1
|
Lv X, Yang C, Li X, Liu Y, Yang Y, Jin T, Chen Z, Jia J, Wang M, Li L. Ferroptosis and hearing loss: from molecular mechanisms to therapeutic interventions. J Enzyme Inhib Med Chem 2025; 40:2468853. [PMID: 39992186 PMCID: PMC11852237 DOI: 10.1080/14756366.2025.2468853] [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: 08/27/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
Hearing loss profoundly affects social engagement, mental health, cognition, and brain development, with sensorineural hearing loss (SNHL) being a major concern. Linked to ototoxic medications, ageing, and noise exposure, SNHL presents significant treatment challenges, highlighting the need for effective prevention and regeneration strategies. Ferroptosis, a distinct form of cell death featuring iron-dependent lipid peroxidation, has garnered interest due to its potential role in cancer, ageing, and neuronal degeneration, especially hearing loss. The emerging role of ferroptosis as a crucial mediator in SNHL suggests that it may offer a novel therapeutic target for otoprotection. This review aims to summarise the intricate connection between ferroptosis and SNHL, offering a fresh perspective for exploring targeted therapeutic strategies that could potentially mitigate cochlear cells damage and enhance the quality of life for individuals with hearing impairments.
Collapse
Affiliation(s)
- Xingyi Lv
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Chenyi Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Xianying Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yun Liu
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Yu Yang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Tongyan Jin
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhijian Chen
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Jinjing Jia
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Min Wang
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Li Li
- Department of Physiology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
2
|
Dai JZ, Hsu WJ, Lin MH, Shueng PW, Lee CC, Yang CC, Lin CW. YAP-mediated DDX3X confers resistance to ferroptosis in breast cancer cells by reducing lipid peroxidation. Free Radic Biol Med 2025; 232:330-339. [PMID: 40089076 DOI: 10.1016/j.freeradbiomed.2025.03.019] [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: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Metabolic shifts in cancer cells were found to participate in tumorigenesis, especially driving chemotherapeutic resistance. Ferroptosis is a newly discovered form of cell death induced by excessive accumulations of iron and lipid peroxidation. Susceptibility to ferroptosis can be intrinsically regulated by various cellular metabolic pathways. Therefore, inducing ferroptosis might be a promising anticancer therapeutic strategy. DEAD-box helicase 3 X-linked (DDX3X), a critical modulator of RNA metabolism, was identified as an oncogene in breast cancer and also participates in cancer metabolism and chemotherapeutic resistance. However, the molecular regulation of the association between DDX3X and ferroptosis is largely unknown. Herein, we investigated the correlation between resistance to ferroptosis and DDX3X expression in breast cancer cells. We found that elevation of DDX3X was associated with increased resistance to a ferroptosis inducer in breast cancer cells, and manipulating DDX3X expression regulated the sensitivity to the ferroptosis inducer. Importantly, DDX3X upregulated expression of the anti-ferroptotic enzyme glutathione peroxidase 4 (GPX4) gene to confer ferroptosis resistance in breast cancer cells. Moreover, DDX3X was transcriptionally upregulated by the yes-associated protein (YAP). Knockdown of YAP downregulated DDX3X mRNA expression and facilitated lipid peroxidation, but that were restored in the presence of DDX3X. Clinically, coexpression of DDX3X and YAP was found in a variety of malignancy, and their elevation conferred poor survival prognosis in patients with breast cancer. Together, our findings reveal the crucial role of DDX3X in sensitivity to ferroptosis and underscore its potential as a diagnostic marker and therapeutic target. DDX3X renders resistance to ferroptosis and plays a role in mitigating lipid peroxidation, paving the way for therapeutic vulnerability via targeting cancer metabolism.
Collapse
Affiliation(s)
- Jia-Zih Dai
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Jing Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Hsiang Lin
- Graduate Institute of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Ching Lee
- Istanbul Sabahattin Zaim University, Faculty of Engineering and Natural Sciences, Department of Food Engineering, Istanbul, Turkey; Department of Food Technology and Nutrition, Faculty of Technologies, Klaipėda State University of Applied Sciences, Lithuania
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan; Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Cheng-Wei Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
3
|
Zhang W, Hong X, Xiao Y, Wang H, Zeng X. Sorafenib resistance and therapeutic strategies in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2025:189310. [PMID: 40187502 DOI: 10.1016/j.bbcan.2025.189310] [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: 11/08/2024] [Revised: 03/30/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent and lethal cancers globally. While surgical resection and liver transplantation offer potential cures for early-stage HCC, the majority of patients are diagnosed at advanced stages where such interventions are not viable. Sorafenib, a multi-target kinase inhibitor, has been a cornerstone in the treatment of advanced HCC since its approval in 2007. Despite its significant clinical impact, less than half of the treated patients derive long-term benefits due to the emergence of resistance and associated side effects. This review focuses on the role of sorafenib, an FDA-approved multi-target kinase inhibitor, in treating advanced HCC, discusses the mechanisms underlying its therapeutic effects and associated resistance, and explores additional therapeutic strategies being investigated to improve patient outcomes.
Collapse
Affiliation(s)
- Weijing Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Xuechuan Hong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuling Xiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaodong Zeng
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China.
| |
Collapse
|
4
|
Wang L, ChenLiu Z, Wang D, Tang D. Cross-talks of GSH, mitochondria, RNA m6A modification, NRF2, and p53 between ferroptosis and cuproptosis in HCC: A review. Int J Biol Macromol 2025; 302:140523. [PMID: 39894098 DOI: 10.1016/j.ijbiomac.2025.140523] [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: 11/11/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with high morbidity and mortality, as well as poor prognosis. Therefore, it is imperative to explore alternative therapeutic targets for HCC treatment. Ferroptosis and cuproptosis have recently been identified as metal-dependent cell death mechanisms that play significant roles in HCC treatment. This study identified potential cross-talk between ferroptosis and cuproptosis, including the common hub glutathione, common site of occurrence, mitochondria, shared epigenetic modification mode, RNA N6 methyladenosine modification, mutual inhibitor, nuclear factor erythroid 2-related factor 2, and dual regulator, p53. These findings provide a theoretical foundation for the joint induction of HCC cell death and effective inhibition of HCC progression. However, some immune cells are susceptible to ferroptosis or cuproptosis, which may impair or enhance anti-cancer immune function. We propose strategies to target specific targets molecules such as tripartite motif containing 25, ferroptosis suppressor protein 1, and peroxisome proliferator-activated receptor gamma or exploit the unique acidic environment surrounding cancer cells to precisely induce ferroptosis in cancer cells. This approach aims to advance the development of precision medicine for HCC treatment.
Collapse
Affiliation(s)
- Leihan Wang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Zhenni ChenLiu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou School of Clinical Medicine of Dalian Medical University, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou 225000, China.
| |
Collapse
|
5
|
Mete M, Ojha A, Dhar P, Das D. Deciphering Ferroptosis: From Molecular Pathways to Machine Learning-Guided Therapeutic Innovation. Mol Biotechnol 2025; 67:1290-1309. [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] [MESH Headings] [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
|
6
|
Yang J, Hu B, Zhang G, Wu K, Zhang X, Ji M, Zhang B, Shi H, Li D. Protocadherin 17 weakens the lenvatinib resistance of liver cancer through inducing ferroptosis. Exp Cell Res 2025; 447:114495. [PMID: 40049312 DOI: 10.1016/j.yexcr.2025.114495] [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: 11/14/2024] [Revised: 01/23/2025] [Accepted: 03/04/2025] [Indexed: 03/09/2025]
Abstract
Lenvatinib has been employed in the treatment of advanced liver cancer; however, its clinical application is significantly impeded by frequent drug resistance. Recent studies have revealed that lenvatinib treatment triggers ferroptosis in liver cancer cells, providing a novel approach to addressing lenvatinib resistance. In this study, we initially validated the induction of ferroptosis by lenvatinib in liver cancer cells. Remarkably, protocadherin 17 (PCDH17), an adhesion-related protein, was found to be down-regulated in liver cancer, and overexpression of PCDH17 could induce ferroptosis in liver cancer cells. Importantly, silencing PCDH17 inhibited the impact of lenvatinib on liver cancer cell ferroptosis, while overexpression of PCDH17 had the opposite effect. These findings were further confirmed using a xenograft tumor model in BALB/c nude mice. Additionally, lenvatinib-resistant (LR) liver cancer cells were generated for additional validation purposes. It was observed that LR-liver cancer cells lost their susceptibility to ferroptosis induction by lenvatinib; however, overexpression of PCDH17 reactivated their sensitivity to ferroptosis. Corresponding results were also verified in BALB/c nude mice models. In conclusion, these results suggest that as a novel regulator of ferroptosis, PCDH17 can alleviate lenvatinib resistance and potentially enhance the therapeutic efficacy of lenvatinib in treating liver cancer.
Collapse
Affiliation(s)
- Jun Yang
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bin Hu
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of General Surgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guowei Zhang
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kai Wu
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xue Zhang
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mengxuan Ji
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bin Zhang
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Hengliang Shi
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Dechun Li
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
7
|
Liu X, Wang W, Nie Q, Liu X, Sun L, Ma Q, Zhang J, Wei Y. The Role and Mechanisms of Ubiquitin-Proteasome System-Mediated Ferroptosis in Neurological Disorders. Neurosci Bull 2025; 41:691-706. [PMID: 39775589 PMCID: PMC11979074 DOI: 10.1007/s12264-024-01343-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/29/2024] [Indexed: 01/11/2025] Open
Abstract
Ferroptosis is a form of cell death elicited by an imbalance in intracellular iron concentrations, leading to enhanced lipid peroxidation. In neurological disorders, both oxidative stress and mitochondrial damage can contribute to ferroptosis, resulting in nerve cell dysfunction and death. The ubiquitin-proteasome system (UPS) refers to a cellular pathway in which specific proteins are tagged with ubiquitin for recognition and degradation by the proteasome. In neurological conditions, the UPS plays a significant role in regulating ferroptosis. In this review, we outline how the UPS regulates iron metabolism, ferroptosis, and their interplay in neurological diseases. In addition, we discuss the future application of small-molecule inhibitors and identify potential drug targets. Further investigation into the mechanisms of UPS-mediated ferroptosis will provide novel insights and strategies for therapeutic interventions and clinical applications in neurological diseases.
Collapse
Affiliation(s)
- Xin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Wei Wang
- Cancer Biology Institute, Baotou Medical College, Baotou, 014010, China
| | - Qiucheng Nie
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xinjing Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Lili Sun
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qiang Ma
- Cancer Biology Institute, Baotou Medical College, Baotou, 014010, China
| | - Jie Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Yiju Wei
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
8
|
Yang LL, Chen X, Huang KT, Tang ST, Ye GY, Wang JL. BEND3 promotes hepatocellular carcinoma progression and metastasis by activating the PI3K/AKT/mTOR pathway and inducing epithelial-mesenchymal transition. Clin Res Hepatol Gastroenterol 2025; 49:102582. [PMID: 40158797 DOI: 10.1016/j.clinre.2025.102582] [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: 01/29/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE This study aimed to investigate the expression of BEND3 in hepatocellular carcinoma (HCC), its correlation with clinical characteristics, and its functional and mechanistic impacts on HCC progression. METHODS Bioinformatics analyses identified BEND3 as highly expressed in HCC and associated with poor clinical prognosis, which was further validated using qRT-PCR, western blotting and immunohistochemistry. Stable BEND3-overexpressing and silenced cell lines were constructed to evaluate its functional effects. CCK-8 and colony formation assays assessed its influence on cell proliferation, while wound healing and Transwell assays evaluated its role in migration and invasion. WB and immunofluorescence were employed to analyze the effects of BEND3 on epithelial-mesenchymal transition (EMT) and the PI3K/AKT/mTOR signaling pathway. RESULTS Public database analysis, alongside qRT-PCR, western blotting, and immunohistochemical, confirmed that BEND3 expression is significantly elevated in HCC tissues compared to normal liver tissues and is closely associated with poor prognosis. Functional assays demonstrated that BEND3 promotes HCC cell proliferation, migration, and invasion. Mechanistic studies revealed that BEND3 drives HCC progression by inducing EMT and activating the PI3K/AKT/mTOR signaling pathway. CONCLUSION BEND3 is highly expressed in HCC and strongly correlates with poor clinical outcomes. Functional and mechanistic analyses indicate that BEND3 enhances HCC progression by promoting proliferation, migration and invasion via EMT induction and PI3K/AKT/mTOR pathway activation.
