1
|
Wu Y, Zhu BT. Role of protein disulfide isomerase in mediating sulfasalazine-induced ferroptosis in HT22 cells: The PDI-NOS-NO-ROS/lipid-ROS cascade. Arch Biochem Biophys 2025; 768:110366. [PMID: 40023379 DOI: 10.1016/j.abb.2025.110366] [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: 11/03/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Ferroptosis is a form of regulated cell death resulting from excessive lipid peroxidation. Sulfasalazine (SAS), an anti-inflammatory drug, can induce ferroptosis through inhibiting the system Xc- and triggering glutathione depletion. SAS has attracted considerable interest in recent years because of its potential for repurposing as an anticancer agent. Our recent studies have shown that protein disulfide isomerase (PDI) is an upstream mediator of chemically-induced ferroptosis through catalyzing the dimerization of nitric oxide synthase (NOS) and NO accumulation in cultured HT22 hippocampal neuronal cells. The present study aims to investigate SAS-induced ferroptotic cell death in HT22 cells with a focus on determining the role of PDI in mediating SAS-induced ferroptosis. We find that SAS induces ferroptotic cell death in HT22 cells, which is accompanied by a time-dependent sequential increase in the accumulation of cellular NO, ROS and lipid-ROS. We find that treatment of HT22 cells with SAS activates PDI-mediated iNOS activation (dimerization) and NO accumulation. In addition, SAS also strongly upregulates iNOS protein levels in HT22 cells. PDI knockdown or pharmacological inhibition of PDI's activity each suppresses SAS-induced iNOS dimerization, which is associated with abrogation of SAS-induced accumulation of NO, ROS and lipid-ROS, and a strong protection against ferroptotic cell death. On the other hand, PDI activation through the use of a TrxR1 inhibitor can strongly sensitize cells to SAS-induced ferroptosis. Together, these experimental observations demonstrate a crucial role of PDI in SAS-induced ferroptosis in a cell culture model through the activation of the PDI → NOS → NO → ROS/lipid-ROS pathway. Insights gained from this study also provide effective strategies to selectively sensitizing human cancer cells to SAS-induced ferroptosis, such as through the use of NO-releasing agents or TrxR1 inhibitors.
Collapse
Affiliation(s)
- Yufei Wu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China; Shenzhen Bay Laboratory, Shenzhen, 518172, China.
| |
Collapse
|
2
|
Ye R, Mao YM, Fei YR, Shang Y, Zhang T, Zhang ZZ, Liu YL, Li JY, Chen SL, He YB. Targeting ferroptosis for the treatment of female reproductive system disorders. J Mol Med (Berl) 2025; 103:381-402. [PMID: 40100417 DOI: 10.1007/s00109-025-02528-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/28/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
Ferroptosis, a regulated form of cell death driven by iron-dependent lipid peroxidation, has emerged as a critical factor in female reproductive health and has been implicated in disorders such as polycystic ovary syndrome, premature ovarian insufficiency, endometriosis, and ovarian cancer. This review explores the intricate molecular mechanisms underlying ferroptosis, emphasizing its reliance on iron metabolism and oxidative stress, which disrupt key processes in reproductive tissues, including granulosa cell function, folliculogenesis, and embryo implantation. Increasing evidence linking ferroptosis to these conditions offers new therapeutic opportunities, with iron chelators, lipid peroxidation inhibitors, and antioxidants showing the potential to alleviate reproductive dysfunction by modulating ferroptotic pathways. In ovarian cancer, ferroptosis inducers combined with conventional cancer therapies, such as chemotherapy, provide promising strategies to overcome drug resistance. This review synthesizes current knowledge on ferroptosis and highlights its importance as a therapeutic target in reproductive health, emphasizing the need for further research to refine and expand treatment options, evaluate their applicability in clinical settings, and explore their role in fertility preservation. By advancing our understanding of ferroptosis regulation, these therapeutic approaches could lead to novel treatments for reproductive disorders and cancers, offering new hope for improving outcomes in women's health and cancer therapy.
Collapse
Affiliation(s)
- Rui Ye
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi-Ming Mao
- Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, Zhejiang Province, China
| | - Yi-Ran Fei
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Yue Shang
- Reproductive Center, Hainan Branch, Shanghai Children'S Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Ting Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhe-Zhong Zhang
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Yong-Lin Liu
- Reproductive Center, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jun-Yu Li
- Department of Pharmacy, Hainan Branch, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Shi-Liang Chen
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| | - Yi-Bo He
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| |
Collapse
|
3
|
Zhao L, Deng H, Zhang J, Zamboni N, Yang H, Gao Y, Yang Z, Xu D, Zhong H, van Geest G, Bruggmann R, Zhou Q, Schmid RA, Marti TM, Dorn P, Peng RW. Lactate dehydrogenase B noncanonically promotes ferroptosis defense in KRAS-driven lung cancer. Cell Death Differ 2025; 32:632-645. [PMID: 39643712 PMCID: PMC11982314 DOI: 10.1038/s41418-024-01427-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024] Open
Abstract
Ferroptosis is an oxidative, non-apoptotic cell death frequently inactivated in cancer, but the underlying mechanisms in oncogene-specific tumors remain poorly understood. Here, we discover that lactate dehydrogenase (LDH) B, but not the closely related LDHA, subunits of active LDH with a known function in glycolysis, noncanonically promotes ferroptosis defense in KRAS-driven lung cancer. Using murine models and human-derived tumor cell lines, we show that LDHB silencing impairs glutathione (GSH) levels and sensitizes cancer cells to blockade of either GSH biosynthesis or utilization by unleashing KRAS-specific, ferroptosis-catalyzed metabolic synthetic lethality, culminating in increased glutamine metabolism, oxidative phosphorylation (OXPHOS) and mitochondrial reactive oxygen species (mitoROS). We further show that LDHB suppression upregulates STAT1, a negative regulator of SLC7A11, thereby reducing SLC7A11-dependent GSH metabolism. Our study uncovers a previously undefined mechanism of ferroptosis resistance involving LDH isoenzymes and provides a novel rationale for exploiting oncogene-specific ferroptosis susceptibility to treat KRAS-driven lung cancer.
Collapse
Affiliation(s)
- Liang Zhao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Haibin Deng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Jingyi Zhang
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
| | - Haitang Yang
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanyun Gao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Zhang Yang
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou City, Fujian, China
| | - Duo Xu
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haiqing Zhong
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Geert van Geest
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Qinghua Zhou
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Ralph A Schmid
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Thomas M Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Ren-Wang Peng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| |
Collapse
|
4
|
Liang Y, Zhao Y, Qi Z, Li X, Zhao Y. Ferroptosis: CD8 +T cells' blade to destroy tumor cells or poison for self-destruction. Cell Death Discov 2025; 11:128. [PMID: 40169575 PMCID: PMC11962101 DOI: 10.1038/s41420-025-02415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
Ferroptosis represents an emerging, iron-dependent form of cell death driven by lipid peroxidation. In recent years, it has garnered significant attention in the realm of cancer immunotherapy, particularly in studies involving immune checkpoint inhibitors. This form of cell death not only enhances our comprehension of the tumor microenvironment but is also considered a promising therapeutic strategy to address tumor resistance, investigate immune activation mechanisms, and facilitate the development of cancer vaccines. The combination of immunotherapy with ferroptosis provides innovative targets and fresh perspectives for advancing cancer treatment. Nevertheless, tumor cells appear to possess a wider array of ferroptosis evasion strategies compared to CD8+T cells, which have been conclusively shown to be more vulnerable to ferroptosis. Furthermore, ferroptosis in the TME can create a favorable environment for tumor survival and invasion. Under this premise, both inducing tumor cell ferroptosis and inhibiting T cell ferroptosis will impact antitumor immunity to some extent, and even make the final result run counter to our therapeutic purpose. This paper systematically elucidates the dual-edged sword role of ferroptosis in the antitumor process of T cells, briefly outlining the complexity of ferroptosis within the TME. It explores potential side effects associated with ferroptosis-inducing therapies and critically considers the combined application of ferroptosis-based therapies with ICIs. Furthermore, it highlights the current challenges faced by this combined therapeutic approach and points out future directions for development.
