhou 510055, China; (Z.W.); (H.L.); (H.C.); (J.L.); (Y.J.); (F.S.); (C.H.)
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yaoqi Jiang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan Road West, Guangzhou 510055, China; (Z.W.); (H.L.); (H.C.); (J.L.); (Y.J.); (F.S.); (C.H.)
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Fan Song
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan Road West, Guangzhou 510055, China; (Z.W.); (H.L.); (H.C.); (J.L.); (Y.J.); (F.S.); (C.H.)
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Chen Hou
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan Road West, Guangzhou 510055, China; (Z.W.); (H.L.); (H.C.); (J.L.); (Y.J.); (F.S.); (C.H.)
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Jinsong Hou
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Lingyuan Road West, Guangzhou 510055, China; (Z.W.); (H.L.); (H.C.); (J.L.); (Y.J.); (F.S.); (C.H.)
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
41
|
Liu J, Han X, Zhou J, Leng Y. Molecular Mechanisms of Ferroptosis and Their Involvement in Acute Kidney Injury. J Inflamm Res 2023; 16:4941-4951. [PMID: 37936596 PMCID: PMC10627075 DOI: 10.2147/jir.s427505] [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: 06/26/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023] Open
Abstract
Ferroptosis is a novel way of regulating cell death, which occurs in a process that is closely linked to intracellular iron metabolism, lipid metabolism, amino acid metabolism, and multiple signaling pathways. The latest research shows that ferroptosis plays a key role in the pathogenesis of acute kidney injury (AKI). Ferroptosis may be an important target for treating AKI caused by various reasons, such as ischemia-reperfusion injury, rhabdomyolysis syndrome, sepsis, and nephrotoxic drugs. This paper provides a review on the regulatory mechanisms of ferroptosis and its role in AKI, which may help to provide new research ideas for the treatment of AKI and future research.
Collapse
Affiliation(s)
- Jie Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People’s Republic of China
| | - Xiaoxia Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People’s Republic of China
| | - Jia Zhou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People’s Republic of China
| | - Yufang Leng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, People’s Republic of China
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| |
Collapse
|
42
|
Wei X, Li X, Hu S, Cheng J, Cai R. Regulation of Ferroptosis in Lung Adenocarcinoma. Int J Mol Sci 2023; 24:14614. [PMID: 37834062 PMCID: PMC10572737 DOI: 10.3390/ijms241914614] [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: 08/31/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common lung cancer, which accounts for about 35-40% of all lung cancer patients. Despite therapeutic advancements in recent years, the overall survival time of LUAD patients still remains poor, especially KRAS mutant LUAD. Therefore, it is necessary to further explore novel targets and drugs to improve the prognos is for LUAD. Ferroptosis, an iron-dependent regulated cell death (RCD) caused by lipid peroxidation, has attracted much attention recently as an alternative target for apoptosis in LUAD therapy. Ferroptosis has been found to be closely related to LUAD at every stage, including initiation, proliferation, and progression. In this review, we will provide a comprehensive overview of ferroptosis mechanisms, its regulation in LUAD, and the application of targeting ferroptosis for LUAD therapy.
Collapse
Affiliation(s)
| | | | | | - Jinke Cheng
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (X.W.); (X.L.); (S.H.)
| | - Rong Cai
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (X.W.); (X.L.); (S.H.)