Collapse
Affiliation(s)
- Liu-Lin Yang
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China
| | - Xing Chen
- Department of Ultrasonography, Guangxi Medical University First Affiliated Hospital, China
| | - Kai-Ting Huang
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China
| | - Shao-Tong Tang
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China
| | - Gui-Yan Ye
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China
| | - Ji-Long Wang
- Department of Hepatological Surgery, Guangxi Medical University First Affiliated Hospital, China.
| |
Collapse
|
9
|
Yu Z, Luo J, An W, Wei H, Song A, Mao Y, Li M, He L, Xiao F, Gao Q, Wei H. EOGT knockdown promotes ferroptosis and inhibits hepatocellular carcinoma proliferation by regulating SLC7A11 via HEY1. Cell Signal 2025:111772. [PMID: 40154588 DOI: 10.1016/j.cellsig.2025.111772] [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: 12/10/2024] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality, characterized by a complex molecular landscape. EGF Domain Specific O-linked β-N-acetylglucosamine transferase (EOGT) functions as an O-GlcNAc transferase with specific activity towards proteins containing epidermal growth factor (EGF) repeats. Although it is known to potentially play an oncogenic role in HCC, the exact mechanisms remain unclear. Induction of ferroptosis is a primary mechanism by which anticancer drugs such as sorafenib treat HCC. This study aimed to elucidate the expression profile of EOGT in HCC and its relationship with ferroptosis, as well as to investigate the underlying molecular mechanisms. METHODS Utilizing bioinformatics resources, we explored the potential role of EOGT in HCC. The effects of EOGT on HCC cell behavior were examined using cell models and subcutaneous xenograft models in nude mice. Further insights into the molecular mechanisms were obtained through RNA-seq in cell models, hydrodynamic modeling in mice, Western blotting, chromatin immunoprecipitation (ChIP) sequencing, and dual-luciferase reporter assays to analyze the interaction between HEY1 and SLC7A11. Multiple validation steps were employed to thoroughly investigate the roles of these factors in the regulation of ferroptosis in HCC. RESULTS Our findings revealed that EOGT is upregulated in HCC and correlates with poor prognosis and drug resistance. Knockdown of EOGT inhibited HCC cell proliferation and enhanced sensitivity to ferroptosis by downregulating SLC7A11, a process mediated by HEY1. These results were confirmed by cell viability assays, quantitative real-time PCR (qPCR), Western blotting, and dual-luciferase reporter gene assays. CONCLUSIONS EOGT promotes HCC proliferation and inhibits ferroptosis by modulating the HEY1-SLC7A11 axis, suggesting a potential therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Zhe Yu
- Department of Gastroenterology, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Jing Luo
- Department of Gastroenterology, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Wen An
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Herui Wei
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Aqian Song
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yuanpeng Mao
- Department of Gastroenterology, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Mengqi Li
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Lingling He
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Fan Xiao
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Qi Gao
- Beijing Youngen Technology Co., Ltd., Beijing 102629, China.
| | - Hongshan Wei
- Department of Gastroenterology, Peking University Ditan Teaching Hospital, Beijing 100015, China; Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| |
Collapse
|
10
|
Xu M, Trung TS, Zhu Z, Li S, Gong S, Cheng N, Zhou P, Wang S. ESR1-dependent suppression of LCN2 transcription reverses autophagy-linked ferroptosis and enhances sorafenib sensitivity in hepatocellular carcinoma. J Physiol Biochem 2025:10.1007/s13105-025-01073-y. [PMID: 40126852 DOI: 10.1007/s13105-025-01073-y] [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: 10/22/2024] [Accepted: 03/05/2025] [Indexed: 03/26/2025]
Abstract
Sorafenib resistance is a significant hurdle in the treatment landscape of hepatocellular carcinoma (HCC). Lipocalin 2 (LCN2), a secretory glycoprotein that transports lipophilic molecules across cell membranes, is thought to affect the s therapeutic efficacy of sorafenib. Despite its importance, the detailed regulatory pathways involving LCN2 are still being deciphered. We probed the correlation between LCN2 expression and sorafenib resistance in HCC cells. Through the modulation of LCN2 levels, we investigated its role in cell proliferation, apoptosis, and its regulatory effects on autophagy-driven ferroptosis. With the aid of hTFtarget and JASPAR databases, ESR1 was pinpointed as a transcriptional inhibitor of LCN2. The impact of the ESR1-LCN2 axis on sorafenib resistance in HCC was then examined in vitro and validated in a xenograft tumor mouse model. In HCC cells, elevated LCN2 levels were found to be associated with resistance to sorafenib. Depletion of LCN2 resulted in attenuated HCC cell growth and elevated rates of apoptosis and ferroptosis. Overexpression of LCN2 had the opposite effect, promoting cell proliferation and suppressing cell death pathways, a response that could be overridden by autophagy agonists. ESR1 suppressed LCN2 transcription, which in turn activated autophagy-mediated ferroptosis, mitigating sorafenib tolerance in HCC and enhancing the therapeutic index. ESR1 targets LCN2 transcription to initiate autophagy-driven ferroptosis, thereby reducing sorafenib resistance in HCC cells.
Collapse
Affiliation(s)
- Mingfang Xu
- Department of Otolaryngology Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434020, China
| | - Tran Sy Trung
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Zhiyong Zhu
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Shijia Li
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Shicheng Gong
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Nuo Cheng
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Peng Zhou
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China
| | - Shuai Wang
- Department of Hepatobiliary Surgery, Jingzhou Hospital Affiliated to Yangtze University, No. 26 Chuyuan Avenue, Jingzhou District, Jingzhou, 434020, China.
| |
Collapse
|
11
|
Liu K, Wang Z, Guo X, Luo J, Wu X, Wang F, Mei Y. The glutamine starvation-induced lncRNA FERRIN suppresses ferroptosis via the stabilization of SLC7A11 mRNA. Int J Biol Macromol 2025; 308:142388. [PMID: 40127798 DOI: 10.1016/j.ijbiomac.2025.142388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/19/2024] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
As an essential nutrient for cancer cell survival, glutamine plays both promoting and inhibitory roles in ferroptosis; however, the underlying mechanisms remain obscure. Emerging evidence suggests that long noncoding RNAs (lncRNAs) are crucial regulators of ferroptosis. Nevertheless, it remains unclear whether lncRNAs are involved in glutamine-regulated ferroptosis. In this study, we report that the lncRNA FERRIN is induced by the transcription factor ATF4 under glutamine starvation conditions. FERRIN functions as an inhibitor of ferroptosis by upregulating SLC7A11 expression. Mechanistically, FERRIN interacts with the RNA binding protein hnRNPK, facilitating its binding to SLC7A11 mRNA and leading to the stabilization of SLC7A11 mRNA. Consistent with its inhibitory role in ferroptosis, FERRIN promotes in vitro cancer cell proliferation and in vivo xenograft tumor growth through its modulation of SLC7A11. Collectively, these findings establish FERRIN as a critical negative regulator of ferroptosis and suggest that FERRIN may represent an important link between glutamine availability and ferroptosis.
Collapse
Affiliation(s)
- Kaiyue Liu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhongyu Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaorui Guo
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jingjing Luo
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xianning Wu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Fang Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| | - Yide Mei
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
12
|
Shang T, Jia Z, Li J, Cao H, Xu H, Cong L, Ma D, Wang X, Liu J. Unraveling the triad of hypoxia, cancer cell stemness, and drug resistance. J Hematol Oncol 2025; 18:32. [PMID: 40102937 PMCID: PMC11921735 DOI: 10.1186/s13045-025-01684-4] [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: 12/15/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
In the domain of addressing cancer resistance, challenges such as limited effectiveness and treatment resistance remain persistent. Hypoxia is a key feature of solid tumors and is strongly associated with poor prognosis in cancer patients. Another significant portion of the development of acquired drug resistance is attributed to tumor stemness. Cancer stem cells (CSCs), a small tumor cell subset with self-renewal and proliferative abilities, are crucial for tumor initiation, metastasis, and intra-tumoral heterogeneity. Studies have shown a significant association between hypoxia and CSCs in the context of tumor resistance. Recent studies reveal a strong link between hypoxia and tumor stemness, which together promote tumor survival and progression during treatment. This review elucidates the interplay between hypoxia and CSCs, as well as their correlation with resistance to therapeutic drugs. Targeting pivotal genes associated with hypoxia and stemness holds promise for the development of novel therapeutics to combat tumor resistance.
Collapse
Affiliation(s)
- Tongxuan Shang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Ziqi Jia
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiayi Li
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, 100730, China
| | - Heng Cao
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hengyi Xu
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Cong
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| | - Dongxu Ma
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiang Wang
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jiaqi Liu
- Department of Breast Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
13
|
Gao X, Cheng H, Teng M, Zhang H, Chen H, Qu S, Liu G. Optimizing interventional therapy: A homogeneous lipiodol formulation of Tirapazamine and Sorafenib responsive to post-embolization microenvironment. J Control Release 2025; 379:879-889. [PMID: 39880038 DOI: 10.1016/j.jconrel.2025.01.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/10/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Transcatheter arterial chemoembolization (TACE) is the principal treatment option for patients with unresectable hepatocellular carcinoma (HCC). However, the hypoxic microenvironment following TACE can promote angiogenesis and suppress tumor ferroptosis, resulting in an unfavorable prognosis. Tirapazamine (TPZ), a hypoxia-activated prodrug with specific cytotoxicity for hypoxic cells, making it a potential candidate for TACE. To develop an effective hypoxia-responsive drug delivery platform for TACE, we propose a novel lipiodol embolic formulation that integrates TPZ and sorafenib (SFB) by super-stable homogeneous intermixed formulation technology (SHIFT). This approach achieves the manufacture of embolic agents with stable drug dispersion characteristics, fulfilling the need for sustained drug release in TACE. The prolonged tumor penetration of TPZ exhibited embolization-responsive tumor killing, and its combination with SFB can suppress hypoxia-induced angiogenesis and trigger tumor ferroptosis, maintaining low oxygen levels, thereby boosting the therapeutic efficacy of TPZ. Conversely, TPZ can combat the resistance to SFB in hypoxic tumor cells. In summary, this study developed a novel embolization drug formulation based on embolic hypoxic microenvironment. The synergistic mechanism of TPZ and SFB enhances the therapeutic effects of hypoxia-activated prodrugs and mitigates the adverse effects of hypoxia.
Collapse
Affiliation(s)
- Xing Gao
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hongwei Cheng
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China; Zhuhai UM Science & Technology Research Institute, University of Macau, Macau 999078, China.
| | - Minglei Teng
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hongrui Zhang
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hu Chen
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Songnan Qu
- Zhuhai UM Science & Technology Research Institute, University of Macau, Macau 999078, China; Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa, Macau 999078, China
| | - Gang Liu
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
14
|
Tang X, Mo D, Jiang N, Kou Y, Zhang Z, Peng R, Mao X, Wang R, Wang Y, Yan F. Polysaccharides from maggot extracts suppressed colorectal cancer progression by inducing ferroptosis via HMOX1/GPX4 signaling pathway. Int J Biol Macromol 2025; 296:139734. [PMID: 39798758 DOI: 10.1016/j.ijbiomac.2025.139734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/17/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Maggots contain various kinds of polysaccharides and recent studies mostly concentrated on their anti-inflammatory functions. While the molecule mechanisms related to the polysaccharides inhibiting carcinogenesis remains unclear. Here we characterized the polysaccharides extracted from maggot (MEs) determining their anti-colon cancer potentials. ME in this study were composed of glucose, mannose, galactose, arabinose and xylose. ME dose-and time-dependently inhibited viability and obviously induced G0/G1 phase arrest in human colon cancer cells. Additionally, Proteomics and western blotting proved that ME suppressed the expression of GPX4 and increased the expression of HMOX1 in vivo and vitro. ME promoted ferroptosis in HCT116 and LOVO cells, reversing ROS, lipid peroxidation and GSSG/GSH radio level. In general, the findings stated that the polysaccharides provided effects of inducing colon cancer ferroptosis, uncovering potential function of ME from maggot as a candidate compound.