Collapse
Affiliation(s)
- Yuan Liang
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhaoyang Qi
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinru Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
5
|
Guo W, Duan Z, Wu J, Zhou BP. Epithelial-mesenchymal transition promotes metabolic reprogramming to suppress ferroptosis. Semin Cancer Biol 2025; 112:20-35. [PMID: 40058616 DOI: 10.1016/j.semcancer.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular de-differentiation process that provides cells with the increased plasticity and stem cell-like traits required during embryonic development, tissue remodeling, wound healing and metastasis. Morphologically, EMT confers tumor cells with fibroblast-like properties that lead to the rearrangement of cytoskeleton (loss of stiffness) and decrease of membrane rigidity by incorporating high level of poly-unsaturated fatty acids (PUFA) in their phospholipid membrane. Although large amounts of PUFA in membrane reduces rigidity and offers capabilities for tumor cells with the unbridled ability to stretch, bend and twist in metastasis, these PUFA are highly susceptible to lipid peroxidation, which leads to the breakdown of membrane integrity and, ultimately results in ferroptosis. To escape the ferroptotic risk, EMT also triggers the rewiring of metabolic program, particularly in lipid metabolism, to enforce the epigenetic regulation of EMT and mitigate the potential damages from ferroptosis. Thus, the interplay among EMT, lipid metabolism, and ferroptosis highlights a new layer of intricated regulation in cancer biology and metastasis. Here we summarize the latest findings and discuss these mutual interactions. Finally, we provide perspectives of how these interplays contribute to cellular plasticity and ferroptosis resistance in metastatic tumor cells that can be explored for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Wenzheng Guo
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Zhibing Duan
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Jingjing Wu
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Binhua P Zhou
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
6
|
Chen L, Ning J, Linghu L, Tang J, Liu N, Long Y, Sun J, Lv C, Shi Y, Tao T, Xiao D, Cao Y, Wang X, Liu S, Li G, Zhang B, Tao Y. USP13 facilitates a ferroptosis-to-autophagy switch by activation of the NFE2L2/NRF2-SQSTM1/p62-KEAP1 axis dependent on the KRAS signaling pathway. Autophagy 2025; 21:565-582. [PMID: 39360581 PMCID: PMC11849926 DOI: 10.1080/15548627.2024.2410619] [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: 09/01/2023] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
Macroautophagy/autophagyis a lysosomal-regulated degradation process that participates incellular stress and then promotes cell survival or triggers celldeath. Ferroptosis was initially described as anautophagy-independent, iron-regulated, nonapoptotic cell death.However, recent studies have revealed that autophagy is positivelyassociated with sensitivity to ferroptosis. Nonetheless, themolecular mechanisms by which these two types of regulated cell death(RCD) modulate each other remain largely unclear. Here, we screened85 deubiquitinating enzymes (DUBs) and found that overexpression ofUSP13 (ubiquitin specific peptidase 13) could significantlyupregulate NFE2L2/NRF2 (NFE2 like bZIP transcription factor 2)protein levels. In addition, in 39 cases of KRAS-mutated lungadenocarcinoma (LUAD), we found that approximately 76% of USP13overexpression is positively correlated with NFE2L2 overexpression.USP13 interacts with and catalyzes the deubiquitination of thetranscription factor NFE2L2. Additionally, USP13 depletion promotesan autophagy-to-ferroptosis switch invitro andin xenograft tumor mouse models, through the activation of theNFE2L2-SQSTM1/p62 (sequestosome 1)-KEAP1 axis in KRAS mutant cellsand tumor tissues. Hence, targeting USP13 effectively switchedautophagy-to-ferroptosis, thereby inhibiting KRAS (KRASproto-oncogene, GTPase) mutant LUAD, suggesting the therapeuticpromise of combining autophagy and ferroptosis in the KRAS-mutantLUAD.Abbreviation: ACSL4: acyl-CoA synthetase long-chain family member 4; ACTB: actin beta; AL: autolysosomes; AP: autophagosomes; BCL2L1/BCL-xL: BCL2 like 1; CCK8: Cell Counting Kit-8; CQ: chloroquine; CUL3: cullin 3; DMSO: dimethyl sulfoxide; DOX: doxorubicin; DUB: deubiquitinating enzyme; Ferr-1: ferrostatin-1; GPX4: glutathione peroxidase 4; GSEA: gene set enrichment analysis; 4HNE: 4-hydroxynonenal; IKE: imidazole ketone erastin; KEAP1: kelch like ECH associated protein 1; KRAS: KRAS proto-oncogene, GTPase; LCSC: lung squamous cell carcinoma; IF: immunofluorescence; LUAD: lung adenocarcinoma; Lys05: Lys01 trihydrochloride; MAPK1/ERK2/p42: mitogen-activated protein kinase 1; MAPK3/ERK1/p44; MTOR: mechanistic target of rapamycin kinase; NFE2L2/NRF2: NFE2 like bZIP transcription factor, 2; NQO1: NAD(P)H quinone dehydrogenase 1; PG: phagophore; RCD: regulated cell death; RAPA: rapamycin; ROS: reactive oxygen species; SLC7A11/xCT: solute carrier family 7 member 11; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; TUBB/beta-tubulin: tubulin, beta; UPS: ubiquitin-proteasome system; USP13: ubiquitin specific peptidase 13.
Collapse
Affiliation(s)
- Ling Chen
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jieling Ning
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Histology and Embryology, School of Basic Medicine, Central South University, Changsha, China
| | - Li Linghu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jun Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Na Liu
- Department of Neurosurgery, Postdoctoral Research Workstation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yao Long
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jingyue Sun
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Cairui Lv
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ying Shi
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Tania Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangjian Li
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Kunming, China
| | - Bin Zhang
- Department of Histology and Embryology, School of Basic Medicine, Central South University, Changsha, China
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Chen J, Yuan M, Wang J. Research progress of cysteine transporter SLC7A11 in endocrine and metabolic diseases. Mol Biol Rep 2025; 52:185. [PMID: 39899147 DOI: 10.1007/s11033-024-10193-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025]
Abstract
SLC7A11, often called xCT, belongs to the SLC family of transporters, which mediates the cellular influx of cystine and the efflux of glutamate. These transport processes are crucial for synthesizing GSH, enhancing the cell's ability to mitigate oxidative stress (OS). Emerging studies highlight the pivotal role of OS in triggering and exacerbating various metabolic and endocrine disorders, underlining the critical importance of regulating SLC7A11 expression levels. This study reviews the diverse roles of SLC7A11 in endocrine and metabolic diseases, examining its relationship with the metabolism of three key nutrients: proteins and amino acids, carbohydrates, and lipids. Additionally, the involvement of SLC7A11 in the onset and development of various common endocrine and metabolic disorders is analyzed. Additionally, it provides an overview of the current clinical and experimental use of SLC7A11 inhibitors and agonists. This review aims to offer insightful perspectives into the involvement of SLC7A11 in endocrine and metabolic pathologies and to foster the development of innovative therapeutic strategies that target SLC7A11.
Collapse
Affiliation(s)
- Jiaqi Chen
- Department of Endocrinology and Metabolic Disease, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China.
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Mengzhu Yuan
- Department of Endocrinology and Metabolic Disease, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jianping Wang
- Department of Endocrinology and Metabolic Disease, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China.
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
8
|
Zhang S, Ge Y, Liu J, Lu K. Poor therapeutic outcomes in KRAS-mutant non-small cell lung cancer due to chemoresistance conferred by SLC7A11. Clin Transl Oncol 2025; 27:494-506. [PMID: 39060774 DOI: 10.1007/s12094-024-03592-4] [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: 06/04/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024]
Abstract
PURPOSE This study aimed to confirm whether Kirsten rat sarcoma viral oncogene (KRAS) mutations affect the therapeutic efficacy of non-small cell lung cancer (NSCLC) and, if so, to explore what the possible mechanisms might be. METHODS We retrospectively analyzed the efficacy of immunochemotherapy in KRAS-mutant NSCLC patients compared to driver-negative patients. Online data platforms were used to find immunotherapy cases, and survival analysis compared treatments' efficacy. Cytotoxicity assays measured chemosensitivity in KRAS-mutant versus wild-type NSCLC to drugs like paclitaxel, carboplatin, and pemetrexed. Bioinformatics confirmed the KRAS-SLC7A11 link and cell experiments tested SLC7A11's role in chemoresistance. Animal studies verified the antitumor effects of SLC7A11 inhibitors with chemotherapy. RESULTS Patients with KRAS-mutated NSCLC have a shorter therapeutic effectiveness duration with immunochemotherapy than patients with driver gene-negative status. The efficacy of immunotherapy alone is similar between the two groups. The KRAS mutation can enhance chemoresistance by upregulating SLC7A11, and inhibiting SLC7A11 can increase the sensitivity of KRAS-mutated NSCLC to chemotherapy. CONCLUSION This study suggests that KRAS-mutant NSCLC can enhance its acquired chemoresistance by overexpressing SLC7A11, leading to poorer therapeutic outcomes. Targeting the KRAS-SLC7A11 axis could increase sensitivity to chemotherapeutic drugs, providing theoretical support for future treatment directions.
Collapse
Affiliation(s)
- Shiyu Zhang
- Department of Oncology, First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Yutong Ge
- Department of Oncology, First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Jingwen Liu
- Department of Oncology, First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Kaihua Lu
- Department of Oncology, First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China.
| |
Collapse
|
9
|
Jang N, Kim IK, Jung D, Chung Y, Kang YP. Regulation of Ferroptosis in Cancer and Immune Cells. Immune Netw 2025; 25:e6. [PMID: 40078787 PMCID: PMC11896659 DOI: 10.4110/in.2025.25.e6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
Ferroptosis, an iron-dependent form of regulated cell death, is driven by lipid peroxidation and shaped by metabolic and antioxidant pathways. In immune cells, ferroptosis susceptibility varies by cell types, lipid composition, and metabolic demands, influencing immune responses in cancer, infections, and autoimmune diseases. Therapeutically, targeting ferroptosis holds promise in cancer immunotherapy by enhancing antitumor immunity or inhibiting immunosuppressive cells. This review highlights the metabolic pathways underlying ferroptosis, its regulation in immune cells, its dual role in tumor progression and antitumor immunity, and its context-dependent therapeutic implications for optimizing cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Yeonseok Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
10
|
Bai X, Kang J, Wei S, Wang Y, Liu Y, Yuan B, Lu Q, Li H, Yan J, Yang X, Chang J. A pH responsive nanocomposite for combination sonodynamic-immunotherapy with ferroptosis and calcium ion overload via SLC7A11/ACSL4/LPCAT3 pathway. EXPLORATION (BEIJING, CHINA) 2025; 5:20240002. [PMID: 40040833 PMCID: PMC11875445 DOI: 10.1002/exp.20240002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/24/2024] [Indexed: 03/06/2025]
Abstract
Sonodynamic therapy offers a non-invasive approach to induce the death of tumor cells. By harnessing ultrasound waves in tandem with sonosensitizers, this method produces reactive oxygen species (ROS) that inflict oxidative damage upon tumor cells, subsequently causing their demise. Ferroptosis is a regulatory form of cell death that differs from other forms, characterized by iron accumulation, ROS accumulation, and lipid peroxidation. In the presented research, a nanoparticle formulation, parthenolide/ICG-CaCO3@lipid (PTL/ICG-CaCO3@Lip), has been engineered to amplify ferroptosis in tumor cells, positioning it as a potent agent for sonodynamic cancer immunotherapy. This nanoparticle significantly augments ROS levels within tumor cells, inducing oxidative stress that leads to cell death. The therapeutic potential of PTL/ICG-CaCO3@Lip, both in vivo and in vitro, has been convincingly demonstrated. Furthermore, RNA-seq analysis insights revealed that PTL/ICG-CaCO3@Lip facilitates tumor cell ferroptosis by regulating P53 to downregulate SLC7A11 protein expression, thereby inhibiting the glutamate-cystine antiporter system Xc- and stimulating ACSL4/LPCAT3 pathways. This pioneering work uncovers an innovative strategy for combatting tumors, leveraging enhanced oxidative stress to promote cell ferroptosis, and paves the way for groundbreaking cancer immunotherapeutic interventions.