| |
Collapse
|
43
|
Xia P, Dubrovska A. CD98 heavy chain as a prognostic biomarker and target for cancer treatment. Front Oncol 2023; 13:1251100. [PMID: 37823053 PMCID: PMC10562705 DOI: 10.3389/fonc.2023.1251100] [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: 06/30/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023] Open
Abstract
The SLC3A2 gene encodes for a cell-surface transmembrane protein CD98hc (4F2). CD98hc serves as a chaperone for LAT1 (SLC7A5), LAT2 (SLC7A8), y+LAT1 (SLC7A7), y+LAT2 (SLC7A6), xCT (SLC7A11) and Asc1 (SLC7A10) providing their recruitment to the plasma membrane. Together with the light subunits, it constitutes heterodimeric transmembrane amino acid transporters. CD98hc interacts with other surface molecules, such as extracellular matrix metalloproteinase inducer CD147 (EMMPRIN) and adhesion receptors integrins, and regulates glucose uptake. In this way, CD98hc connects the signaling pathways sustaining cell proliferation and migration, biosynthesis and antioxidant defense, energy production, and stem cell properties. This multifaceted role makes CD98hc one of the critical regulators of tumor growth, therapy resistance, and metastases. Indeed, the high expression levels of CD98hc were confirmed in various tumor tissues, including head and neck squamous cell carcinoma, glioblastoma, colon adenocarcinoma, pancreatic ductal adenocarcinoma, and others. A high expression of CD98hc has been linked to clinical prognosis and response to chemo- and radiotherapy in several types of cancer. In this mini-review, we discuss the physiological functions of CD98hc, its role in regulating tumor stemness, metastases, and therapy resistance, and the clinical significance of CD98hc as a tumor marker and therapeutic target.
Collapse
Affiliation(s)
- Pu Xia
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Anna Dubrovska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden: German Cancer Research Center (DKFZ), Heidelberg, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
44
|
Wang F, Sun Z, Zhang Q, Yang H, Yang G, Yang Q, Zhu Y, Wu W, Xu W, Wu X. Curdione induces ferroptosis mediated by m6A methylation via METTL14 and YTHDF2 in colorectal cancer. Chin Med 2023; 18:122. [PMID: 37735401 PMCID: PMC10512537 DOI: 10.1186/s13020-023-00820-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: 03/22/2023] [Accepted: 08/11/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Curdione is a sesquiterpene isolated from Curcumae Rhizoma that possesses high biological activity and extensive pharmacological effects. As a traditional Chinese medicine, Curcumae Rhizoma can inhibit the development of many types of cancer, especially colorectal cancer. However, the anti-colorectal mechanism of its monomer curdione remains unclear. METHODS Colorectal cancer (CRC) cells were treated with curdione at doses of 12.5 μM, 25 μM, and 50 μM, and then the cells' activity was measured with methyl thiazolyl tetrazolium (MTT). Nude mice were administered different doses of curdione subcutaneously and oxaliplatin by tail vein injection, and then hematoxylin-eosin (HE) staining was adopted to examine tumor histology. Moreover, flow cytometry was applied to detect reactive oxygen species in cells and tissues. Kits were employed to detect the levels of iron ions, malondialdehyde, lipid hydroperoxide, and glutathione. Polymerase chain reaction (PCR) and Western blotting were adopted to detect ferroptosis and m6A modification-related factors. A methylation spot hybridization assay was performed to measure changes in overall methylation. SLC7A11 and HOXA13 were measured by MeRIP-qPCR. The shRNA-METTL14 plasmid was constructed to verify the inhibitory effect of curdione on CRC. RESULTS A dose-dependent decrease in activity was observed in curdione-treated cells. Curdione increased the accumulation of reactive oxygen species in CRC cells and tumor tissues, greatly enhanced the levels of malondialdehyde, lipid hydroperoxide and Fe2+, and lowered the activity of glutathione. According to the qPCR and Western blot results, curdione promoted the expression of METTL14 and YTHDF2 in CRC cells and tissues, respectively, and decreased the expression of SLC7A11, SLC3A2, HOXA13, and glutathione peroxidase 4. Additionally, in animal experiments, the curdione-treated group showed severe necrosis of tumor cells, as displayed by HE staining. Furthermore, compared with the control group, levels of m6A modifying factors (namely, SLC7A11 and HOXA13) were increased in the tissues after drug intervention. METTL14 knockdown was followed by an increase in CRC cell activity and glutathione levels. However, the levels of reactive oxygen species, malondialdehyde, and iron ions decreased. The expression levels of SLC7A11, SLC3A2, HOXA13, and GPX4 were all increased after METTL14 knockdown. CONCLUSION The results suggest that curdione induces ferroptosis in CRC by virtue of m6A methylation.