Collapse
Affiliation(s)
- Xun Tang
- Department of Clinical Laboratory, the Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing 210009, China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing 210009, China; State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - Dongping Mo
- Department of Clinical Laboratory, the Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing 210009, China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing 210009, China
| | - Ning Jiang
- Department of Clinical Laboratory, the Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Yingying Kou
- Good Clinical Practice Office, the Affiliated Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Zhe Zhang
- Department of Pathology, the Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Rui Peng
- Department of General Surgery, the Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xuelian Mao
- Department of Clinical Laboratory, the Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing 210009, China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing 210009, China
| | - Rong Wang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210000, Jiangsu, PR China.
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China.
| | - Feng Yan
- Department of Clinical Laboratory, the Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing 210009, China; Jiangsu Key Laboratory of Molecular and Translational Cancer Research, 42 Baiziting Road, Xuanwu District, Nanjing 210009, China.
| |
Collapse
|
15
|
Zhang Q, Zhang Y, Guo S, Wang H. Emerging insights into the role of microRNAs regulation of ferroptosis in hepatocellular carcinoma. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167642. [PMID: 39734007 DOI: 10.1016/j.bbadis.2024.167642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Hepatocellular carcinoma (HCC) is a major type of liver cancer and an important cause of cancer death. It has been reported that the hepatocyte death plays an important role in HCC. Ferroptosis is an iron-dependent programmed cell death characterized by the accumulation of free iron and lipid peroxidation. A series of studies have shown that ferroptosis contributes to the occurrence and development of HCC. MicroRNAs (miRNAs) are non-coding RNAs with a length of approximately 222 nt. In recent years, miRNAs have been shown to participate in regulating ferroptosis to play a vital role in HCC, but the related mechanisms are not fully understood. This review summarized the current understanding of ferroptosis, as well as the biogenesis and function of miRNAs, and focused on the role of miRNAs regulation of ferroptosis in HCC, with the hope of providing new targets and ideas for the treatment of HCC.
Collapse
Affiliation(s)
- Qi Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yingdan Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
16
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
17
|
Zeng H, Yu J, Wang H, Shen M, Zou X, Zhang Z, Liu L. Cancer ATF4-mediated CD58 endocytosis impairs anti-tumor immunity and immunotherapy. J Transl Med 2025; 23:225. [PMID: 40001116 PMCID: PMC11863482 DOI: 10.1186/s12967-025-06245-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Co-stimulatory molecules are imperative for CD8+ T cells to eliminate target cell and maintain sustained cytotoxicity. Despite an advanced understanding of the co-stimulatory molecules deficiency that results in tumor escape, the tumor cell-intrinsic mechanisms that regulate co-stimulatory molecules remain enigmatic, and an in-depth dissection could facilitate the improvement of treatment options. To this end, in this study, we report that the deficiency of the critical costimulatory molecule CD58, mediated by the expression of ATF4 in tumor cells, impairs the formation of immunological synapses (IS) and leads to the deterioration of antitumor immune function of CD8+ T cells. Mechanistically, ATF4 transcriptionally upregulated dynamin 1 (DNM1) expression leading to DNM1-dependent endocytosis (DDE)-mediated degradation of CD58. Furthermore, administration of DDE inhibitor prochlorperazine or ATF4 knockdown effectively restored CD58 expression, boosting CD8+ T cell cytotoxicity and immunotherapy efficiency. Thus, our study reveals that ATF4 in tumor cells weakens CD58 expression to interfere with complete IS formation, and indicates potential approaches to improve the cytolytic function of CD8+ T cell in tumor immunotherapy.
Collapse
Affiliation(s)
- Hanyi Zeng
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
- State Key Laboratory of Organ Failure Research, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangzhou, China
- Guangdong Institute of Hepatology, Guangzhou, China
| | - Jiaping Yu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
- State Key Laboratory of Organ Failure Research, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangzhou, China
- Guangdong Institute of Hepatology, Guangzhou, China
| | - Haijian Wang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
- State Key Laboratory of Organ Failure Research, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangzhou, China
- Guangdong Institute of Hepatology, Guangzhou, China
| | - Mengying Shen
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
- State Key Laboratory of Organ Failure Research, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangzhou, China
- Guangdong Institute of Hepatology, Guangzhou, China
| | - Xuejing Zou
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
- State Key Laboratory of Organ Failure Research, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangzhou, China
- Guangdong Institute of Hepatology, Guangzhou, China
| | - Ziyong Zhang
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China
- State Key Laboratory of Organ Failure Research, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangzhou, China
- Guangdong Institute of Hepatology, Guangzhou, China
| | - Li Liu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, China.
- State Key Laboratory of Organ Failure Research, Guangzhou, China.
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Viral Hepatitis, Guangzhou, China.
- Guangdong Institute of Hepatology, Guangzhou, China.
| |
Collapse
|
18
|
Shao J, Wang W, Li S, Yin G, Han L, Wang X, Cai M, Yang T, Wang Y, Qu W, Jiao Y, Wang P, Xu H, Zhu X, Ying S, Xu S, Sheng Q, Fang J, Jiang T, Wei C, Shen Y, Shen Y. Nuclear Overexpression of SAMHD1 Induces M Phase Stalling in Hepatoma Cells and Suppresses HCC Progression by Interacting with the Cohesin Complex. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411988. [PMID: 39679869 PMCID: PMC11809348 DOI: 10.1002/advs.202411988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/26/2024] [Indexed: 12/17/2024]
Abstract
Emerging evidence suggests that the sterile alpha-motif (SAM) and histidine-aspartate (HD) domain-containing protein 1 (SAMHD1) is implicated in various cancers, including hepatocellular carcinoma (HCC). However, its precise role in tumor cells and the underlying mechanisms remain unclear. This study aimed to investigate the expression patterns, prognostic values, and functional role of SAMHD1 in HCC progression. We constructed liver tissue microarrays using tumor and paired paratumor tissue specimens from 187 patients with primary HCC. Our findings indicate that nuclear SAMHD1 protein levels are increased in tumors compared to paratumor tissues. Moreover, nuclear SAMHD1 levels decline in advanced tumor stages, with higher SAMHD1 nuclear staining correlating with favorable prognostic outcomes. Hepatocyte-specific SAMHD1 knockout mice, generated by crossing SAMHD1fl/fl mice with Alb-cre mice, showed accelerated tumor progression in a diethylnitrosamine (DEN)-induced HCC model. In hepatoma cell lines, nuclear overexpression of SAMHD1 inhibited cell proliferation by stalling mitosis, independent of its deoxynucleotide triphosphohydrolase (dNTPase) function. Mechanistically, SAMHD1 interacts with the cohesin complex in nucleus, enhancing sister chromatid cohesion during cell division, which delays metaphase progression. Our findings suggest that nuclear SAMHD1 plays a critical role in slowing HCC progression by regulating mitosis, highlighting its potential as a therapeutic target by manipulating cohesin dynamics.
Collapse
Affiliation(s)
- Juntang Shao
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Wei Wang
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical University218 Jixi RoadHefei230022China
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Anhui Medical University218 Jixi RoadHefei230022China
| | - Shiyu Li
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Guangfa Yin
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Lili Han
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Xinyu Wang
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Meng Cai
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Tao Yang
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Ying Wang
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Wenyan Qu
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Yanhong Jiao
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Peng Wang
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Hanyang Xu
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Xu Zhu
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Songcheng Ying
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Sa Xu
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Qiang Sheng
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Jian Fang
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Tongcui Jiang
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Chuansheng Wei
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Yujun Shen
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
| | - Yuxian Shen
- School of Basic Medical Sciences and Biopharmaceutical Research InstituteAnhui Medical University81 Meishan RoadHefei230032China
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical University218 Jixi RoadHefei230022China
| |
Collapse
|
19
|
Lai L, Tan M, Hu M, Yue X, Tao L, Zhai Y, Li Y. Important molecular mechanisms in ferroptosis. Mol Cell Biochem 2025; 480:639-658. [PMID: 38668809 DOI: 10.1007/s11010-024-05009-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/07/2024] [Indexed: 02/19/2025]
Abstract
Ferroptosis is a type of cell death that is caused by the oxidation of lipids and is dependent on the presence of iron. It was first characterized by Brent R. Stockwell in 2012, and since then, research in the field of ferroptosis has rapidly expanded. The process of ferroptosis-induced cell death is genetically, biochemically, and morphologically distinct from other forms of cellular death, such as apoptosis, necroptosis, and non-programmed cell death. Extensive research has been devoted to comprehending the intricate process of ferroptosis and the various factors that contribute to it. While the majority of these studies have focused on examining the effects of lipid metabolism and mitochondria on ferroptosis, recent findings have highlighted the significant involvement of signaling pathways and associated proteins, including Nrf2, P53, and YAP/TAZ, in this process. This review provides a concise summary of the crucial signaling pathways associated with ferroptosis based on relevant studies. It also elaborates on the drugs that have been employed in recent years to treat ferroptosis-related diseases by targeting the relevant signaling pathways. The established and potential therapeutic targets for ferroptosis-related diseases, such as cancer and ischemic heart disease, are systematically addressed.
Collapse
Affiliation(s)
- Lunmeng Lai
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Menglei Tan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Mingming Hu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Xiyue Yue
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Lulu Tao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Yanru Zhai
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Yunsen Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China.
| |
Collapse
|
20
|
Wang D, Wang J, Yang L, Wang X, Huang S. Dexmedetomidine plays a protective role in sepsis-associated myocardial injury by repressing PRMT5-mediated ferroptosis. Toxicol Res (Camb) 2025; 14:tfaf010. [PMID: 39902345 PMCID: PMC11787764 DOI: 10.1093/toxres/tfaf010] [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: 07/24/2024] [Revised: 11/26/2024] [Accepted: 01/16/2025] [Indexed: 02/05/2025] Open
Abstract
Sepsis rapidly contributed to multiorgan failure, most typically damaging the cardiovascular system, and there were no effective treatments. Dexmedetomidine (Dex) has good therapeutic effects on sepsis-induced organ injury. Our work aimed to probe the pharmacological effects of Dex on ferroptosis in sepsis-associated myocardial injury (S-MI) and define underlying mechanism of action. Cardiomyocytes were exposed to lipopolysaccharide (LPS) for mimicking S-MI model in vitro. The septic mice were constructed by cecum ligation and puncture operation. The mRNA and protein expressions were assessed using quantitative real-time polymerase chain reaction or western blot. Cell survival was determined by cell counting kit-8, lactic dehydrogenase release, and flow cytometry assays. 2',7'-Dichlorodihydrofluorescein diacetate staining measured cellular reactive oxygen species level. The secretion levels of inflammatory cytokines, ferroptosis-related indicators were analyzed by enzyme-linked immunosorbent assay. The N6-methyladenosine (m6A) modification level of protein arginine methyltransferase 5 (PRMT5) mRNA was examined by methylated RNA binding protein immunoprecipitation (Me-RIP) assay. The interaction between methyltransferase like 3 (METTL3)/fat mass and obesity-associated protein (FTO) and PRMT5 was analyzed by RNA immunoprecipitation assay. Dex treatment alleviated LPS-induced cardiomyocyte injury and ferroptosis, while these effects of Dex were reversed by Erastin treatment. Mechanically, Dex ameliorated PRMT5 expression in LPS-induced cardiomyocytes by regulating METTL3/FTO catalyzed m6A modification on PRMT5 mRNA. Rescue experiments confirmed that PRMT5 overexpression abolished Dex-mediated inhibitory roles on LPS-induced cardiomyocyte injury and ferroptosis. Moreover, Dex administration alleviated inflammation, ferroptosis, and myocardial injury in septic mice. Taken together, Dex repressed PMRT5 expression in a m6A-dependent manner, thus lightening LPS-triggered ferroptosis to alleviate cardiomyocyte injury.