Collapse
Affiliation(s)
- Xue Bai
- School of Life SciencesTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin UniversityTianjinChina
| | - Jun Kang
- School of Life SciencesTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin UniversityTianjinChina
- Department of Biological SciencesUniversity of Toronto ScarboroughTorontoCanada
| | | | - Yun Wang
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
| | - Yangsui Liu
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
| | - Bo Yuan
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
| | - Qian Lu
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
- Hepatopancreatobiliary CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
- School of Clinical MedicineTsinghua UniversityBeijingChina
| | - Huansong Li
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
| | - Jun Yan
- Hepatobiliary Pancreatic CenterXuzhou Central HospitalXuzhouChina
- Hepatopancreatobiliary CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
- School of Clinical MedicineTsinghua UniversityBeijingChina
| | - Xi Yang
- Department of Oral Maxillofacial‐Head and Neck OncologyShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
| | - Jin Chang
- School of Life SciencesTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular StructuresSchool of Life SciencesTianjin UniversityTianjinChina
| |
Collapse
|
11
|
Tian RL, Wang TX, Huang ZX, Yang Z, Guan KL, Xiong Y, Wang P, Ye D. Temsirolimus inhibits FSP1 enzyme activity to induce ferroptosis and restrain liver cancer progression. J Mol Cell Biol 2025; 16:mjae036. [PMID: 39313331 PMCID: PMC11879044 DOI: 10.1093/jmcb/mjae036] [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: 02/28/2024] [Revised: 06/02/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024] Open
Abstract
Ferroptosis is a non-apoptotic mode of cell death characterized by iron-dependent accumulation of lipid peroxidation. While lipid radical elimination reaction catalyzed by glutathione peroxidase 4 (GPX4) is a major anti-ferroptosis mechanism, inhibiting this pathway pharmaceutically shows promise as an antitumor strategy. However, certain tumor cells exhibit redundancy in lipid radical elimination pathways, rendering them unresponsive to GPX4 inhibitors. In this study, we conducted screens across different cancer cell lines and Food and Drug Administration-approved drugs, leading to the identification of temsirolimus in combination with the GPX4 inhibitor RSL3 as a potent inducer of ferroptosis in liver cancer cells. Mechanistically, temsirolimus sensitized liver cancer cells to ferroptosis by directly binding to and inhibiting ferroptosis suppressor protein 1 (FSP1) enzyme. Notably, while temsirolimus is recognized as a potent mammalian target of rapamycin (mTOR) inhibitor, its ferroptosis-inducing effect is primarily attributed to the inhibition of FSP1 rather than mTOR activity. By employing in vitro colony formation assays and in vivo tumor xenograft models, we demonstrated that the combination of temsirolimus and RSL3 effectively suppressed liver tumor progression. This tumoricidal effect was associated with increased lipid peroxidation and induction of ferroptosis. In conclusion, our findings underscore the potential of combining multitarget ferroptosis-inducing agents to circumvent the resistance to ferroptosis of liver cancer cells and highlight temsirolimus as a promising FSP1 inhibitor and ferroptosis inducer, which also deserves further investigation in translational medicine.
Collapse
Affiliation(s)
- Rui-Lin Tian
- Huashan Hospital, Fudan University; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Key Laboratory of Metabolism and Molecular Medicine (Ministry of Education), and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Tian-Xiang Wang
- School of Life Sciences, Westlake University, and Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Zi-Xuan Huang
- Huashan Hospital, Fudan University; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Key Laboratory of Metabolism and Molecular Medicine (Ministry of Education), and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Zhen Yang
- Center for Medical Research and Innovation of Pudong Hospital, and Intelligent Medicine Institute, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Kun-Liang Guan
- School of Life Sciences, Westlake University, and Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
| | - Yue Xiong
- Cullgen Inc., 12730 High Bluff Drive, San Diego, CA 92130, USA
| | - Pu Wang
- Huashan Hospital, Fudan University; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Key Laboratory of Metabolism and Molecular Medicine (Ministry of Education), and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Dan Ye
- Huashan Hospital, Fudan University; Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Key Laboratory of Metabolism and Molecular Medicine (Ministry of Education), and Molecular and Cell Biology Lab, Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Cao J, Zhou T, Wu T, Lin R, Huang J, Shi D, Yu J, Ren Y, Qian C, He L, Wu G, Dong Z, Yuan S, Gu H. Targeting estrogen-regulated system x c- promotes ferroptosis and endocrine sensitivity of ER+ breast cancer. Cell Death Dis 2025; 16:30. [PMID: 39833180 PMCID: PMC11756422 DOI: 10.1038/s41419-025-07354-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Estrogen receptor positive (ER+) breast cancer accounts for approximately 70% of cases. Endocrine therapies targeting estrogen are the first line therapies for ER+ breast cancer. However, resistance to these therapies occurs in about half of patients, leading to decreased survival rates. Inducing ferroptosis is a promising therapeutic strategy for cancer treatment for refractory and malignant cancers including triple-negative breast cancer. Nevertheless, ER+ breast cancer is relatively resistant to ferroptosis inducers. Here, we uncovered that ERα suppressed ferroptosis in ER+ breast cancer. Silencing ERα triggered ferroptosis, which was attenuated by ferroptosis inhibitor Ferrostatin-1, and was enhanced by ferroptosis inducer Erastin. Mechanistically, ERα transcriptionally upregulated the expression of SLC7A11 and SLC3A2, two subunits of the system xc-, which is one key inhibitory regulator of ferroptosis. Overexpression of the exogenous SLC7A11 and SLC3A2 was able to mitigate ferroptosis induced by ERα inhibition. Moreover, SLC7A11 and SLC3A2 levels were elevated in endocrine-resistant breast cancer cells and tumors. Importantly, the system xc- inhibitor Sorafenib or Imidazole ketone erastin effectively inhibited the growth of tamoxifen-resistant breast cells in vitro and in vivo. In conclusion, our data reveal that targeting estrogen-regulated SLC7A11 and SLC3A2 enhances ferroptosis in ER+ breast cancer, offering a novel therapeutic option for patients with ER+ breast cancer, particularly those with endocrine resistance.
Collapse
Affiliation(s)
- Jiawei Cao
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China.
| | - Tong Zhou
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China
| | - Tao Wu
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China
| | - Rixu Lin
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, 325035, Wenzhou, China
| | - Ju Huang
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China
| | - Dejin Shi
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China
| | - Jiawei Yu
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China
| | - Yinrui Ren
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China
| | - Changrui Qian
- School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Licai He
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China
| | - Guang Wu
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China
| | - Zhixiong Dong
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China
| | - Shaofei Yuan
- Department of Medical Oncology, Rui'an People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, 325200, Wenzhou, China.
| | - Haihua Gu
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education, Wenzhou Medical University, 325035, Wenzhou, China.
| |
Collapse
|
13
|
Tang J, Liu W, Li Z, Shen C, Zhang L, Wang C, Wang F, Zai Z, Qian X, Hu W, Zhang X, Peng X, Xu Y, Chen F. Inhibition of ASIC1a reduces ferroptosis in rheumatoid arthritis articular chondrocytes via the p53/NRF2/SLC7A11 pathway. FASEB J 2025; 39:e70298. [PMID: 39760183 PMCID: PMC11712546 DOI: 10.1096/fj.202402134rr] [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: 09/09/2024] [Revised: 12/15/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
The activation of acid-sensing ion channel 1a (ASIC1a) in response to extracellular acidification leads to an increase in extracellular calcium influx, thereby exacerbating the degeneration of articular chondrocytes in rheumatoid arthritis (RA). It has been suggested that the inhibition of extracellular calcium influx could potentially impede chondrocyte ferroptosis. The cystine transporter, solute carrier family 7 member 11 (SLC7A11), is recognized as a key regulator of ferroptosis. Recent studies suggest that the tumor suppressor gene p53 facilitates the induction of ferroptosis by suppressing the upregulation of SLC7A11. This process is mediated by the nuclear factor erythroid 2-related factor 2 (NRF2), a key transcription factor integral to the maintenance of cellular redox homeostasis and the regulation of inflammatory responses. This study aims to investigate the role of ASIC1a in the ferroptosis of RA chondrocytes and to determine the involvement of the p53/NRF2/SLC7A11 pathway in its underlying mechanism. In vitro experiments revealed that acidosis induces ferroptosis and reduces the expression of NRF2 and SLC7A11 in chondrocytes. Moreover, acidification significantly increased p53 protein levels in chondrocytes. Pifithrin-α (PFN-α), a p53 inhibitor, mitigated acidosis-induced ferroptosis and restored the diminished expression of NRF2 and SLC7A11. Furthermore, PcTx-1, an ASIC1a inhibitor, inhibited acidification-induced ferroptosis, enhanced the protein levels of SLC7A11 and NRF2, and reduced p53 expression. In vivo experiments demonstrated that the ASIC1a-specific inhibitor PcTx-1 ameliorated histopathological characteristics of ankle joints in collagen-induced arthritis (CIA) mice, decreased p53 expression, and enhanced NRF2 and SLC7A11 expression in chondrocytes. These findings suggest that ASIC1a inhibition may mitigate acidification-induced ferroptosis in articular chondrocytes in RA, potentially via the p53/NRF2/SLC7A11 pathway.