Collapse
Affiliation(s)
- Fang Wang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Zheng Sun
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Qunyao Zhang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Hao Yang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Gang Yang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Qi Yang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Yimiao Zhu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Wenya Wu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, China
| | - Wenwen Xu
- Department of Gynecology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| | - Xiaoyu Wu
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
45
|
Chen X, Zhang L, He Y, Huang S, Chen S, Zhao W, Yu D. Regulation of m 6A modification on ferroptosis and its potential significance in radiosensitization. Cell Death Discov 2023; 9:343. [PMID: 37714846 PMCID: PMC10504338 DOI: 10.1038/s41420-023-01645-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/28/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
Radiotherapy is often used to treat various types of cancers, but radioresistance greatly limits the clinical efficiency. Recent studies have shown that radiotherapy can lead to ferroptotic cancer cell deaths. Ferroptosis is a new type of programmed cell death caused by excessive lipid peroxidation. The induction of ferroptosis provides a potential therapeutic strategy for radioresistance. As the most common post-transcriptional modification of mRNA, m6A methylation is widely involved in the regulation of various physiopathological processes by regulating RNA function. Dynamic m6A modification controlled by m6A regulatory factors also affects the susceptibility of cells to ferroptosis, thereby determining the radiosensitivity of tumor cells to radiotherapy. In this review, we summarize the mechanism and significance of radiotherapy induced ferroptosis, analyze the regulatory characteristics of m6A modification on ferroptosis, and discuss the possibility of radiosensitization by enhancing m6A-mediated ferroptosis. Clarifying the regulation of m6A modification on ferroptosis and its significance in the response of tumor cells to radiotherapy will help us identify novel targets to improve the efficacy of radiotherapy and reduce or overcome radioresistance.
Collapse
Affiliation(s)
- Xun Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China
| | - Lejia Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China
| | - Yi He
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China
| | - Siyuan Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China
| | - Shangwu Chen
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory for Biocontrol, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China
| | - Wei Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China.
| | - Dongsheng Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China.
| |
Collapse
|
46
|
Zhang X, Ma L, Wang J. Cross-Regulation Between Redox and Epigenetic Systems in Tumorigenesis: Molecular Mechanisms and Clinical Applications. Antioxid Redox Signal 2023; 39:445-471. [PMID: 37265163 DOI: 10.1089/ars.2023.0253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Significance: Redox and epigenetics are two important regulatory processes of cell physiological functions. The cross-regulation between these processes has critical effects on the occurrence and development of various types of tumors. Recent Advances: The core factor that influences redox balance is reactive oxygen species (ROS) generation. The ROS functions as a double-edged sword in tumors: Low levels of ROS promote tumors, whereas excessive ROS induces various forms of tumor cell death, including apoptosis and ferroptosis as well as necroptosis and pyroptosis. Many studies have shown that the redox balance is influenced by epigenetic mechanisms such as DNA methylation, histone modification, chromatin remodeling, non-coding RNAs (microRNA, long non-coding RNA, and circular RNA), and RNA N6-methyladenosine modification. Several oxidizing or reducing substances also affect the epigenetic state. Critical Issues: In this review, we summarize research on the cross-regulation between redox and epigenetics in cancer and discuss the relevant molecular mechanisms. We also discuss the current research on the clinical applications. Future Directions: Future research can use high-throughput methods to analyze the molecular mechanisms of the cross-regulation between redox and epigenetics using both in vitro and in vivo models in more detail, elucidate regulatory mechanisms, and provide guidance for clinical treatment. Antioxid. Redox Signal. 39, 445-471.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Lifang Ma
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Jiayi Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
47
|
Zhang J, Qiu T, Yao X, Sun X. Insights into the role of N6-methyladenosine in ferroptosis. Biomed Pharmacother 2023; 165:115192. [PMID: 37487443 DOI: 10.1016/j.biopha.2023.115192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/03/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023] Open
Abstract
N6-methyladenosine (m6A) methylation modification is one of the most prevalent epigenetic modifications of eukaryotic RNA. m6A methylation is widely associated with many biological processes through the modification of RNA metabolism and is associated with multiple disease states. As a newly discovered regulatory cell death in recent years, ferroptosis is an iron-dependent cell death characterized by excessive lipid peroxidation. Emerging evidence supports that ferroptosis has a significant role in the progression of diverse diseases. Besides, the key regulators of ferroptosis exhibit aberrant m6A levels under different pathological conditions. However, the correlation between m6A-modified ferroptosis and multiple diseases has not been well elucidated. In this review, we summarized the functions of m6A in ferroptosis, which are associated with the initiation and progression of multiple diseases. Investigating the role of m6A in ferroptosis might both facilitate a better understanding of the pathogenesis of these diseases and provide new opportunities for targeted treatment.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Occupational and Environmental Health Department, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China.