Collapse
Affiliation(s)
- Dan Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, Hunan Province 410011, P.R. China
| | - Jun Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, Hunan Province 410011, P.R. China
| | - Li Yang
- Department of Neurology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, Hunan Province 410011, P.R. China
| | - Xin Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, Hunan Province 410011, P.R. China
| | - Sijian Huang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, Hunan Province 410011, P.R. China
| |
Collapse
|
21
|
Liu F, Yin P, Lu L, Yao J, Jiao B. Identification of PIF1 as a Ferroptosis-Related Prognostic Biomarker Correlated with Immune Infiltration in Hepatocellular Carcinoma. Appl Biochem Biotechnol 2025:10.1007/s12010-024-05161-5. [PMID: 39888492 DOI: 10.1007/s12010-024-05161-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2024] [Indexed: 02/01/2025]
Abstract
Hepatocellular carcinoma (HCC) is a primary liver malignancy characterized by high morbidity and mortality. Recently, ferroptosis has been recognized as an important factor in regulating cell growth in HCC. However, the role of ferroptosis-related genes in HCC remains unclear. The SRP119173 dataset from the Sequence Read Archive database was used to screen differentially expressed genes (DEGs) related to ferroptosis. Meanwhile, weighted gene co-expression network analysis was conducted to identify the HCC-related gene modules in the TCGA-liver hepatocellular carcinoma (LIHC) cohort. Next, the candidate genes related to HCC progression and ferroptosis were identified by Venn analysis. Kaplan-Meier, multivariate COX regression, and CIBERSORT analyses were then performed. Our results found that the levels of PIF1 5'-to-3' DNA helicase (PIF1) were notably elevated in HCC tissues relative to normal tissues. Additionally, HCC patients with high PIF1 expression had worse overall survival outcomes than patients with low PIF1 expression. Additionally, the PIF1 gene could independently predict HCC patients' prognosis. Meanwhile, HCC patients with high PIF1 levels had a higher proportion of regulatory T cells (Tregs) and M0 macrophages, as well as higher expression of immune checkpoints such as PD-1 (PDCD1) and PD-L1 (CD274), compared with patients with low PIF1 levels. Our data suggested that a ferroptosis-related gene PIF1 may serve as a potential biomarker for predicting prognosis in HCC patients.
Collapse
Affiliation(s)
- Feng Liu
- Department of Head and Neck Surgery, Shanxi Provincial Cancer Hospital/Shanxi Hospital Cancer Hospital of Chinese Academy of Medical Sciences, Taiyuan, 030001, China
| | - Pengyu Yin
- Department of Gastroenterology, General Hospital of Tisco, The Sixth Hospital of Shanxi Medical University, Taiyuan, 030008, China
| | - Lifang Lu
- Department of General Surgery, Shanxi Provincial Cancer Hospital/Shanxi Hospital Cancer Hospital of Chinese Academy of Medical Sciences, No. 3, Gongye New Street , Xinhualing District, Taiyuan, 030001, China
| | - Jingchun Yao
- Department of Head and Neck Surgery, Shanxi Provincial Cancer Hospital/Shanxi Hospital Cancer Hospital of Chinese Academy of Medical Sciences, Taiyuan, 030001, China
| | - Baoping Jiao
- Department of General Surgery, Shanxi Provincial Cancer Hospital/Shanxi Hospital Cancer Hospital of Chinese Academy of Medical Sciences, No. 3, Gongye New Street , Xinhualing District, Taiyuan, 030001, China.
| |
Collapse
|
22
|
Li C, Xiong L, Yang Y, Jiang P, Wang J, Li M, Wei S, Tian S, Wang Y, Zhang M, Tang J. Sorafenib enhanced the function of myeloid-derived suppressor cells in hepatocellular carcinoma by facilitating PPARα-mediated fatty acid oxidation. Mol Cancer 2025; 24:34. [PMID: 39876004 PMCID: PMC11773820 DOI: 10.1186/s12943-025-02238-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Sorafenib, an FDA-approved drug for advanced hepatocellular carcinoma (HCC), faces resistance issues, partly due to myeloid-derived suppressor cells (MDSCs) that enhance immunosuppression in the tumor microenvironment (TME). METHODS Various murine HCC cell lines and MDSCs were used in a series of in vitro and in vivo experiments. These included subcutaneous tumor models, cell viability assays, flow cytometry, immunohistochemistry, and RNA sequencing. MDSCs were analyzed for chemotaxis, immunosuppressive functions, fatty acid oxidation (FAO), and PPARα expression. The impact of sorafenib on tumor growth, MDSC infiltration, differentiation, and immunosuppressive function was assessed, alongside the modulation of these processes by PPARα. RESULTS Here, we revealed increased infiltration and enhanced function of MDSCs in TME after treatment with sorafenib. Moreover, our results indicated that sorafenib induced the accumulation of MDSCs mediated by CCR2, and pharmacological blockade of CCR2 markedly reduced MDSCs migration and tumor growth. Mechanistically, sorafenib promoted the effect and fatty acid uptake ability of MDSCs and modulated peroxisome proliferator-activated receptor α (PPARα)-mediated fatty acid oxidation (FAO). In addition, tumor-bearing mice fed a high-fat diet (HFD) at the beginning of sorafenib administration had worse outcomes than mice fed a regular diet. Genetic deficiency of PPARα weakens the effect of sorafenib on MDSCs in mice with HCC. Pharmacological inhibition of PPARα has a synergistic anti-tumor effect with sorafenib, which is attenuated by the inhibition of MDSCs. Mechanistically, sorafenib significantly inhibited the differentiation of macrophages by upregulating PPARα expression and suppressing the PU.1-CSF1R pathway. CONCLUSION Overall, our study demonstrated that sorafenib enhanced the function of MDSCs by facilitating PPARα-mediated FAO and further augmenting sorafenib resistance, which sheds light on dietary management and improves the therapeutic response in HCC.
Collapse
Affiliation(s)
- Chunxiao Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| | - Liting Xiong
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Yuhan Yang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Ping Jiang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| | - Mengyuan Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Shuhua Wei
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Suqing Tian
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Yuexuan Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Mi Zhang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Jie Tang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| |
Collapse
|
23
|
Yang M, Cui W, Lv X, Xiong G, Sun C, Xuan H, Ma W, Cui X, Cheng Y, Han L, Chu B. S100P is a ferroptosis suppressor to facilitate hepatocellular carcinoma development by rewiring lipid metabolism. Nat Commun 2025; 16:509. [PMID: 39779666 PMCID: PMC11711731 DOI: 10.1038/s41467-024-55785-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Ferroptosis is a newly identified programmed cell death induced by iron-driven lipid peroxidation and implicated as a potential approach for tumor treatment. However, emerging evidence indicates that hepatocellular carcinoma (HCC) cells are generally resistant to ferroptosis and the underlying molecular mechanism is poorly understood. Here, our study confirms that S100 calcium binding protein P (S100P), which is significantly up-regulated in ferroptosis-resistant HCC cells, efficiently inhibits ferroptosis. Mechanistically, S100P facilitates lysosomal degradation of acetyl-CoA carboxylase alpha (ACC1), which is indispensable for de novo biosynthesis of lipids. Loss of S100P elevates the expression of ACC1 and promotes ferroptotic sensitivity of HCC cells. S100P-mediated ACC1 degradation relies on RAB5C, which directs ACC1 to lysosome via P62-dependent selective autophagy. Knockdown of RAB5C or P62 abrogates S100P-induced lysosomal degradation of ACC1 and restores resistance of HCC cells to ferroptosis. Our work reveals an alternative anti-ferroptosis pathway and suggests S100P as a promising druggable target for ferroptosis-related therapy of HCC.
Collapse
Grants
- National Key R&D Program of China(2022YFA0912600, B.C.), National Natural Science Foundation of China (32000515 and 32370800, B.C.; 82472725, 81972275 and 82171748, L.H.), Natural Science Foundation of Shandong Province (ZR2020QC074, B.C.), Joint Fund of Shandong Provincial Natural Science Foundation (ZR2023LZL010, L.H.), Distinguished Professor of Taishan Scholars (tstp20221109, L.H.)
Collapse
Affiliation(s)
- Min Yang
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Weiwei Cui
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiaoting Lv
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Gaozhong Xiong
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Caiyu Sun
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Haocheng Xuan
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Wei Ma
- Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xiuling Cui
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yeping Cheng
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Lihui Han
- Department of Immunology, Shandong Provincial Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
24
|
Xi C, Zhou J, Zheng X, Fu X, Xie M. Sodium aescinate-induced hepatotoxicity via ATF4/GSH/GPX4 axis-mediated ferroptosis. Sci Rep 2025; 15:1141. [PMID: 39774712 PMCID: PMC11706965 DOI: 10.1038/s41598-024-79723-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
Sodium aescinate (SA), a natural plant extract with various bioactivities, is widely used to treat oedema and inflammation in clinics. However, adverse events, including liver injury, kidney injury, and phlebitis, have been reported in patients with SA in recent years. In this study, we used BALB/c mice and L02 cells to evaluate the role of ferroptosis in SA-induced liver injury. SA significantly increased AST, ALT, MDA and Fe2+, decreased GSH levels, and induced pathological changes in the liver in vivo. SA also reduced the viability of L02 cells and induced LDH release, intracellular cysteine reduction, GSH depletion, iron accumulation, ROS production, and lipid peroxidation, indicating that SA causes ferroptosis. In addition, SA inhibited transcriptional activity of activating transcription factor 4 (ATF4) and subsequently reduced the expression of the downstream genes xCT (solute carrier family 7a member 11, SLC7A11) and Cystathionine gamma-lyase (CTH) which play vital roles in GSH biosynthesis. Interestingly, the cytotoxic effects of SA were effectively attenuated by ATF4 overexpression, while they were significantly aggravated by ATF4 silencing. These results revealed that SA triggers hepatocyte ferroptosis by inhibiting the activity of ATF4, which causes an oxidative imbalance.
Collapse
Affiliation(s)
- Chen Xi
- Pharmaceutical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China.
| | - Xin Zheng
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| | - Xiaoyi Fu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| | - Minjuan Xie
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, Jiangxi, P.R. China
| |
Collapse
|
25
|
Wang J, Liao L, Miao B, Yang B, Li B, Ma X, Fitz A, Wu S, He J, Zhang Q, Ji S, Jin G, Zhang J, Cao Y, Wang H, Qin W, Sun C, Bernards R, Wang C. Deciphering the role of the MALT1-RC3H1 axis in regulating GPX4 protein stability. Proc Natl Acad Sci U S A 2025; 122:e2419625121. [PMID: 39739814 PMCID: PMC11725786 DOI: 10.1073/pnas.2419625121] [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: 09/24/2024] [Accepted: 11/25/2024] [Indexed: 01/02/2025] Open
Abstract
Ferroptosis, a unique form of iron-dependent cell death triggered by lipid peroxidation accumulation, holds great promise for cancer therapy. Despite the crucial role of GPX4 in regulating ferroptosis, our understanding of GPX4 protein regulation remains limited. Through FACS-based genome-wide CRISPR screening, we identified MALT1 as a regulator of GPX4 protein. Inhibition of MALT1 expression enhances GPX4 ubiquitination-mediated degradation by up-regulating the E3 ubiquitin ligase RC3H1. Using both rescue assays and functional genetic screening, we demonstrate that pharmacologically targeting MALT1 triggers ferroptosis in liver cancer cells. Moreover, we show that targeting MALT1 synergizes with sorafenib or regorafenib to induce ferroptosis across multiple cancer types. These findings elucidate the modulatory effects of the MALT1-RC3H1 axis on GPX4 stability, revealing a molecular mechanism that could be exploited to induce ferroptosis for cancer therapy.