Collapse
Affiliation(s)
- Jie Tang
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Wenqiang Liu
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Zihan Li
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Can Shen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Longbiao Zhang
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Cheng Wang
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Fengshuo Wang
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Zhuoyan Zai
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Xuewen Qian
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Weirong Hu
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Xiaoyue Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Xiaoqing Peng
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| | - Yayun Xu
- Shenzhen Institute of Translational MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's HospitalShenzhenChina
| | - Feihu Chen
- School of PharmacyAnhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐Inflammatory and Immune MedicinesMinistry of EducationHefeiChina
| |
Collapse
|
14
|
Su C, Xue Y, Fan S, Sun X, Si Q, Gu Z, Wang J, Deng R. Ferroptosis and its relationship with cancer. Front Cell Dev Biol 2025; 12:1423869. [PMID: 39877159 PMCID: PMC11772186 DOI: 10.3389/fcell.2024.1423869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Marked by iron buildup and lipid peroxidation, ferroptosis is a relatively new regulatory cell death (RCD) pathway. Many diseases like cancer, myocardial ischemia-reperfusion injury (MIRI), neurological disorders and acute renal failure (AKI) are corelated with ferroptosis. The main molecular processes of ferroptosis discovered yet will be presented here, along with the approaches in which it interacts with tumour-associated signaling pathways and its uses in systemic therapy, radiation therapy, and immunotherapy managing tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Runzhi Deng
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| |
Collapse
|
15
|
Ma Q, Zhang W, Wu K, Shi L. The roles of KRAS in cancer metabolism, tumor microenvironment and clinical therapy. Mol Cancer 2025; 24:14. [PMID: 39806421 PMCID: PMC11727292 DOI: 10.1186/s12943-024-02218-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
KRAS is one of the most mutated genes, driving alternations in metabolic pathways that include enhanced nutrient uptaking, increased glycolysis, elevated glutaminolysis, and heightened synthesis of fatty acids and nucleotides. However, the beyond mechanisms of KRAS-modulated cancer metabolisms remain incompletely understood. In this review, we aim to summarize current knowledge on KRAS-related metabolic alterations in cancer cells and explore the prevalence and significance of KRAS mutation in shaping the tumor microenvironment and influencing epigenetic modification via various molecular activities. Given that cancer cells rely on these metabolic changes to sustain cell growth and survival, targeting these processes may represent a promising therapeutic strategy for KRAS-driven cancers.
Collapse
Affiliation(s)
- Qinglong Ma
- RNA Oncology Group, School of Public Health, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Wenyang Zhang
- RNA Oncology Group, School of Public Health, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Lei Shi
- RNA Oncology Group, School of Public Health, Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK.
| |
Collapse
|
16
|
Shi Y, Zheng H, Wang T, Zhou S, Zhao S, Li M, Cao B. Targeting KRAS: from metabolic regulation to cancer treatment. Mol Cancer 2025; 24:9. [PMID: 39799325 PMCID: PMC11724471 DOI: 10.1186/s12943-024-02216-3] [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: 09/06/2024] [Accepted: 12/25/2024] [Indexed: 01/15/2025] Open
Abstract
The Kirsten rat sarcoma viral oncogene homolog (KRAS) protein plays a key pathogenic role in oncogenesis, cancer progression, and metastasis. Numerous studies have explored the role of metabolic alterations in KRAS-driven cancers, providing a scientific rationale for targeting metabolism in cancer treatment. The development of KRAS-specific inhibitors has also garnered considerable attention, partly due to the challenge of acquired treatment resistance. Here, we review the metabolic reprogramming of glucose, glutamine, and lipids regulated by oncogenic KRAS, with an emphasis on recent insights into the relationship between changes in metabolic mechanisms driven by KRAS mutant and related advances in targeted therapy. We also focus on advances in KRAS inhibitor discovery and related treatment strategies in colorectal, pancreatic, and non-small cell lung cancer, including current clinical trials. Therefore, this review provides an overview of the current understanding of metabolic mechanisms associated with KRAS mutation and related therapeutic strategies, aiming to facilitate the understanding of current challenges in KRAS-driven cancer and to support the investigation of therapeutic strategies.
Collapse
Affiliation(s)
- Yanyan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, China
| | - Huiling Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing, 100191, China
| | - Tianzhen Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction (Peking University), Peking University Third Hospital, Ministry of Education, Beijing, 100191, China
| | - Shengpu Zhou
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, China
| | - Shiqing Zhao
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, China
| | - Mo Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction (Peking University), Peking University Third Hospital, Ministry of Education, Beijing, 100191, China.
| | - Baoshan Cao
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
17
|
Meng Y, Zhou Q, Dian Y, Zeng F, Deng G, Chen X. Ferroptosis: A Targetable Vulnerability for Melanoma Treatment. J Invest Dermatol 2025:S0022-202X(24)03024-0. [PMID: 39797894 DOI: 10.1016/j.jid.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 01/13/2025]
Abstract
Melanoma is a devastating form of skin cancer characterized by a high mutational burden, limited treatment success, and dismal prognosis. Although immunotherapy and targeted therapies have significantly revolutionized melanoma treatment, the majority of patients fail to achieve durable responses, highlighting the urgent need for novel therapeutic strategies. Ferroptosis, an iron-dependent form of regulated cell death driven by the overwhelming accumulation of lipid peroxides, has emerged as a promising therapeutic approach in preclinical melanoma models. A deeper understanding of the ferroptosis landscape in melanoma based on its biology characteristics, including phenotypic plasticity, metabolic state, genomic alterations, and epigenetic changes, as well as the complex role and mechanisms of ferroptosis in immune cells could provide a foundation for developing effective treatments. In this review, we outline the molecular mechanisms of ferroptosis, decipher the role of melanoma biology in ferroptosis regulation, reveal the therapeutic potential of ferroptosis in melanoma, and discuss the pressing questions that should guide future investigations into ferroptosis in melanoma.
Collapse
Affiliation(s)
- Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China; Furong Laboratory, Changsha, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| |
Collapse
|
18
|
Su Z, Liu Y, Wang L, Gu W. Regulation of SLC7A11 as an unconventional checkpoint in tumorigenesis through ferroptosis. Genes Dis 2025; 12:101254. [PMID: 39569390 PMCID: PMC11577153 DOI: 10.1016/j.gendis.2024.101254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 11/22/2024] Open
Abstract
Although cell-cycle arrest, senescence, and apoptosis are well accepted as the classic barriers in tumorigenesis, recent studies indicate that metabolic regulation is equally important as a major checkpoint in cancer development. It is well accepted that ferroptosis, an iron-dependent programmed cell death, acts as a new type of tumor suppression mechanism tightly linked with numerous metabolic pathways. SLC7A11 is a transmembrane cystine/glutamate transporter protein that plays a vital role in controlling ferroptosis in vivo. The levels of SLC7A11 are dynamically regulated by various types of stresses, such as oxidative stress, nutrient deprivation, endoplasmic reticulum stress, radiation, oncogenic stress, DNA damage, and immune stress. SLC7A11 can be transcriptionally regulated by both activators such as ATF4, NRF2, and ETS1, and repressors including BACH1, p53, ATF3, and STAT1 during stress responses. Moreover, SLC7A11 activity and its protein stability and cellular localization are also modulated upon stress. Patients' data show that SLC7A11 is overexpressed in various types of human cancers, and higher levels of SLC7A11 predict poorer overall survival. Growing evidence also suggests that targeting SLC7A11 is a promising approach in cancer therapy by effectively inhibiting tumor proliferation, invasion, and metastasis, as well as counteracting cancer stem cells and overcoming chemoresistance. This review highlights the regulation of SLC7A11 as an unconventional checkpoint in tumorigenesis through modulating ferroptotic responses under various types of stress.