| | - Tianming Qiu
- Occupational and Environmental Health Department, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China.
| | - Xiaofeng Yao
- Occupational and Environmental Health Department, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China.
| | - Xiance Sun
- Occupational and Environmental Health Department, School of Public Health, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian 116044, PR China; Global Health Research Center, Dalian Medical University, No. 9 West Section Lvshun South Road, Dalian, 116044, PR China.
| |
Collapse
|
48
|
Yang Z, Su W, Wei X, Qu S, Zhao D, Zhou J, Wang Y, Guan Q, Qin C, Xiang J, Zen K, Yao B. HIF-1α drives resistance to ferroptosis in solid tumors by promoting lactate production and activating SLC1A1. Cell Rep 2023; 42:112945. [PMID: 37542723 DOI: 10.1016/j.celrep.2023.112945] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/06/2023] [Accepted: 07/21/2023] [Indexed: 08/07/2023] Open
Abstract
Solid tumors have developed robust ferroptosis resistance. The mechanism underlying ferroptosis resistance regulation in solid tumors, however, remains elusive. Here, we report that the hypoxic tumor microenvironment potently promotes ferroptosis resistance in solid tumors in a hypoxia-inducible factor 1α (HIF-1α)-dependent manner. In combination with HIF-2α, which promotes tumor ferroptosis under hypoxia, HIF-1α is the main driver of hypoxia-induced ferroptosis resistance. Mechanistically, HIF-1α-induced lactate contributes to ferroptosis resistance in a pH-dependent manner that is parallel to the classical SLC7A11 and FSP1 systems. In addition, HIF-1α also enhances transcription of SLC1A1, an important glutamate transporter, and promotes cystine uptake to promote ferroptosis resistance. In support of the role of hypoxia in ferroptosis resistance, silencing HIF-1α sensitizes mouse solid tumors to ferroptosis inducers. In conclusion, our results reveal a mechanism by which hypoxia drives ferroptosis resistance and identify the combination of hypoxia alleviation and ferroptosis induction as a promising therapeutic strategy for solid tumors.
Collapse
Affiliation(s)
- Zhou Yang
- National Experimental Teaching Center of Basic Medical Science, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China; Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Su
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiyi Wei
- The State Key Lab of Reproductive Medicine, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuang Qu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Dan Zhao
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingwan Zhou
- National Experimental Teaching Center of Basic Medical Science, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yunjun Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Guan
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chao Qin
- The State Key Lab of Reproductive Medicine, Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Jun Xiang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, China.