Collapse
Affiliation(s)
- Jun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Long Liao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Beiping Miao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
- German Cancer Research Center, Division Immune Regulation in Cancer, Heidelberg69120, Germany
| | - Bo Yang
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei230027, China
| | - Botai Li
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Xuhui Ma
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam1066 CX, The Netherlands
| | - Annika Fitz
- German Cancer Research Center, Division Immune Regulation in Cancer, Heidelberg69120, Germany
| | - Shanshan Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Jia He
- Department of Medical Oncology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai200072, China
| | - Qianqian Zhang
- National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200025, China
| | - Shuyi Ji
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai200032, China
| | - Guangzhi Jin
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200336, China
| | - Jianming Zhang
- Institute of Translational Medicine, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Ying Cao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Chong Sun
- German Cancer Research Center, Division Immune Regulation in Cancer, Heidelberg69120, Germany
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam1066 CX, The Netherlands
| | - Cun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| |
Collapse
|
26
|
Chen Y, Gao Q, Wang D, Zou X, Li X, Ji J, Liu B. An Overview of Research Advances in Oncology Regarding the Transcription Factor ATF4. Curr Drug Targets 2025; 26:59-72. [PMID: 39350552 DOI: 10.2174/0113894501328461240921062056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/09/2024] [Accepted: 09/10/2024] [Indexed: 02/19/2025]
Abstract
This review provides a comprehensive overview of the recent advancements in research on ATF4 (Activating Transcription Factor 4) within the field of oncology. As a crucial transcription factor, ATF4 has garnered increasing attention for its role in cancer research. The review begins with an exploration of the regulatory mechanisms of ATF4, including its transcriptional control, post-translational modifications, and interactions with other transcription factors. It then highlights key research findings on ATF4's involvement in various aspects of tumor biology, such as cell proliferation, differentiation, apoptosis and survival, invasion and metastasis, and the tumor microenvironment. Furthermore, the review discusses the potential of targeting ATF4 as a novel therapeutic strategy for cancer treatment. It also explores how ATF4's interactions with existing anticancer drugs could inform the development of more effective therapeutic agents. By elucidating the role of ATF4 in tumor biology and its potential clinical applications, this review aims to provide new insights and strategies for cancer treatment.
Collapse
Affiliation(s)
- Yulu Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Qi Gao
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Dan Wang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xun Zou
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiuming Li
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Bin Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| |
Collapse
|
27
|
Li X, Zhu D. Role of disulfide death in cancer (Review). Oncol Lett 2025; 29:55. [PMID: 39606569 PMCID: PMC11600708 DOI: 10.3892/ol.2024.14801] [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: 07/24/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
The research field of regulated cell death is growing extensively. Following the recognition of ferroptosis, other unique and distinct forms of regulated cell death, including cuproptosis and disulfide death, have been identified. Disulfide death occurs due to the abnormal accumulation of disulfides within cells in environments lacking glucose, leading to contraction of the actin cytoskeleton, which ultimately triggers various signaling pathways and cell death. The induction of disulfide death in the treatment of cancer may exhibit significant therapeutic potential. Therefore, in the present review, a comprehensive and critical analysis of the current understanding of the molecular mechanisms and regulatory networks of disulfide death is presented. In addition, the potential physiological functions of disulfide death in tumor suppression and immune surveillance as well as its pathological roles and therapeutic potential are described. The core focus areas for future research into this form of cell death are also explored. Given the current lack of extensive clinical findings and well-defined key concepts, these may be regarded as pivotal points of interest in future studies.
Collapse
Affiliation(s)
- Xue Li
- Oncology Department, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| | - Danxia Zhu
- Oncology Department, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
28
|
Li Y, Zhang H, Yang F, Zhu D, Chen S, Wang Z, Wei Z, Yang Z, Jia J, Zhang Y, Wang D, Ma M, Kang X. Mechanisms and therapeutic potential of disulphidptosis in cancer. Cell Prolif 2025; 58:e13752. [PMID: 39354653 DOI: 10.1111/cpr.13752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
SLC7A11 plays a pivotal role in tumour development by facilitating cystine import to enhance glutathione synthesis and counteract oxidative stress. Disulphidptosis, an emerging form of cell death observed in cells with high expression of SLC7A11 under glucose deprivation, is regulated through reduction-oxidation reactions and disulphide bond formation. This process leads to contraction and collapse of the F-actin cytoskeleton from the plasma membrane, ultimately resulting in cellular demise. Compared to other forms of cell death, disulphidptosis exhibits distinctive characteristics and regulatory mechanisms. This mechanism provides novel insights and innovative strategies for cancer treatment while also inspiring potential therapeutic approaches for other diseases. Our review focuses on elucidating the molecular mechanism underlying disulphidptosis and its connection with the actin cytoskeleton, identifying alternative metabolic forms of cell death, as well as offering insights into disulphidptosis-based cancer therapy. A comprehensive understanding of disulphidptosis will contribute to our knowledge about fundamental cellular homeostasis and facilitate the development of groundbreaking therapies for disease treatment.
Collapse
Affiliation(s)
- Yanhu Li
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Haijun Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
- The Second People's Hospital of Gansu Province, Lanzhou, PR China
| | - Fengguang Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Daxue Zhu
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Shijie Chen
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhaoheng Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Ziyan Wei
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Zhili Yang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Jingwen Jia
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Yizhi Zhang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Dongxin Wang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Mingdong Ma
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| | - Xuewen Kang
- Lanzhou University Second Hospital, Lanzhou, PR China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou, PR China
| |
Collapse
|
29
|
Shi CJ, Pang FX, Lei YH, Deng LQ, Pan FZ, Liang ZQ, Xie T, Wu XL, Wang YY, Xian YF, Zeng WQ, Lin HL, Zhang JF. 5-methylcytosine methylation of MALAT1 promotes resistance to sorafenib in hepatocellular carcinoma through ELAVL1/SLC7A11-mediated ferroptosis. Drug Resist Updat 2025; 78:101181. [PMID: 39657434 DOI: 10.1016/j.drup.2024.101181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
Emerging evidence demonstrates that long non-coding RNAs (lncRNAs) play a crucial role in sorafenib resistance in hepatocellular carcinoma (HCC), and lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a dysregulated lncRNA in sorafenib-resistant HCC cells. However, the underlying regulatory mechanisms of MALAT1 in sorafenib-resistant HCC cells remain unclear. In the present study, we demonstrated that 5-methylcytosine (m5C) methylation catalyzed by NSUN2 and ALYREF contributed to the RNA stability and upregulation of MALAT1. The NSUN2/ALYREF/MALAT1 signaling axis was activated in sorafenib-resistant cells, and the upregulation of MALAT1 inhibited sorafenib-induced ferroptosis to drive sorafenib resistance. Mechanistically, MALAT1 maintained the mRNA stability of SLC7A11 by directly binding to ELAVL1 and stimulating its cytoplasmic translocation. Furthermore, we explored a new synergetic strategy for the treatment of HCC by combining MALAT1 inhibitor MALAT1-IN1 with sorafenib. The results demonstrated that MALAT1-IN1 significantly enhanced sorafenib efficacy for the treatment of HCC both in vitro and in vivo. Collectively, our work brings new insights into the epigenetic mechanisms of sorafenib resistance and offers an alternative therapeutic strategy targeting ferroptosis for sorafenib-resistant HCC patients.
Collapse
MESH Headings
- Sorafenib/pharmacology
- Sorafenib/therapeutic use
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Ferroptosis/drug effects
- Ferroptosis/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- 5-Methylcytosine/metabolism
- 5-Methylcytosine/analogs & derivatives
- ELAV-Like Protein 1/metabolism
- ELAV-Like Protein 1/genetics
- Amino Acid Transport System y+/genetics
- Amino Acid Transport System y+/metabolism
- Animals
- Mice
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Gene Expression Regulation, Neoplastic/drug effects
- Methylation/drug effects
- Cell Line, Tumor
Collapse
Affiliation(s)
- Chuan-Jian Shi
- Cancer Center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, PR China; Shenzhen Traditional Chinese Medicine Oncology Medical Center, Shenzhen, Guangdong 518000, PR China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Feng-Xiang Pang
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, PR China
| | - Yu-He Lei
- Research Institute, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, PR China
| | - Li-Qiang Deng
- Cancer Center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, PR China; Shenzhen Traditional Chinese Medicine Oncology Medical Center, Shenzhen, Guangdong 518000, PR China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Fu-Zhen Pan
- Cancer Center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, PR China; Shenzhen Traditional Chinese Medicine Oncology Medical Center, Shenzhen, Guangdong 518000, PR China
| | - Zhi-Qing Liang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 510086, PR China
| | - Tian Xie
- Research Institute, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, PR China
| | - Xian-Lin Wu
- Cancer Center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, PR China; Shenzhen Traditional Chinese Medicine Oncology Medical Center, Shenzhen, Guangdong 518000, PR China
| | - Yu-Yan Wang
- Cancer Center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, PR China; Shenzhen Traditional Chinese Medicine Oncology Medical Center, Shenzhen, Guangdong 518000, PR China
| | - Yan-Fang Xian
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Wei-Qiang Zeng
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Han-Li Lin
- Research Institute, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, PR China
| | - Jin-Fang Zhang
- Cancer Center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, PR China; Shenzhen Traditional Chinese Medicine Oncology Medical Center, Shenzhen, Guangdong 518000, PR China.
| |
Collapse
|
30
|
Chen YS, Lee CH, Hsieh YH, Chiou HL, Hung MC, Lee HL. Sorafenib, a Tyrosine Kinase Inhibitor, Synergistically Enhances the Ferroptosis Effects of Asiatic Acid in Hepatocellular Carcinoma Cells. ENVIRONMENTAL TOXICOLOGY 2025; 40:79-87. [PMID: 39264136 DOI: 10.1002/tox.24415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 09/13/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most common cancers worldwide. Asiatic acid (AA) is a natural triterpene, which is recognized as effect of antioxidant and antitumor. Sorafenib (Sor), an orally target drug, has been applicate for the HCC therapy. However, the synergistic effect of AA and Sor on human HCC is still unclear. Here, we explore the effect of combined treatment with AA and Sor in the HCC cell line SK-HEP-1 and HepG2. Compared with treating alone, our results demonstrated that AA combined with Sor synergistically inhibited proliferative rates in MTT assay and colony formation assay. We also found that AA combined with Sor in HCC cells strongly caused cell cycle arrest in G0/G1 phase and affected the protein level of cyclin D1 and SKP2. Furthermore, combination treatment strongly enhanced ferroptosis through cellular accumulation of iron ions, lipid peroxidation, and ferroptosis-related proteins (GPX4 and FTH1) in HCC cells. In addition, the combined treatment resulted in higher phosphorylation of JNK1/2 in the promotion of ferroptosis than drug treatment alone. These results indicate that AA combined with Sor synergistically improved ferroptosis in HCC cells through the regulation of JNK1/2 signaling. Taken together, the combinatorial strategy may serve as the potential treatment in HCC.
Collapse
Affiliation(s)
- Yong-Syuan Chen
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Hsing Lee
- Division of Pediatric Surgery, Children's Hospital of China Medical University, Taichung, Taiwan
- Department of Surgery, Children's Hospital of China Medical University, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Chun Hung
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiang-Lin Lee
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
31
|
Li Y, Liu C, Fang B, Chen X, Wang K, Xin H, Wang K, Yang SM. Ferroptosis, a therapeutic target for cardiovascular diseases, neurodegenerative diseases and cancer. J Transl Med 2024; 22:1137. [PMID: 39710702 DOI: 10.1186/s12967-024-05881-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The identification of ferroptosis represents a pivotal advancement in the field of cell death research, revealing an entirely novel mechanism of cellular demise and offering new insights into the initiation, progression, and therapeutic management of various diseases. Ferroptosis is predominantly induced by intracellular iron accumulation, lipid peroxidation, or impairments in the antioxidant defense system, culminating in membrane rupture and consequent cell death. Studies have associated ferroptosis with a wide range of diseases, and by enhancing our comprehension of its underlying mechanisms, we can formulate innovative therapeutic strategies, thereby providing renewed hope for patients.