Collapse
Affiliation(s)
- Zhenyi Su
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yanqing Liu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lin Wang
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
19
|
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
|
20
|
Xie F, Niu Y, Chen X, Kong X, Yan G, Zhuang A, Li X, Lian L, Qin D, Zhang Q, Zhang R, Yang K, Xia X, Chen K, Xiao M, Yang C, Wu T, Shen Y, Yu C, Luo C, Lin SH, Li W. Ursodeoxycholic acid inhibits the uptake of cystine through SLC7A11 and impairs de novo synthesis of glutathione. J Pharm Anal 2025; 15:101068. [PMID: 39902457 PMCID: PMC11788867 DOI: 10.1016/j.jpha.2024.101068] [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: 03/19/2024] [Revised: 07/18/2024] [Accepted: 08/03/2024] [Indexed: 02/03/2025] Open
Abstract
Ursodeoxycholic acid (UDCA) is a naturally occurring, low-toxicity, and hydrophilic bile acid (BA) in the human body that is converted by intestinal flora using primary BA. Solute carrier family 7 member 11 (SLC7A11) functions to uptake extracellular cystine in exchange for glutamate, and is highly expressed in a variety of human cancers. Retroperitoneal liposarcoma (RLPS) refers to liposarcoma originating from the retroperitoneal area. Lipidomics analysis revealed that UDCA was one of the most significantly downregulated metabolites in sera of RLPS patients compared with healthy subjects. The augmentation of UDCA concentration (≥25 μg/mL) demonstrated a suppressive effect on the proliferation of liposarcoma cells. [15N2]-cystine and [13C5]-glutamine isotope tracing revealed that UDCA impairs cystine uptake and glutathione (GSH) synthesis. Mechanistically, UDCA binds to the cystine transporter SLC7A11 to inhibit cystine uptake and impair GSH de novo synthesis, leading to reactive oxygen species (ROS) accumulation and mitochondrial oxidative damage. Furthermore, UDCA can promote the anti-cancer effects of ferroptosis inducers (Erastin, RSL3), the murine double minute 2 (MDM2) inhibitors (Nutlin 3a, RG7112), cyclin dependent kinase 4 (CDK4) inhibitor (Abemaciclib), and glutaminase inhibitor (CB839). Together, UDCA functions as a cystine exchange factor that binds to SLC7A11 for antitumor activity, and SLC7A11 is not only a new transporter for BA but also a clinically applicable target for UDCA. More importantly, in combination with other antitumor chemotherapy or physiotherapy treatments, UDCA may provide effective and promising treatment strategies for RLPS or other types of tumors in a ROS-dependent manner.
Collapse
Affiliation(s)
- Fu'an Xie
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yujia Niu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiaobing Chen
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Beijing, 102206, China
| | - Xu Kong
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Guangting Yan
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Aobo Zhuang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xi Li
- School of Public Health, Harvard University, Boston, MA, 02115, USA
| | - Lanlan Lian
- Department of Laboratory Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361102, China
| | - Dongmei Qin
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Quan Zhang
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ruyi Zhang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Kunrong Yang
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Xiaogang Xia
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Kun Chen
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Mengmeng Xiao
- Department of General Surgery, Peking University People's Hospital, Beijing, 100032, China
| | - Chunkang Yang
- Department of Gastrointestinal Surgical Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Ting Wu
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Beijing, 102206, China
| | - Ye Shen
- Department of Management, Jiang Xia Blood Technology Co., Ltd., Shanghai, 200000, China
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chenghua Luo
- Department of General Surgery, Peking University People's Hospital, Beijing, 100032, China
| | - Shu-Hai Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wengang Li
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| |
Collapse
|
21
|
Wang J, Guo C, Wang J, Zhang X, Qi J, Huang X, Hu Z, Wang H, Hong B. Tumor Mutation Signature Reveals the Risk Factors of Lung Adenocarcinoma with EGFR or KRAS Mutation. Cancer Control 2025; 32:10732748241307363. [PMID: 39760242 DOI: 10.1177/10732748241307363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
INTRODUCTION EGFR and KRAS mutations are frequently detected in lung adenocarcinoma (LUAD). Tumor mutational signature (TMS) determination is an approach to identify somatic mutational patterns associated with pathogenic factors. In this study, through the analysis of TMS, the underlying pathogenic factors of LUAD with EGFR and KRAS mutations were traced. METHODS This was a retrospective study. TMS of LUAD with KRAS and EGFR mutations from the TCGA, OncoSG, and MSK datasets was determined by two bioinformatics tools, namely the "MutationalPatterns" and "FitMS" packages. Elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) of LUAD clinical specimens was analyzed using capillary electrophoresis. RESULTS In LUAD with KRAS mutations, TMS analysis indicated that the smoking-related SBS4 signature was enriched. For LUAD with EGFR L858R mutation, the smoking-related SBS4 signature was enriched in the Western population from the TCGA database; however, the smoking-related SBS4 signature was not obvious in Asian LUAD patients. LUAD with EGFR exon19 deletion (19Del) exhibited stronger SBS15 signature, which was related to defective DNA mismatch repair. Capillary electrophoresis analysis showed that an EMAST locus was frequently instable in LUAD with EGFR 19Del. Different from the Western population, Asian LUAD patients with EGFR mutations exhibited the enrichment of SBS1, SBS2, and SBS13 signatures, which were associated with the endogenous mutation process of cytidine deamination. CONCLUSIONS TMS analysis reveals that smoking is associated with LUAD with KRAS mutations. Defective DNA mismatch repair and endogenous cytidine deamination are associated with LUAD with EGFR mutations, especially for the EGFR 19Del. The endogenous mutational process is stronger in Asian LUAD patients than Western LUAD patients.
Collapse
Affiliation(s)
- Jialiang Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Chang Guo
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Jiexiao Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Xiaopeng Zhang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Jian Qi
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Xiang Huang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Zongtao Hu
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Hongzhi Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| | - Bo Hong
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, China
| |
Collapse
|
22
|
Azizi R, Ahmed HH, Abdul Kareem R, Taher Waam WM, Alwan M, Jawad MJ, Hamad AK, Darzi S. SLC7A11 Inhibitors Represent a Promising Therapeutic Target by Facilitating the Induction of Ferroptosis in Breast Cancer. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2025; 14:496-516. [PMID: 40123584 PMCID: PMC11927159 DOI: 10.22088/ijmcm.bums.14.1.496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 10/23/2024] [Indexed: 03/25/2025]
Abstract
It is predicted with near certainty that an estimated 310,720 women will be diagnosed with invasive breast carcinoma in 2024, while the number of men will be significantly lower at around 2,800, highlighting the alarming prevalence of this cancer across the sexes. The Solute Carrier Family 7 Member 11(SLC7A11) gene is vital for the exchange of extracellular cystine for glutamate at a 1:1 ratio and its expression is significantly increased in various tumors. Numerous research studies have shown that SLC7A11 expression is fine-tuned at several levels, contributing to its pharmacological functions in tumors, such as maintaining cellular redox balance, promoting cell proliferation, and influencing ferroptosis. Many studies suggest that reducing SLC7A11 expression and activity may be beneficial for cancer treatment, making it a promising target for therapy. However, recent findings also suggest that inhibiting SLC7A11 in certain scenarios may increase the survival of cancer cells and promote drug resistance. This review begins with a brief overview of the properties of SLC7A11, including its structural features and physiological functions, followed by a summary of its potential regulators. We then delve deeper into its role in cancer, particularly breast cancer, and explore the relationships between SLC7A11 and ferroptosis, proliferation, metastasis, and therapeutic resistance. Consequently, more customized therapeutic approaches should be considered when targeting SLC7A11 in the context of breast cancer. Thus, high expression of SLC7A11 is associated with poor prognosis in breast cancer, and various inhibitors have been identified that can effectively target this transporter. Innovative therapeutic strategies, including immunotherapies targeting SLC7A11, can potentially reduce tumor growth and metastasis in breast cancer models.
Collapse
Affiliation(s)
- Rojin Azizi
- Master in Molecular and Cellular Biology in Department of Science at Islamic Azad University, Yadegar Emam Branch Tehran, Iran.
| | | | | | | | | | | | | | - Satinik Darzi
- Abnormal Uterine Bleeding Research Center, Semnan University of Medical Sciences Semnan, Iran.
| |
Collapse
|
23
|
Nath P, Alfarsi LH, El-Ansari R, Masisi BK, Erkan B, Fakroun A, Ellis IO, Rakha EA, Green AR. The amino acid transporter SLC7A11 expression in breast cancer. Cancer Biol Ther 2024; 25:2291855. [PMID: 38073087 PMCID: PMC10761065 DOI: 10.1080/15384047.2023.2291855] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer (BC), characterized by its diverse molecular profiles and clinical outcomes, presents a significant challenge in the development of effective therapeutic strategies. Metabolic reprogramming, a defining characteristic of cancer, has emerged as a promising target for novel therapies. SLC7A11, an amino acid transporter that facilitates cysteine uptake in exchange for glutamate, plays a crucial role in sustaining the altered metabolism of cancer cells. This study delves into the comprehensive analysis of SLC7A11 at the genomic, transcriptomic, and protein levels in extensive BC datasets to elucidate its potential role in different BC subtypes. SLC7A11 gene copy number and mRNA expression were evaluated using the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) cohort (n = 1,980) and Breast Cancer Gene Expression Miner (n = 4,712). SLC7A11 protein was assessed using immunohistochemistry in a large BC cohort (n = 1,981). Additionally, The Cancer Genome Atlas (TCGA) dataset was used to explore SLC7A11 DNA methylation patterns using MethSurv (n = 782) and association of SLC7A11 mRNA expression with immune infiltrates using TIMER (n = 1,100). High SLC7A11 mRNA and SLC7A11 protein expression were significantly associated with high tumor grade (p ≤ .02), indicating a potential role in cancer progression. Interestingly, SLC7A11 copy number gain was observed in HER2+ tumors (p = .01), suggesting a subtype-specific association. In contrast, SLC7A11 mRNA expression was higher in the basal-like/triple-negative (TN; p < .001) and luminal B tumors (p = .02), highlighting its differential expression across BC subtypes. Notably, high SLC7A11 protein expression was predominantly observed in Estrogen Receptor (ER)-negative and Triple Negative (TN) BC, suggesting a role in these aggressive subtypes. Further analysis revealed that SLC7A11 was positively correlated with other amino acid transporters and enzymes associated with glutamine metabolism, implying a coordinated role in metabolic regulation. Additionally, SLC7A11 gene expression was positively associated with neutrophil and macrophage infiltration, suggesting a potential link between SLC7A11 and tumor immunity. Our findings suggest that SLC7A11 plays a significant role in BC metabolism, demonstrating differential expression across subtypes and associations with poor patient outcomes. Further functional studies are warranted to elucidate the precise mechanisms by which SLC7A11 contributes to BC progression and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Preyanka Nath
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Lutfi H. Alfarsi
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Rokaya El-Ansari
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Brendah K. Masisi
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Busra Erkan
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Ali Fakroun
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Ian O. Ellis
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A. Rakha
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
- Cellular Pathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R. Green
- Nottingham Breast Cancer Research Centre, Academic Unit of Translational Medical Sciences, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, UK
| |
Collapse
|
24
|
Wu R, Zhu H, He Q, Yuan T, Yang B. Metabolic reprogramming in KRAS-mutant cancers: Proven targetable vulnerabilities and potential therapeutic strategies. Drug Discov Today 2024; 29:104220. [PMID: 39481592 DOI: 10.1016/j.drudis.2024.104220] [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: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Kras (Ki-ras2 Kirsten rat sarcoma viral oncogene homolog), one of the most frequently mutated oncogenes in the human genome, is considered 'untargetable'. Although specific KRASG12C inhibitors have been developed, their overall impact is limited, highlighting the need for further research on targeting KRAS-mutant cancers. Metabolic abnormalities are key hallmarks of cancer, with KRAS-driven tumors exhibiting traits like glycolysis upregulation, glutamine addiction, lipid droplet accumulation, highly active macropinocytosis, and metabolic reprogramming-associated tumor microenvironment remodeling. Targeting these unique metabolic characteristics offers a promising strategy for new cancer treatments. This review summarizes recent advances in our understanding of the metabolic network in KRAS-mutated tumor cells, discusses potential targetable vulnerabilities, and outlines clinical developments in relevant therapies, while also addressing challenges to improve strategies against these aggressive cancers.