| | - Bing Yao
- National Experimental Teaching Center of Basic Medical Science, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China; Department of Medical Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
49
|
Ma H, Chen X, Mo S, Zhang Y, Mao X, Chen J, Liu Y, Tong WM, Lu Z, Yu S, Chen J. Targeting N-glycosylation of 4F2hc mediated by glycosyltransferase B3GNT3 sensitizes ferroptosis of pancreatic ductal adenocarcinoma. Cell Death Differ 2023; 30:1988-2004. [PMID: 37479744 PMCID: PMC10406883 DOI: 10.1038/s41418-023-01188-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 06/09/2023] [Accepted: 06/26/2023] [Indexed: 07/23/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a highly fatal malignancy partially due to the acquired alterations related to aberrant protein glycosylation that pathologically remodel molecular biological processes and protect PDAC cells from death. Ferroptosis driven by lethal lipid peroxidation provides a targetable vulnerability for PDAC. However, the crosstalk between glycosylation and ferroptosis remains unclear. Here, we identified 4F2hc, a subunit of the glutamate-cystine antiporter system Xc-, and its asparagine (N)-glycosylation is involved in PDAC ferroptosis by N- and O-linked glycoproteomics. Knockdown of SLC3A2 (gene name of 4F2hc) or blocking the N-glycosylation of 4F2hc potentiates ferroptosis sensitization of PDAC cells by impairing the activity of system Xc- manifested by a marked decrease in intracellular glutathione. Mechanistically, we found that the glycosyltransferase B3GNT3 catalyzes the glycosylation of 4F2hc, stabilizes the 4F2hc protein, and enhances the interaction between 4F2hc and xCT. Knockout of B3GNT3 or deletion of enzymatically active B3GNT3 sensitizes PDAC cells to ferroptosis. Reconstitution of 4F2hc-deficient cells with wildtype 4F2hc restores ferroptosis resistance while glycosylation-mutated 4F2hc does not. Additionally, upon combination with a ferroptosis inducer, treatment with the classical N-glycosylation inhibitor tunicamycin (TM) markedly triggers the overactivation of lipid peroxidation and enhances the sensitivity of PDAC cells to ferroptosis. Notably, we confirmed that genetic perturbation of SLC3A2 or combination treatment with TM significantly augments ferroptosis-induced inhibition of orthotopic PDAC. Clinically, high expression of 4F2hc and B3GNT3 contributes to the progression and poor survival of PDAC patients. Collectively, our findings reveal a previously unappreciated function of N-glycosylation of 4F2hc in ferroptosis and suggest that dual targeting the vulnerabilities of N-glycosylation and ferroptosis may be an innovative therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Heng Ma
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Xianlong Chen
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Shengwei Mo
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Yue Zhang
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Xinxin Mao
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Jingci Chen
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Yilin Liu
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Wei-Min Tong
- Department of Pathology, Institute of Basic Medical Sciences, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Zhaohui Lu
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China
| | - Shuangni Yu
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China.
| | - Jie Chen
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, 100730, China.
| |
Collapse
|
50
|
Fan X, Fan YT, Zeng H, Dong XQ, Lu M, Zhang ZY. Role of ferroptosis in esophageal cancer and corresponding immunotherapy. World J Gastrointest Oncol 2023; 15:1105-1118. [PMID: 37546564 PMCID: PMC10401468 DOI: 10.4251/wjgo.v15.i7.1105] [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: 12/28/2022] [Revised: 02/28/2023] [Accepted: 04/24/2023] [Indexed: 07/12/2023] Open
Abstract
Esophageal cancer (EC) is one of the most common digestive system malignancies in the world. The combined modality treatment of EC is usually surgery and radiation therapy, however, its clinical efficacy for advanced patients is relatively limited. Ferroptosis, a new type of iron-dependent programmed cell death, is different from apoptosis, necrosis and autophagy. In recent years, many studies have further enlightened that ferroptosis plays an essential role in the occurrence, development and metastasis of tumors. Targeting ferroptosis stimulates a new direction for further exploration of oncologic treatment regimens. Furthermore, ferroptosis has a critical role in the immune microenvironment of tumors. This paper reviews the mechanism of ferroptosis and the ferroptosis research progress in the treatment of EC. We further elaborate the interaction between ferroptosis and immunotherapy, and the related mechanisms of ferroptosis participation in the immunotherapy of EC, so as to provide new directions and ideas for the treatment of EC.
Collapse
Affiliation(s)
- Xin Fan
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China
| | - Yan-Ting Fan
- The First Clinical Medical College, Nanchang University, Nanchang 330000, Jiangxi Province, China
| | - Hui Zeng
- Department of Stomatology, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China
| | - Xi-Qi Dong
- The First Clinical Medical College, Nanchang University, Nanchang 330000, Jiangxi Province, China
| | - Min Lu
- Department of Emergency Medicine, Shangrao Hospital Affiliated to Nanchang University, Shangrao 334000, Jiangxi Province, China
| | - Zhi-Yuan Zhang
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, China
| |
Collapse
|