Collapse
Affiliation(s)
- Yinghui Li
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Cuiyun Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Bo Fang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xinzhe Chen
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Kai Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266021, China.
| | - Kun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Su-Min Yang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
32
|
Liu R, Cui H, Li D, Guo X, Zhang Z, Tan S, Zhu X. Roles and Mechanisms of Ferroptosis in Sorafenib Resistance for Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:2493-2504. [PMID: 39717509 PMCID: PMC11665174 DOI: 10.2147/jhc.s500084] [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: 10/10/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent malignant tumor, characterized by a poor prognosis. In recent decades, both the incidence and mortality rates of HCC have risen sharply. Sorafenib has emerged as the first conventional drug approved by the US Food and Drug Administration for first-line treatment in advanced HCC patients due to its favorable safety profile. However, its effectiveness is severely hindered by acquired drug resistance, which leads to only approximately 30% of HCC patients benefited from sorafenib therapy. Sorafenib resistance involves various mechanisms that inhibit cellular uptake of iron and reactive oxygen species (ROS). Consequently, ferroptosis a novel form of cell death contingent upon the accumulation of intracellular iron and ROS plays a critical role in mediating sorafenib resistance through the Hippo YAP pathway or Keap1-Nrf2 system. This review aimed to comprehensively elucidate the mechanisms underlying sorafenib resistance in HCC, particularly focusing on ferroptosis and its pathways, to provide valuable insights into targeting ferroptosis or its pathways for sorafenib-resistant HCC treatment.
Collapse
Affiliation(s)
- Ruyuan Liu
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Huanyu Cui
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Di Li
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Xuefeng Guo
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Zhengbao Zhang
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Shengkui Tan
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
- Guangxi Clinical Medical Research Center for Hepatobiliary Diseases, the Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533000, People’s Republic of China
| | - Xiaonian Zhu
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, 541199, People’s Republic of China
| |
Collapse
|
33
|
Wang L, Zeng T, Wang Y, Wang G, Yu W, Zhang J, Shi Y, Li J, Ding J. K90 lactylation orchestrates YAP nuclear sequestration by impairing binding with exportin CRM1 and enhances HCC malignancy. Cancer Lett 2024; 611:217386. [PMID: 39645025 DOI: 10.1016/j.canlet.2024.217386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 09/13/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Affiliation(s)
- Li Wang
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, China.
| | - Tanlun Zeng
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, China
| | - Yichuan Wang
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, China
| | - Guang Wang
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, China
| | - Weichen Yu
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jian Zhang
- The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yihai Shi
- Department of Gastroenterology, Shanghai Pudong New Area Gongli Hospital, Shanghai, 200135, China
| | - Jixi Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Huashan Hospital, Shanghai Engineering Research Center of Industrial Microorganisms, Fudan University, Shanghai, 200438, China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
34
|
Lai HC, Weng JC, Huang HC, Ho JX, Kuo CL, Cheng JC, Huang ST. Solanum torvum induces ferroptosis to suppress hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118670. [PMID: 39117020 DOI: 10.1016/j.jep.2024.118670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Solanum torvum Sw. (ST) is used to clear heat toxins, promote blood circulation, and alleviate blood stasis. Therefore, this plant has traditionally been used as an ethnomedicine for common cold, chronic gastritis, and tumors. AIM OF THE STUDY This study aimed to elucidate the mechanism by which ST induces ferroptosis in hepatocellular carcinoma (HCC), the combination effect with lenvatinib, and the impact on lenvatinib-resistant cells. MATERIALS AND METHODS Cell viability assays were performed using different hepatoma cell lines treated with ST. Lipid peroxidation and iron assays were performed using flow cytometry. Molecules involved in the ferroptosis pathway were detected by Western blotting. Finally, a lenvatinib-resistant cell line was established to evaluate the antiproliferative effects of ST. RESULTS ST ethanol extract inhibited the growth of various hepatoma cell lines. A significant reduction in glutathione peroxidase 4 (GPX4) expression was observed following ST treatment, which was accompanied by increased lipid peroxidation and Fe2+ accumulation. ST induced ferroptosis mainly through heme oxygenase-1 (HO-1) expression. HO-1 knockdown reduced ST-induced lipid peroxidation and reversed GPX4 suppression. Acyl-CoA synthetase long-chain family member 4 (ACSL4) also participated in ST-induced ferroptosis. ST and lenvatinib combination showed an additive effect, and ST retained its potential anti-HCC efficacy in a lenvatinib-resistant cell line. CONCLUSION This study demonstrated that the ethanol extract of ST inhibits hepatoma cell growth by inducing ferroptosis. ST displayed an additive effect with lenvatinib in Hep 3B cells and showed remarkable anti-HCC activity in lenvatinib-resistant Hep 3B cells. Collectively, the study shows that ST might have the potential to reduce lenvatinib use in clinical practice and salvage cases of lenvatinib resistance.
Collapse
Affiliation(s)
- Hsiang-Chun Lai
- Graduate Institute of Chinese Medicine, School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Jui-Chun Weng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Hui-Chi Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Jin-Xuan Ho
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Chao-Lin Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan.
| | - Sheng-Teng Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan; Cancer Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
35
|
Rahdan F, Abedi F, Dianat-Moghadam H, Sani MZ, Taghizadeh M, Alizadeh E. Autophagy-based therapy for hepatocellular carcinoma: from standard treatments to combination therapy, oncolytic virotherapy, and targeted nanomedicines. Clin Exp Med 2024; 25:13. [PMID: 39621122 PMCID: PMC11611955 DOI: 10.1007/s10238-024-01527-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024]
Abstract
Human hepatocellular carcinoma (HCC) has been identified as a significant cause of mortality worldwide. In recent years, extensive research has been conducted to understand the underlying mechanisms of autophagy in the pathogenesis of the disease, with the aim of developing novel therapeutic agents. Targeting autophagy with conventional therapies in invasive HCC has opened up new opportunities for treatment. However, the emergence of resistance and the immunosuppressive tumor environment highlight the need for combination therapy or specific targeting, as well as an efficient drug delivery system to ensure targeted tumor areas receive sufficient doses without affecting normal cells or tissues. In this review, we discuss the findings of several studies that have explored autophagy as a potential therapeutic approach in HCC. We also outline the potential and limitations of standard therapies for autophagy modulation in HCC treatment. Additionally, we discuss how different combination therapies, nano-targeted strategies, and oncolytic virotherapy could enhance autophagy-based HCC treatment in future research.
Collapse
Affiliation(s)
- Fereshteh Rahdan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Abedi
- Clinical Research Development, Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran.
- Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran.
| | - Maryam Zamani Sani
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Taghizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
36
|
Li X, Zhou Z, Tao Y, He L, Zhan F, Li J. Linking homocysteine and ferroptosis in cardiovascular disease: insights and implications. Apoptosis 2024; 29:1944-1958. [PMID: 39044092 DOI: 10.1007/s10495-024-01999-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 07/25/2024]
Abstract
Homocysteine (Hcy) is a metabolic intermediate product derived from methionine. Hyperhomocysteinemia is a condition associated with various diseases. Hcy is recognized as a risk factor for cardiovascular disease (CVD). Ferroptosis, a novel form of cell death, is primarily characterized by substantial iron accumulation and lipid peroxidation. Recent research indicates a close association between ferroptosis and the pathophysiological processes of tumors, neurological diseases, CVD, and other ailments. However, limited research has been conducted on the impact of Hcy on ferroptosis. Therefore, this paper aimed to investigate the potential roles and mechanisms of homocysteine and ferroptosis in the context of cardiovascular disease. By conducting comprehensive literature research and analysis, we aimed to summarize recent advancements in understanding the effects of homocysteine on ferroptosis in cardiovascular diseases. This research contributes to a profound understanding of this critical domain.
Collapse
Affiliation(s)
- Xiaozhong Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China
| | - Zheng Zhou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China
| | - Yu Tao
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lei He
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Fenfang Zhan
- Jiangxi Key Laboratory of Molecular Medicine, Nanchang, 330006, China
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Juxiang Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
37
|
Li J, Ma X, Xu F, Yan Y, Chen W. Babaodan overcomes cisplatin resistance in cholangiocarcinoma via inhibiting YAP1. PHARMACEUTICAL BIOLOGY 2024; 62:314-325. [PMID: 38571483 PMCID: PMC10997361 DOI: 10.1080/13880209.2024.2331060] [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: 07/17/2023] [Accepted: 03/06/2024] [Indexed: 04/05/2024]
Abstract
CONTEXT Cholangiocarcinoma with highly heterogeneous, aggressive, and multidrug resistance has a poor prognosis. Although babaodan (BBD) combined with cisplatin improved non-small cell lung cancer efficacy, its impact on overcoming resistance in cholangiocarcinoma remains unexplored. OBJECTIVE This study explored the role and mechanism of BBD on cisplatin resistance in cholangiocarcinoma cells (CCAs). MATERIALS AND METHODS Cisplatin-resistant CCAs were exposed to varying concentrations of cisplatin (25-400 μg/mL) or BBD (0.25-1.00 mg/mL) for 48 h. IC50 values, inhibition ratios, apoptosis levels, DNA damage, glutathione (GSH) levels, oxidized forms of GSH, total GSH content, and glutaminase relative activity were evaluated using the cell counting kit 8, flow cytometry, comet assay, and relevant assay kits. RESULTS BBD-reduced the cisplatin IC50 in CCAs from 118.8 to 61.83 μg/mL, leading to increased inhibition rate, apoptosis, and DNA damage, and decreased expression of B-cell lymphoma-2, p-Yes-associated protein 1/Yes-associated protein 1, solute carrier family 1 member 5, activating transcription factor 4, and ERCC excision repair 1 in a dose-dependent manner with maximum reductions of 78.97%, 51.98%, 54.03%, 56.59%, and 63.22%, respectively; bcl2-associated X and gamma histone levels were increased by 0.43-115.77% and 22.15-53.39%. The impact of YAP1 knockdown on cisplatin-resistant CCAs resembled BBD. GSH, oxidized GSH species, total GSH content, and glutaminase activity in cisplatin-resistant CCAs with BBD treatment also decreased, while YAP1 overexpression countered BBD's effects. DISCUSSION AND CONCLUSION This study provides a scientific basis for BBD clinical application and provides a new direction for BBD biological mechanism research.
Collapse
Affiliation(s)
- Jiong Li
- Department of Traditional Chinese Medicine, The First People’s Hospital of Lin’an District, Hangzhou, China
| | - Xiangjun Ma
- Department of Traditional Chinese Medicine, The First People’s Hospital of Lin’an District, Hangzhou, China
| | - Faying Xu
- College of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weiqing Chen
- Department of General Surgery, The First People’s Hospital of Lin’an District, Hangzhou, China
| |
Collapse
|
38
|
Malnassy G, Ziolkowski L, Macleod KF, Oakes SA. The Integrated Stress Response in Pancreatic Development, Tissue Homeostasis, and Cancer. Gastroenterology 2024; 167:1292-1306. [PMID: 38768690 PMCID: PMC11570703 DOI: 10.1053/j.gastro.2024.05.009] [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: 12/14/2023] [Revised: 04/06/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
Present in all eukaryotic cells, the integrated stress response (ISR) is a highly coordinated signaling network that controls cellular behavior, metabolism, and survival in response to diverse stresses. The ISR is initiated when any 1 of 4 stress-sensing kinases (protein kinase R-like endoplasmic reticulum kinase [PERK], general control non-derepressible 2 [GCN2], double-stranded RNA-dependent protein kinase [PKR], heme-regulated eukaryotic translation initiation factor 2α kinase [HRI]) becomes activated to phosphorylate the protein translation initiation factor eukaryotic translation initiation factor 2α (eIF2α), shifting gene expression toward a comprehensive rewiring of cellular machinery to promote adaptation. Although the ISR has been shown to play an important role in the homeostasis of multiple tissues, evidence suggests that it is particularly crucial for the development and ongoing health of the pancreas. Among the most synthetically dynamic tissues in the body, the exocrine and endocrine pancreas relies heavily on the ISR to rapidly adjust cell function to meet the metabolic demands of the organism. The hardwiring of the ISR into normal pancreatic functions and adaptation to stress may explain why it is a commonly used pro-oncogenic and therapy-resistance mechanism in pancreatic ductal adenocarcinoma and pancreatic neuroendocrine tumors. Here, we review what is known about the key roles that the ISR plays in the development, homeostasis, and neoplasia of the pancreas.