Collapse
Affiliation(s)
- Ruilin Wu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China.
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China; School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
25
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
26
|
Lee J, Roh JL. Unveiling therapeutic avenues targeting xCT in head and neck cancer. Cell Oncol (Dordr) 2024; 47:2019-2030. [PMID: 39361147 DOI: 10.1007/s13402-024-00997-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2024] [Indexed: 01/11/2025] Open
Abstract
Head and neck cancer (HNC) remains a major global health burden, prompting the need for innovative therapeutic strategies. This review examines the role of the cystine/glutamate antiporter (xCT) in HNC, specifically focusing on how xCT contributes to cancer progression through mechanisms such as redox imbalance, ferroptosis, and treatment resistance. The central questions addressed include how xCT dysregulation affects tumor biology and the potential for targeting xCT to enhance treatment outcomes. We explore recent developments in xCT-targeted current and emerging therapies, including xCT inhibitors and novel treatment modalities, and their role in addressing therapeutic challenges. This review aims to provide a comprehensive analysis of xCT as a therapeutic target and to outline future directions for research and clinical application.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, 13496, Gyeonggi-do, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, 13496, Gyeonggi-do, Republic of Korea.
- Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea.
| |
Collapse
|
27
|
Zhang Y, Sun C, Wang B, Gu A, Zhou Z, Gu C. Brain-Derived Exosomal miR-9-5p Induces Ferroptosis in Traumatic Brain Injury-Induced Acute Lung Injury by Targeting Scd1. CNS Neurosci Ther 2024; 30:e70189. [PMID: 39723576 DOI: 10.1111/cns.70189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/11/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024] Open
Abstract
AIMS This study aimed to explore the role and underlying mechanisms of brain-derived exosomes in traumatic brain injury-induced acute lung injury (TBI-induced ALI), with a particular focus on the potential regulation of ferroptosis through miRNAs and Scd1. METHODS To elucidate TBI-induced ALI, we used a TBI mouse model. Exosomes were isolated from the brains of these mice and characterized using TEM and NTA. LC-MS analysis revealed an increase in the level of ferroptosis in the lung tissues of mice with TBI. Subsequent miRNA and mRNA sequencing revealed the upregulation of miR-9-5p and the downregulation of Scd1 in the pulmonary tissues of these mice. Ferroptosis was assessed by quantifying the levels of ROS, MDA, and Fe2+ and the expression of proteins associated with ferroptosis. RESULTS TBI led to the release of exosomes enriched with miR-9-5p, which targeted Scd1 in lung tissue, thereby promoting ferroptosis. Treatment with antagomir 9-5p reduced the level of ALI in TBI mice, indicating that exosomal miR-9-5p plays a significant role in TBI-induced ALI. CONCLUSION This study revealed that brain-derived exosomal miR-9-5p mediates ferroptosis in TBI-induced ALI by targeting Scd1. These findings may provide new insights into the complex interplay between TBI and ALI and highlight the potential of miR-9-5p as a target for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Chang Sun
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bailun Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Angran Gu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ziyi Zhou
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Changping Gu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
28
|
Du L, Xu G, Zhang X, Zhang Z, Yang Y, Teng H, Yang T. AQP4-AS1 Can Regulate the Expression of Ferroptosis-Related Regulator ALOX15 through Competitive Binding with miR-4476 in Lung Adenocarcinoma. Glob Med Genet 2024; 11:241-250. [PMID: 39155888 PMCID: PMC11329318 DOI: 10.1055/s-0044-1789199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Abstract
Background The AQP4-AS1/miR-4476-ALOX15 regulatory axis was discovered in previous studies. We aimed to investigate the regulatory mechanism of the ferroptosis-related regulator ALOX15 by AQP4-AS1 and miR-4476 in lung adenocarcinoma (LUAD) and find new targets for clinical treatment. Methods After bioinformatics analysis, we contained one ferroptosis-related gene (FRG), namely ALOX15. MicroRNAs (miRNAs) and long noncoding RNAs were predicted by miRWalk. Furthermore, we constructed overexpressed LUAD cell lines. Real-time quantitative polymerase chain reaction and western blot were used to determine the expression of mRNA and protein, respectively. Cell Counting Kit-8 (CCK-8) and EdU assay were used to detect the cell proliferation. Double luciferase assay was used to detect the binding relationship between AQP4-AS1 and miR-4464. Results ALOX15 was the most significantly downregulated FRG compared with normal tissues. Furthermore, protein-protein interaction network analysis indicated that the AQP4-AS1-miR-4476-ALOX15 regulatory axis might be involved in the occurrence and development of LUAD and there might be direct interaction between AQP4-AS1 and miR-4476, and miR-4476 and ALOX15. Furthermore, AQP4-AS1 and ALOX15 were significantly downregulated in the LUAD tissue and cell lines, whereas miR-4476 showed the opposite results ( p < 0.001). AQP4-AS1 overexpression improved the ALOX15 expression in LUAD cell lines. CCK-8 and EdU assay revealed that overexpression of AQP4-AS1 and ALOX15 inhibited the LUAD cell proliferation. Double luciferase assay results indicated that there was a combination between AQP4-AS1 and miRNA-4476. In addition, we found that overexpressed AQP4-AS1 activates the ferroptosis in LUAD cell lines. Conclusions AQP4-AS1 can regulate the expression of ALOX15 through competitive binding with miR-4476, further activate ferroptosis and inhibit the proliferation of LUAD cells.
Collapse
Affiliation(s)
- Lin Du
- Department of Thoracic Surgery, Tianjin Fifth Center Hospital, Tianjin, China
| | - Geng Xu
- Department of Thoracic Surgery, Tianjin Fifth Center Hospital, Tianjin, China
| | - Xiuqiang Zhang
- Department of Thoracic Surgery, Tianjin Fifth Center Hospital, Tianjin, China
| | - Zhiwei Zhang
- Department of Thoracic Surgery, Tianjin Fifth Center Hospital, Tianjin, China
| | - Yang Yang
- Department of Thoracic Surgery, Tianjin Fifth Center Hospital, Tianjin, China
| | - Hongsheng Teng
- Department of Thoracic Surgery, Tianjin Fifth Center Hospital, Tianjin, China
| | - Tao Yang
- Department of Thoracic Surgery, Tianjin Fifth Center Hospital, Tianjin, China
| |
Collapse
|
29
|
Kędzia K, Szmajda-Krygier D, Krygier A, Jabłoński S, Balcerczak E, Wcisło S. Altered carnitine transporter genes ( SLC22A5, SLC22A16, SLC6A14) expression pattern among lung cancer patients. Transl Lung Cancer Res 2024; 13:2903-2917. [PMID: 39670016 PMCID: PMC11632432 DOI: 10.21037/tlcr-24-448] [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: 05/22/2024] [Accepted: 08/20/2024] [Indexed: 12/14/2024]
Abstract
Background Despite the decrease of morbidity rate of non-small cell lung cancer (NSCLC) in recent years, it is still a cancer with poor prognosis. Lung cancers (LCs) are usually diagnosed at a late stage of the disease due to non-specific clinical symptoms. Proper regulation of carnitine levels is important in the context of development and increased risk of cancer cells proliferation. The expression profiles and clinical value of SLC family members in LC remain largely unexplored. The aim of the study was the assessment of SLC22A16, SLC22A5 and SLC6A14 mRNA expression level among patients suffering from NSCLC. The obtained results were compared with the clinical and the pathological features of NSCLC patients. Methods Through reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and bioinformatics studies, the evaluation of carnitine transporting genes (SLC22A16, SLC22A5 and SLC6A14) mRNA levels was performed in order to elucidate their connection to clinical features of patients and influence on overall survival (OS). Results The analysis showed a significant difference for the SLC22A5 gene of NSCLC patients and for SLC6A14 and SLC22A5 genes in LUSC patients in terms of sex (P=0.002, P=0.02 and P=0.001, respectively) and in terms of tobacco smoking (P=0.04). Analysis also revealed a significant negative correlation for SLC22A5 and SLC22A16 genes expression level in the lung adenocarcinoma (LUAD) subtype with standardized uptake value (SUV) (r=-0.40, P=0.02 and r=-0.43, P=0.04). The significant downregulation of gene expression compared to normal adjacent tissue was observed for SLC22A5 in lung squamous cell carcinoma (LUSC) and for SLC6A14 in both LUAD and LUSC subtypes. The effect of the SLC22A5, SLC22A16 and SLC6A14 gene expression at the time of diagnosis on the OS time of LC patients revealed that lower expression correlated with a shorter 5 years OS (all P values <0.01). The effects were distinct after division for LUAD and LUSC subtypes. Conclusions The expression levels of genes encoding carnitine transporters are diverse, hinting at a potentially altered carnitine metabolism in LC patients. Notably, this variance is not uniform and exhibits specificity across LC subtypes, with marked distinctions between LUAD and LUSC. The correlation between gene expression levels and OS of patients underlines the prognostic significance of SLC genes within these cancer subtypes.