Collapse
Affiliation(s)
- Greg Malnassy
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Leah Ziolkowski
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinoi; Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Kay F Macleod
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinoi; Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois; Committee on Cancer Biology, University of Chicago, Chicago, Illinois.
| | - Scott A Oakes
- Department of Pathology, University of Chicago, Chicago, Illinois; Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois; Committee on Cancer Biology, University of Chicago, Chicago, Illinois.
| |
Collapse
|
39
|
Xue X, Zhu X, Zhou L, Sun X, Gu M, Liang Y, Tan M, Hou Q, Wang S, Dai C. The Hippo Coactivator TAZ Exacerbates Cisplatin-Induced Acute Renal Injury. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:421-435. [PMID: 39664333 PMCID: PMC11631110 DOI: 10.1159/000540973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/12/2024] [Indexed: 12/13/2024]
Abstract
Introduction Transcriptional coactivator with PDZ-binding motif (TAZ), a Hippo signaling pathway effector, maintains the balance of cell proliferation, differentiation, and death. However, the role of TAZ in tubular cell survival and acute kidney injury (AKI) remains largely unknown. Methods We used the RNA-seq database, Western blot, and immunohistochemistry to examine TAZ expression in kidneys from cisplatin-induced AKI. We generated tubular-specific TAZ knockout mice to assess the role of TAZ in cisplatin-induced renal toxicity. Immunoprecipitation-mass spectrometry followed standard procedures. Results TAZ was activated in tubular cells in kidneys injected with cisplatin. Conditional deletion of TAZ in tubular cells confers ferroptosis resistance and protects kidneys from cisplatin-induced AKI, whereas overexpression of TAZ(S89A) exacerbates cisplatin-induced ferroptosis. Inhibition of ferroptosis with ferrostatin-1 potently preserves renal function and alleviates morphological injury and tubular cell ferroptosis induced by cisplatin. Mechanistically, in a PPARδ-dependent manner, but not TEAD, TAZ reduces the expression of glutathione peroxidase 4 (GPX4), thus exacerbating cisplatin-induced ferroptosis. Conclusions Our findings show that cisplatin-induced AKI and tubular cell ferroptosis are mediated by TAZ-PPARδ interaction through regulation of GPX4, highlighting TAZ as a potential therapeutic candidate for AKI.
Collapse
Affiliation(s)
- Xian Xue
- Center for Kidney Disease, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xingwen Zhu
- Department of Clinical Genetics, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Lu Zhou
- Department of Clinical Genetics, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoli Sun
- Center for Kidney Disease, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Mengru Gu
- Department of Clinical Genetics, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Liang
- Department of Clinical Genetics, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Mengzhu Tan
- Department of Clinical Genetics, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Qing Hou
- Department of Clinical Genetics, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Sudan Wang
- Department of Clinical Genetics, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Chunsun Dai
- Center for Kidney Disease, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Genetics, The 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Zhang C, Gao L, Zhang Y, Jin X, Wang M, Wang Q, Zhao W, Wu N, Zhang Y, Liu Y, Zhang Y, Ma L, Chen Y. Corosolic acid inhibits EMT in lung cancer cells by promoting YAP-mediated ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156110. [PMID: 39369568 DOI: 10.1016/j.phymed.2024.156110] [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: 07/08/2024] [Revised: 09/15/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Corosolic acid (CA), a naturally occurring pentacyclic triterpenoid is renowned for its anticancer attributes. Previous studies have predominantly centered on the anticancer properties of CA in lung cancer, specifically its role in inducing apoptosis, however, investigations regarding its involvement in ferroptosis have been scarce. METHODS The apoptotic and proliferative effects were evaluated by CCK8 and colony formation assay. Cell death and ROS generation were measured to assess the response of CA to iron death induction. Scratch and invasion assays were performed to verify the effect of CA on the invasive ability of lung cancer cells. Protein and mRNA expression were analyzed using Western blotting and qPCR. The CHX assay was carried out to detect protein half-life. Metabolite levels were measured with appropriate kits. Protein expression was detected through IF and IHC. A xenograft tumor model was established to investigate the inhibitory effect of CA on lung cancer in vivo. RESULTS The current findings revealed that CA exerts its anticancer effect by inducing cell death, accompanied by the accumulation of lipid reactive oxygen species (ROS), hinting at the possible involvement of ferroptosis. Our experimental results further substantiated the significance of ferroptosis in the CA anticancer mechanism, as ferroptosis inhibitors were found to effectively rescue CA-induced cell death. Significantly, we demonstrated for the first time that CA could induce ferroptosis further by suppressing EMT in lung cancer cells. Additionally, CA could regulate GPX4 to induce ferroptosis, interestingly, CA downregulated GSH synthetase by inhibiting YAP rather than GPX4, thereby reducing GSH, inducing ferroptosis, and further suppressing EMT in lung cancer cells.We also discovered that GSS is a crucial downstream target of YAP in regulating GSH. Moreover, a xenograft mouse model indicated that CA could trigger ferroptosis in lung cancer cells by regulating YAP expression and GSH levels. CONCLUSION CA inhibited lung cancer cell metastasis by inducing ferroptosis. Our data offer the first evidence that CA induces ferroptosis in lung cancer cells by regulating YAP/GSS to modulate GSH, thereby further suppressing EMT. These results imply the potential of CA as an inducer of ferroptosis to inhibit lung cancer metastasis.
Collapse
Affiliation(s)
- Congcong Zhang
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China
| | - Lingli Gao
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China
| | - Yinghui Zhang
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China
| | - Xiaoqin Jin
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China
| | - Mengyu Wang
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China.
| | - Qianna Wang
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China
| | - Wenyu Zhao
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China
| | - Nan Wu
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China
| | - Yasu Zhang
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China
| | - Yaru Liu
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China
| | - Yanyu Zhang
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China.
| | - Liangliang Ma
- Rehabilitation Center, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450014, PR China.
| | - Yulong Chen
- Rehabilition Medicine College, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, PR China.
| |
Collapse
|
41
|
Liu X, Liu T, Zhou Z, Bian K, Qiu C, Zhang F. Brusatol improves the efficacy of sorafenib in Huh7 cells via ferroptosis resistance dependent Nrf2 signaling pathway. Biochem Biophys Res Commun 2024; 734:150762. [PMID: 39353360 DOI: 10.1016/j.bbrc.2024.150762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common malignancy with a poor prognosis. The recommended treatment of unresectable HCC involves targeted therapy, for example sorafenib, combined with immunotherapy. A recent article reported that sorafenib could induce ferroptosis escape in HCC. Brusatol is a novel Nrf2 inhibitor that takes effects in various diseases. In our study, we aimed to identify whether the addition of Brusatol to sorafenib could reverse ferroptosis escape in Huh7 cells. METHODS The cultured Huh7 cells treated by sorafenib with or without Brusatol addition were harvested for ferroptotic phenotype experiments and ferroptosis-related markers such as GPX4 and SLC7A11 were detected. In vivo experiments were conducted to discover the effect of Brusatol in combination with sorafenib in liver tumor bearing mice. Mechanism signaling pathways were detected by RNA-sequencing. RESULTS Brusatol alone could induce Huh7 cell death and sorafenib could moderately mediate Huh7 cell ferroptosis by paradoxically inhibiting GPX4. However, sorafenib simultaneously upregulates Nrf2 signaling in Huh7 cells fighting against ferroptosis to result in sorafenib resistance. The addition of Brusatol could potentiate ferroptosis in Huh7 cells through downregulating Nrf2 and the downstream HO-1 and NQO1, thus enhancing the efficacy of sorafenib, which could be reversed by ferrostatin-1 treatment. CONCLUSION In conclusion, Brusatol improves the efficacy of sorafenib by inducing ferroptosis via hindering Nrf2 signaling activation in HCC.
Collapse
Affiliation(s)
- Xujin Liu
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China
| | - Tianyi Liu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, PR China
| | - Zhonghua Zhou
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China
| | - Kai Bian
- Department of Burn and Plastic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China; Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, PR China
| | - Cheng Qiu
- Department of Orthopaedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, Shandong, PR China
| | - Fan Zhang
- Department of Burn and Plastic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
42
|
Liao C, He Y, Luo X, Deng G. Ferroptosis: insight into the treatment of hepatocellular carcinoma. Cancer Cell Int 2024; 24:376. [PMID: 39538215 PMCID: PMC11562710 DOI: 10.1186/s12935-024-03559-z] [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: 03/27/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignances in the world, with high morbidity and mortality. Due to the hidden onset of symptoms, there are huge obstacles in early diagnosis, recurrence, metastasis and drug resistance. Although great strides have been made in the treatment of HCC, effective treatment options are still limited and achieving longer survival for patients remains urgent. Ferroptosis is a novel type of programmed cell death that is mainly caused by iron-dependent oxidative damage. With further investigations, ferroptosis has been proved to be associated with the occurrence and development of various tumors. This article reviews the regulatory mechanism and signal transduction pathways of ferroptosis, investigates the complex relationship between autophagy, sorafenib resistance and immunotherapy with ferroptosis involved in HCC, providing new ideas and directions for the treatment of HCC.
Collapse
Affiliation(s)
- Chuanjie Liao
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, China
| | - Youwu He
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, China
| | - Xinning Luo
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, China
| | - Ganlu Deng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, 530021, China.
| |
Collapse
|
43
|
Su L, Bu J, Yu J, Jin M, Meng G, Zhu X. Comprehensive review and updated analysis of DNA methylation in hepatocellular carcinoma: From basic research to clinical application. Clin Transl Med 2024; 14:e70066. [PMID: 39462685 PMCID: PMC11513202 DOI: 10.1002/ctm2.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary malignant tumour, ranking second in global mortality rates and posing significant health threats. Epigenetic alterations, particularly DNA methylation, have emerged as pivotal factors associated with HCC diagnosis, therapy, prognosis and malignant progression. However, a comprehensive analysis of the DNA methylation mechanism driving HCC progression and its potential as a therapeutic biomarker remains lacking. This review attempts to comprehensively summarise various aspects of DNA methylation, such as its mechanism, detection methods and biomarkers aiding in HCC diagnosis, treatment and prognostic assessment of HCC. It also explores the role of DNA methylation in regulating HCC's malignant progression and sorafenib resistance, alongside elaborating the therapeutic effects of DNA methyltransferase inhibitors on HCC. A detailed examination of these aspects underscores the significant research on DNA methylation in tumour cells to elucidate malignant progression mechanisms, identify diagnostic markers and develop new tumour-specific inhibitors for HCC. KEY POINTS: A comprehensive summary of various aspects of DNA methylation, such as its mechanism, detection methods and biomarkers aiding in diagnosis and treatment. The role of DNA methylation in regulating hepatocellular carcinoma's (HCC) malignant progression and sorafenib resistance, alongside elaborating therapeutic effects of DNA methyltransferase inhibitors. Deep research on DNA methylation is critical for discovering novel tumour-specific inhibitors for HCC.
Collapse
Affiliation(s)
- Lin Su
- Department of Pain ManagementShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiawen Bu
- Department of Colorectal SurgeryShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiahui Yu
- Department of UltrasoundShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Mila Jin
- Department of Operation RoomThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Guanliang Meng
- Department of UrologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xudong Zhu
- Department of OncologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
- Department of General SurgeryCancer Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
44
|
Wang S, Qin M, Fan X, Jiang C, Hou Q, Ye Z, Zhang X, Yang Y, Xiao J, Wallace K, Rastegar-Kashkooli Y, Peng Q, Jin D, Wang J, Wang M, Ding R, Tao J, Kim YT, Bhawal UK, Wang J, Chen X, Wang J. The role of metal ions in stroke: Current evidence and future perspectives. Ageing Res Rev 2024; 101:102498. [PMID: 39243890 DOI: 10.1016/j.arr.2024.102498] [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: 03/18/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Metal ions play a pivotal role in maintaining optimal brain function within the human body. Nevertheless, the accumulation of these ions can result in irregularities that lead to brain damage and dysfunction. Disruptions of metal ion homeostasis can result in various pathologies, including inflammation, redox dysregulation, and blood-brain barrier disruption. While research on metal ions has chiefly focused on neurodegenerative diseases, little attention has been given to their involvement in the onset and progression of stroke. Recent studies have identified cuproptosis and confirmed ferroptosis as significant factors in stroke pathology, underscoring the importance of metal ions in stroke pathology, including abnormal ion transport, neurotoxicity, blood-brain barrier damage, and cell death. Additionally, it provides an overview of contemporary metal ion chelators and detection techniques, which may offer novel approaches to stroke treatment.
Collapse
Affiliation(s)
- Shaoshuai Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Non-commissioned Officer School of Army Medical University, Shijiazhuang, Hebei 050000, China
| | - Mengzhe Qin
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Qingchuan Hou
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ziyi Ye
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinru Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yunfan Yang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jingyu Xiao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kevin Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qinfeng Peng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Dongqi Jin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruoqi Ding
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jin Tao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ujjal K Bhawal
- Center for Global Health Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India; Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba 271-8587, Japan
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
45
|
Zou Y, Yang A, Chen B, Deng X, Xie J, Dai D, Zhang J, Tang H, Wu T, Zhou Z, Xie X, Wang J. crVDAC3 alleviates ferroptosis by impeding HSPB1 ubiquitination and confers trastuzumab deruxtecan resistance in HER2-low breast cancer. Drug Resist Updat 2024; 77:101126. [PMID: 39243601 DOI: 10.1016/j.drup.2024.101126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 09/09/2024]
Abstract
AIMS With the wide application of trastuzumab deruxtecan (T-DXd), the survival of HER2-low breast cancer patients is dramatically improved. However, resistance to T-DXd still exists in a subset of patients, and the molecular mechanism remains unclear. METHODS An in vivo shRNA lentiviral library functional screening was performed to identify potential circular RNA (crRNA) that mediates T-DXd resistance. RNA pull-down, mass spectrometry, RNA immunoprecipitation, and co-immunoprecipitation assays were conducted to investigate the molecular mechanism. Ferroptosis was detected using C11-BODIPY, Liperfluo, FerroOrange staining, glutathione quantification, malondialdehyde quantification, and transmission electron microscopy. Molecular docking, virtual screening, and patient-derived xenograft (PDX) models were used to validate therapeutic agents. RESULTS VDAC3-derived crRNA (crVDAC3) ranked first in functional shRNA library screening. Knockdown of crVDAC3 increased the sensitivity of HER2-low breast cancer cells to T-DXd treatment. Further mechanistic research revealed that crVDAC3 specifically binds to HSPB1 protein and inhibits its ubiquitination degradation, leading to intracellular accumulation and increased levels of HSPB1 protein. Notably, suppression of crVDAC3 dramatically increases excessive ROS levels and labile iron pool accumulation. Inhibition of crVDAC3 induces ferroptosis in breast cancer cells by reducing HSPB1 expression, thereby mediating T-DXd resistance. Through virtual screening and experimental validation, we identified that paritaprevir could effectively bind to crVDAC3 and prevent its interaction with HSPB1 protein, thereby increasing ubiquitination degradation of HSPB1 protein to overcome T-DXd resistance. Finally, we validated the enhanced therapeutic efficacy of T-DXd by paritaprevir in a HER2-low PDX model. CONCLUSION This finding reveals the molecular mechanisms underlying T-DXd resistance in HER2-low breast cancer. Our study provides a new strategy to overcome T-DXd resistance by inhibiting the interaction between crVDAC3 and HSPB1 protein.
Collapse
Affiliation(s)
- Yutian Zou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Anli Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bo Chen
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xinpei Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jindong Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Danian Dai
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Jinhui Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Tao Wu
- Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, China.
| | - Zhigang Zhou
- Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, China.
| | - Xiaoming Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Jin Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
46
|
Wang M, Wang X, Wang Y, Gai Y, Ye J, Xu X, You X. Advances in the study of the mechanism of action of miR‑22 in liver lesions (Review). Oncol Lett 2024; 28:541. [PMID: 39310022 PMCID: PMC11413475 DOI: 10.3892/ol.2024.14674] [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: 05/10/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Globally, nearly 2 million deaths annually are attributed to the development of liver diseases, with liver cancer and cirrhosis being particularly prominent, which makes liver disease a significant global health concern. Cirrhosis is closely linked to the evolution of hepatitis, hepatic fibrosis and fatty liver. However, most liver diseases have an insidious onset, are challenging to treat and the prognosis and efficacy of current therapies are unsatisfactory, which can result in irreversible functional damage to the liver. Therefore, there is an urgent need to explore the molecular mechanisms underlying liver disease and identify new biomarkers and therapeutic targets. In previous years, microRNAs (miRs), a class of short non-coding RNAs comprising 17-25 nucleotides, have attracted attention for their roles in various types of liver diseases. Among them, miR-22 serves a unique role in mediating multiple pathway mechanisms and epigenetic modifications and can act both as an inhibitor of liver cancer and a metabolic blocker. Given its close association with the liver, several studies have reported that the differential expression of miR-22 regulates the metabolic process of liver cancer and is involved in the evolution of hepatic fibrosis and steatohepatitis, making it a potential target for early diagnosis and treatment. The present manuscript aimed to comprehensively review the key role of miR-22 in the evolution of liver diseases and offer valuable references and guidance for subsequent studies by identifying its specific mechanism of action and future development prospects.
Collapse
Affiliation(s)
- Minghe Wang
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Xuejing Wang
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Yanqi Wang
- College of Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Yikuo Gai
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Jingran Ye
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Xinyan Xu
- College of Second Clinical Medical, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Xue You
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong 272067, P.R. China
| |
Collapse
|
47
|
Zhang C, Wu Q, Yang H, Zhang H, Liu C, Yang B, Hu Q. Ferroptosis-related gene signature for predicting prognosis and identifying potential therapeutic drug in EGFR wild-type lung adenocarcinoma. Commun Biol 2024; 7:1416. [PMID: 39478024 PMCID: PMC11525656 DOI: 10.1038/s42003-024-07117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Epidermal growth factor receptor wild type lung adenocarcinoma (EGFRWT LUAD) still has limited treatment options and unsatisfactory clinical outcomes. Ferroptosis, as a form of cell death, has been reported to play a dual role in regulating tumor cell survival. In this study, we constructed a 3-ferroptosis-gene signature, FeSig, and verified its accuracy and efficacy in predicting EGFRWT LUAD prognosis at both the RNA and protein levels. Patients with higher FeSig scores were found to have worse clinical outcomes. Additionally, we explored the relationship between FeSig and tumor microenvironment, revealing that enhanced interactions between fibroblasts and tumor cells in FeSighigh patients causing tumor resistance to ferroptosis. To address this challenge, we screened potential drugs from NCI-60 (The US National Cancer Institute 60 human tumour cell line anticancer drug screen) and Connectivity map database, ultimately identifying 6-mercatopurine (6-MP) as a promising candidate. Both in vitro and in vivo experiments demonstrated its efficacy in treating FeSighigh EGFRWT LUAD tumor models. In summary, we develop a novel FeSig for predicting prognosis and guiding drug application.
Collapse
Affiliation(s)
- Chuankai Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qi Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hongwei Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hui Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Changqing Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bo Yang
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qingsong Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
| |
Collapse
|
48
|
Chi ZC. Progress in research of ferroptosis in gastrointestinal tumors. Shijie Huaren Xiaohua Zazhi 2024; 32:699-715. [DOI: 10.11569/wcjd.v32.i10.699] [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: 08/02/2024] [Revised: 09/04/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Ferroptosis is a non-apoptotic and oxidation-damaged regulated cell death caused by iron accumulation, lipid peroxidation, and subsequent plasma membrane rupture. Ferroptosis is the main cause of tissue damage caused by iron overload and lipid peroxidation. With the deepening of the research in recent years, the understanding of the occurrence and treatment of tumors has made a major breakthrough, which brings new strategies for anti-cancer treatment. This paper reviews the relationship between ferroptosis and gastrointestinal tumors, the research of ferroptosis in cancer prevention and treatment, and the role of ferroptosis in the prevention and treatment of gastrointestinal tumors.
Collapse
Affiliation(s)
- Zhao-Chun Chi
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266011, Shandong Province, China
| |
Collapse
|
49
|
Yang G, Yang Y, Song Z, Chen L, Liu F, Li Y, Jiang S, Xue S, Pei J, Wu Y, He Y, Chu B, Wu H. Spliceosomal GTPase Eftud2 deficiency-triggered ferroptosis leads to Purkinje cell degeneration. Neuron 2024; 112:3452-3469.e9. [PMID: 39153477 DOI: 10.1016/j.neuron.2024.07.020] [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: 03/21/2023] [Revised: 03/20/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024]
Abstract
Spliceosomal GTPase elongation factor Tu GTP binding domain containing 2 (EFTUD2) is a causative gene for mandibulofacial dysostosis with microcephaly (MFDM) syndrome comprising cerebellar hypoplasia and motor dysfunction. How EFTUD2 deficiency contributes to these symptoms remains elusive. Here, we demonstrate that specific ablation of Eftud2 in cerebellar Purkinje cells (PCs) in mice results in severe ferroptosis, PC degeneration, dyskinesia, and cerebellar atrophy, which recapitulates phenotypes observed in patients with MFDM. Mechanistically, Eftud2 promotes Scd1 and Gch1 expression, upregulates monounsaturated fatty acid phospholipids, and enhances antioxidant activity, thereby suppressing PC ferroptosis. Importantly, we identified transcription factor Atf4 as a downstream target to regulate anti-ferroptosis effects in PCs in a p53-independent manner. Inhibiting ferroptosis efficiently rescued cerebellar deficits in Eftud2 cKO mice. Our data reveal an important role of Eftud2 in maintaining PC survival, showing that pharmacologically or genetically inhibiting ferroptosis may be a promising therapeutic strategy for EFTUD2 deficiency-induced disorders.
Collapse
Affiliation(s)
- Guochao Yang
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China; Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, 226019 Nantong, China
| | - Yinghong Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250100 Jinan, China
| | - Zhihong Song
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Liping Chen
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Fengjiao Liu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Ying Li
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Shaofei Jiang
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Saisai Xue
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Jie Pei
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Yan Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | - Yuanlin He
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250100 Jinan, China.
| | - Haitao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China; Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, 226019 Nantong, China; Chinese Institute for Brain Research, 102206 Beijing, China.
| |
Collapse
|
50
|
Wu W, Wen F, Hu J, Li L. Overexpression of ATF4 Inhibits Ferroptosis to Alleviate Anxiety Disorders by Activating the TGF-β Signaling Pathway. Neuropsychiatr Dis Treat 2024; 20:1969-1983. [PMID: 39430656 PMCID: PMC11491069 DOI: 10.2147/ndt.s480782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024] Open
Abstract
Background Anxiety disorders seriously impair patients' mental health and quality of life, with limited effectiveness of current treatments. Dysregulation of activating transcription factor 4 (ATF4) is involved in various mental diseases, but the research on its potential roles in alleviating anxiety disorders remains limited. Methods ATF4 was screened out by bioinformatic analysis and its expression was verified in vivo. Mice were treated with 21 d of chronic restraint stress to establish the anxiety mice model. The anxiolytic effect of ATF4 was assessed by a battery of behavior tests and evaluation of hippocampal tissue damage after overexpressing ATF4. Ferroptosis-related indicators were detected by enzyme-linked immunosorbent assay and Western blotting. Then the transforming growth factor beta (TGF-β) signaling pathway was predicted as the downstream regulatory pathway of ATF4 by bioinformatic methods. Western blotting was conducted to detect the protein expression level of TGF-β1, small mothers against decapentaplegic 3 (Smad3), and phospho-Smad3 (p-Smad3). Results ATF4 was screened out as a ferroptosis-related anxiolytic gene after bioinformatics analysis and was down-regulated in the anxiety mice model. Mice with ATF4 overexpression spent more time in the open arms in the elevated plus-maze test, appeared more frequently in the central area in the open-field test, and decreased the immobility time in the forced swimming and tail suspension tests. Hippocampal tissue damage was alleviated, ferroptosis was suppressed, and the levels of TGF-β1 and p-Smad3/Smad3 were increased by AFT4 overexpression. Conclusion ATF4 overexpression can repress ferroptosis to improve anxiety disorders by activating the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Wentao Wu
- Department of Psychiatry, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Fei Wen
- Department of Psychiatry, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Jiaxin Hu
- Department of Psychiatry, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, People’s Republic of China
| | - Leijun Li
- Department of Psychiatry, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou City, Guangdong Province, People’s Republic of China
| |
Collapse
|