Collapse
Affiliation(s)
- Konrad Kędzia
- Department of Thoracic, General and Oncological Surgery, Medical University of Lodz and Military Medical Academy Memorial Teaching Hospital of the Medical University of Lodz-Central Veteran Hospital, Lodz, Poland
| | - Dagmara Szmajda-Krygier
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Lodz, Poland
| | - Adrian Krygier
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Lodz, Poland
| | - Sławomir Jabłoński
- Department of Thoracic, General and Oncological Surgery, Medical University of Lodz and Military Medical Academy Memorial Teaching Hospital of the Medical University of Lodz-Central Veteran Hospital, Lodz, Poland
| | - Ewa Balcerczak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Lodz, Poland
| | - Szymon Wcisło
- Department of Thoracic, General and Oncological Surgery, Medical University of Lodz and Military Medical Academy Memorial Teaching Hospital of the Medical University of Lodz-Central Veteran Hospital, Lodz, Poland
| |
Collapse
|
30
|
Zhang Y, Cao S, Zeng F, Pan D, Cai L, Zhou Y, Wang H, Qin G, Zhang C, Chen W. Dihydroartemisinin enhances the radiosensitivity of breast cancer by targeting ferroptosis signaling pathway through hsa_circ_0001610. Eur J Pharmacol 2024; 983:176943. [PMID: 39182549 DOI: 10.1016/j.ejphar.2024.176943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVE This study aimed to elucidate how DHA enhances the radiosensitivity of BC and to explain its potential mechanisms of action. METHODS The circular structure of hsa_circ_0001610 was confirmed by Sanger sequencing, RNase R treatment, RT-PCR analysis using gDNA or cDNA. Cellular localization of hsa_circ_0001610 and microRNA-139-5p (miR-139-5p) was detected by fluorescence in situ hybridization. Cell counting kit-8 assay, wound healing and colony formation tests for assessing cell proliferation, while flow cytometry was utilized to estimate cell cycle progression and apoptosis. Reactive oxygen species and malondialdehyde experiments were conducted to validate ferroptosis of BC cells. The expression of ncRNAs and mRNAs was quantified via qRT-PCR, and protein expression was analyzed using Western blot. The effects of hsa_circ_0001610 and DHA on radiosensitivity of BC in vivo were studied by establishing BC mice model. RESULTS In vivo and in vitro experimental results indicate that DHA promotes ferroptosis of BC cells at least partly by inhibiting hsa_circ_0001610/miR-139-5p/SLC7A11 pathway, thereby enhancing the radiosensitivity of BC cells. CONCLUSIONS Our findings showed that DHA can induce ferroptosis of BC cells by down-regulation of hsa_circ_0001610, thus enhancing radiosensitivity, suggesting a promising therapeutic strategy for enhancing BC radiosensitivity that is worthy of further exploration.
Collapse
Affiliation(s)
- YiWen Zhang
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - ShuYi Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, 510000, China
| | - FengXia Zeng
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - DeRun Pan
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - LongMei Cai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - YingYing Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - HongMei Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - GengGeng Qin
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China.
| | - Chao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, 510000, China.
| | - WeiGuo Chen
- Department of Radiology, NanFang Hospital, Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
31
|
Huang P, Zhao H, Dai H, Li J, Pan X, Pan W, Xia C, Liu F. FXR deficiency induced ferroptosis via modulation of the CBP-dependent p53 acetylation to suppress breast cancer growth and metastasis. Cell Death Dis 2024; 15:826. [PMID: 39543094 PMCID: PMC11564727 DOI: 10.1038/s41419-024-07222-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Farnesoid X receptor (NR1H4/FXR) functions as a scavenger of lipid peroxide products and drives the proliferation and metastasis of various cancers. However, the underlying molecular mechanisms remain poorly understood. In our study, we found that the expression levels of FXR, vimentin and SLC7A11 were significantly higher in breast cancer tissues, particularly in metastatic cancer tissues compared to non-metastatic ones. Furthermore, the increased FXR expression was positively correlated with vimentin and SLC7A11 in clinical tumor specimens. In addition, a high level of FXR correlated with poor prognosis in patients with breast cancer. Both Z-Guggulsterone (Z-GS), as a pharmacological inhibitor of FXR, and silencing FXR curbed proliferation and migration of breast cancer cells by promoting ferroptosis. Notably, our results showed that FXR competitively bound to CREB-binding protein (CBP) to suppress the interaction between p53 and CBP in the nucleus, and thus prevented p53 acetylation at lys382, which was essential for upregulating the expression of SLC7A11. Conversely, FXR knockdown increased the interaction between p53 and CBP and promoted p53 acetylation, which ultimately led to facilitating ferroptosis in breast cancer cells. More importantly, we also found that Z-GS inhibited TGF-β1-induced tumor growth and metastasis of breast cancer primarily through ferroptosis via regulating CBP-dependent p53 acetylation in nude mice. In conclusion, the FXR was first reported as a tumor promoter that enhanced the proliferation and metastasis of breast cancer cells through regulating CBP-dependent p53 K382 acetylation. It proposes that FXR may serve as a potential therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Ping Huang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Han Zhao
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Hua Dai
- Department of Pathology, the First Affiliated Hospital of Nanchang University, Nanchang, 330038, P. R. China
| | - Jinying Li
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Xiafang Pan
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Wentian Pan
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China
| | - Chunhua Xia
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
- Jiangxi Province Key Laboratory of New Drug Evaluation and Transformation, Nanchang, 330031, P. R. China.
| | - Fanglan Liu
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, P. R. China.
- Jiangxi Province Key Laboratory of New Drug Evaluation and Transformation, Nanchang, 330031, P. R. China.
| |
Collapse
|
32
|
Bian Y, Shan G, Bi G, Liang J, Hu Z, Sui Q, Shi H, Zheng Z, Yao G, Wang Q, Fan H, Zhan C. Targeting ALDH1A1 to enhance the efficacy of KRAS-targeted therapy through ferroptosis. Redox Biol 2024; 77:103361. [PMID: 39317105 PMCID: PMC11465744 DOI: 10.1016/j.redox.2024.103361] [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: 08/29/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024] Open
Abstract
KRAS is among the most commonly mutated oncogenes in human malignancies. Although the advent of sotorasib and adagrasib, has lifted the "undruggable" stigma of KRAS, the resistance to KRAS inhibitors quickly becomes a major issue. Here, we reported that aldehyde dehydrogenase 1 family member A1 (ALDH1A1), an enzyme in retinoic acid biosynthesis and redox balance, increases in response to KRAS inhibitors and confers resistance in a range of cancer types. KRAS inhibitors' efficacy is significantly improved in sensitive or drug-resistant cells, patient-derived organoids (PDO), and xenograft models by ALDH1A1 knockout, loss of enzyme function, or inhibitor. Furthermore, we discovered that ALDH1A1 suppresses the efficacy of KRAS inhibitors by counteracting ferroptosis. ALDH1A1 detoxicates deleterious aldehydes, boosts the synthesis of NADH and retinoic acid (RA), and improves RARA function. ALDH1A1 also activates the CREB1/GPX4 pathway, stimulates the production of lipid droplets in a pH-dependent manner, and subsequently prevents ferroptosis induced by KRAS inhibitors. Meanwhile, we established that GTF2I is dephosphorylated at S784 via ERK by KRAS inhibitors, which hinders its nuclear translocation and mediates ALDH1A1's upregulation in response to KRAS inhibitors. In summary, the results offer valuable insights into targeting ALDH1A1 to enhance the effectiveness of KRAS-targeted therapy through ferroptosis in cancer treatment.
Collapse
Affiliation(s)
- Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qihai Sui
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haochun Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaolin Zheng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangyu Yao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Thoracic Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
33
|
Wu A, Yang H, Xiao T, Gu W, Li H, Chen P. COPZ1 regulates ferroptosis through NCOA4-mediated ferritinophagy in lung adenocarcinoma. Biochim Biophys Acta Gen Subj 2024; 1868:130706. [PMID: 39181476 DOI: 10.1016/j.bbagen.2024.130706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Ferroptosis, a type of autophagy-dependent cell death, has been implicated in the pathogenesis of lung adenocarcinoma (LUAD). This study aimed to investigate the involvement of coatomer protein complex I subunit zeta 1 (COPZ1) in ferroptosis and ferritinophagy in LUAD. METHODS Publicly available human LUAD sample data were obtained from the TCGA database to analyze the association of COPZ1 expression with LUAD grade and patient survival. Clinical samples of LUAD and para-carcinoma tissues were collected. COPZ1-deficient LUAD cell model and xenograft model were established. These models were analyzed to evaluate tumor growth, lipid peroxidation levels, mitochondrial structure, autophagy activation, and iron metabolism. RESULTS High expression of COPZ1 was indicative of malignancy and poor overall survival. Clinical LUAD tissues showed increased COPZ1 expression and decreased nuclear receptor coactivator 4 (NCOA4) expression. COPZ1 knockdown inhibited xenograft tumor growth and induced apoptosis. COPZ1 knockdown elevated the levels of ROS, Fe2+ and lipid peroxidation. COPZ1 knockdown also caused mitochondrial shrinkage. Liproxstatin-1, deferoxamine, and z-VAD-FMK reversed the effects of COPZ1 knockdown on LUAD cell proliferation and ferroptosis. Furthermore, COPZ1 was directly bound to NCOA4. COPZ1 knockdown restricted FTH1 expression and promoted NCOA4 and LC3 expression. NCOA4 knockdown reversed the regulation of iron metabolism, lipid peroxidation, and mitochondrial structure induced by COPZ1 knockdown. COPZ1 knockdown induced the translocation of ferritin to lysosomes for degradation, whereas NCOA4 knockdown disrupted this process. CONCLUSION This study provides novel evidence that COPZ1 regulates NCOA4-mediated ferritinophagy and ferroptosis. These findings provide new insights into the pathogenesis and potential treatment of LUAD.
Collapse
Affiliation(s)
- Anbang Wu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hongmin Yang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Tengfei Xiao
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wangnin Gu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - He Li
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; College of pharmacy, Changsha Medical University, Changsha 410219, China.
| | - Pan Chen
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
| |
Collapse
|
34
|
Nissi L, Tuominen S, Routila J, Huusko T, Ketonen P, Sundvall M, Leivo I, Irjala H, Minn H, Grönroos T, Ventelä S. xCT as a Predictor for Survival in a Population-Based Cohort of Head and Neck Squamous Cell Carcinoma. Cancer Med 2024; 13:e70371. [PMID: 39487701 PMCID: PMC11530867 DOI: 10.1002/cam4.70371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND xCT, also known as SLC7A11 (solute carrier Family 7 Member 11), is a cystine/glutamate antiporter protein that mediates regulated cell death and antioxidant defense. The aim of this study was to investigate the effect of xCT on the outcome of patients diagnosed with new head and neck squamous cell carcinoma (HNSCC). METHODS This retrospective cohort study utilized a population-based dataset, comprising all patients (n = 1033) diagnosed with new HNSCC during 2005-2015 in a population of 697,000 people. All patients (n = 585) with a tumor tissue sample available for immunohistochemical (IHC) staining were included. The follow-up rates were 97% and 81% at 3 and 5 years, respectively. Also, the specificity of the anti-xCT antibody was validated. RESULTS The expression level and prognostic significance of xCT were strongly dependent on tumor location. In oropharyngeal squamous cell carcinoma (OPSCC) patients, xCT expression was a significant prognostic factor for 5-year overall survival (OAS) (HR: 2.71; 95% CI 1.67-4.39; p < 0.001), disease-specific survival (DSS) (HR: 2.58; 95% CI 1.47-4.54; p = 0.001), and disease-free survival (DFS) (HR: 2.69; 95% CI 1.55-4.64; p < 0.001). Five-year survival rates for OPSCC patients with high and low levels of xCT were OAS 34% versus 62%; DSS 51% versus 73%; DFS 43% versus 73%, respectively. According to a multivariate model adjusted for age, T-class, nodal positivity, and tobacco consumption, xCT was an independent prognostic factor for 3-year survival, in which it outperformed p16 IHC. Similar associations were not observed in squamous cell carcinomas of oral cavity or larynx. Regarding treatment modalities, xCT was most predictive in HNSCC patients who received radiotherapy. CONCLUSIONS High xCT expression was associated with poor prognosis in OPSCC. Our findings suggest that joint analysis of xCT and p16 may add significant value in OPSCC treatment stratification.
Collapse
Affiliation(s)
- Linda Nissi
- Department of Oncology, and FICAN West Cancer CentreTurku University Hospital and University of TurkuTurkuFinland
| | - Sanni Tuominen
- Preclinical Imaging Turku PET CentreUniversity of TurkuTurkuFinland
- Medicity Research LaboratoryUniversity of TurkuTurkuFinland
| | - Johannes Routila
- Department of Otorhinolaryngology—Head and Neck SurgeryTurku University Hospital and University of TurkuTurkuFinland
| | - Teemu Huusko
- Department of Otorhinolaryngology—Head and Neck SurgeryTurku University Hospital and University of TurkuTurkuFinland
| | - Petra Ketonen
- Preclinical Imaging Turku PET CentreUniversity of TurkuTurkuFinland
- Medicity Research LaboratoryUniversity of TurkuTurkuFinland
| | - Maria Sundvall
- Department of Oncology, and FICAN West Cancer CentreTurku University Hospital and University of TurkuTurkuFinland
- Cancer Research UnitInstitute of BiomedicineUniversity of TurkuTurkuFinland
| | - Ilmo Leivo
- Department of PathologyTurku University Hospital and University of TurkuTurkuFinland
| | - Heikki Irjala
- Department of Otorhinolaryngology—Head and Neck SurgeryTurku University Hospital and University of TurkuTurkuFinland
| | - Heikki Minn
- Department of Oncology, and FICAN West Cancer CentreTurku University Hospital and University of TurkuTurkuFinland
| | - Tove J. Grönroos
- Department of Oncology, and FICAN West Cancer CentreTurku University Hospital and University of TurkuTurkuFinland
- Preclinical Imaging Turku PET CentreUniversity of TurkuTurkuFinland
- Medicity Research LaboratoryUniversity of TurkuTurkuFinland
| | - Sami Ventelä
- Department of Otorhinolaryngology—Head and Neck SurgeryTurku University Hospital and University of TurkuTurkuFinland
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| |
Collapse
|
35
|
Xia B, Qiu L, Yue J, Si J, Zhang H. The metabolic crosstalk of cancer-associated fibroblasts and tumor cells: Recent advances and future perspectives. Biochim Biophys Acta Rev Cancer 2024; 1879:189190. [PMID: 39341468 DOI: 10.1016/j.bbcan.2024.189190] [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: 12/26/2023] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Tumor cells grow in a microenvironment with a lack of nutrients and oxygen. Cancer-associated fibroblasts (CAFs) as one major component of tumor microenvironment have strong ability to survive under stressful conditions through metabolic remodelling. Furthermore, CAFs are educated by tumor cells and help them adapt to the hostile microenvironment through their metabolic communication. By inducing catabolism, CAFs release nutrients into the microenvironment which are taken up by tumor cells to satisfy their metabolic requirements. Furthermore, CAFs can recycle toxic metabolic wastes produced by cancer cells into energetic substances, allowing cancer cells to undergo biosynthesis. Their metabolic crosstalk also enhances CAFs' pro-tumor phenotype and reshape the microenvironment facilitating tumor cells' metastasis and immune escape. In this review, we have analyzed the effect and mechanisms of metabolic crosstalk between tumor cells and CAFs. We also analyzed the future perspectives in this area from the points of CAFs heterogeneity, spatial metabonomics and patient-derived tumor organoids (PDOs). These information may deepen the knowledge of tumor metabolism regulated by CAFs and provide novel insights into the development of metabolism-based anti-cancer strategies.
Collapse
Affiliation(s)
- Bing Xia
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou Cancer Hospital, Hangzhou 310002, China
| | - Liqing Qiu
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, 310002, China
| | - Jing Yue
- Hangzhou Cancer Institution, Hangzhou Cancer Hospital, 310002, China
| | - Jingxing Si
- Cancer Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Hongfang Zhang
- Hangzhou Cancer Institution, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou Cancer Hospital, 310002, China.
| |
Collapse
|
36
|
Xiao Z, Li S, Wu X, Chen X, Yan D, He J. GATA-4 overexpressing BMSC-derived exosomes suppress H/R-induced cardiomyocyte ferroptosis. iScience 2024; 27:110784. [PMID: 39391723 PMCID: PMC11466636 DOI: 10.1016/j.isci.2024.110784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/01/2024] [Accepted: 08/19/2024] [Indexed: 10/12/2024] Open
Abstract
Bone marrow mesenchymal stem cell (BMSC)-derived exosomes overexpressing GATA-4 (Exosoe-GATA-4) can protect cardiac function. Mitochondrial permeability transition pore (mPTP) has a crucial role in ferroptosis. This study aimed to assess the mechanism of Exosoe-GATA-4 in myocardial ischemia/reperfusion (I/R) injury. Exos were successfully excreted, and 185 differential expression miRNAs were obtained using bioinformatics. The Exosoe-GATA-4 effectively suppressed hypoxia/reoxygenation (H/R)-induced cardiomyocytes' ferroptosis, while the effects were reversed by miR-330-3p inhibitor. miR-330-3p targeted negative regulated BAP1. The effects of miR-330-3p inhibitor were reversed by knock-down BAP1. Also, BAP1 reversed the effects of Exosoe-GATA-4 on H/R-induced cardiomyocytes' ferroptosis by downregulating SLC7A11. Mechanistically, BAP1 interacted with IP3R and increased cardiomyocytes' Ca2+ level, causing mPTP opening and mitochondrial dysfunction, promoting H/R-induced cardiomyocytes' ferroptosis. Moreover, hydrogen sulfide (H2S) content was increased and regulated the keap1/Nrf2 signaling pathway by Exosoe-GATA-4 treated. Exosoe-GATA-4 effectively suppresses H/R-induced cardiomyocytes' ferroptosis by upregulating miR-330-3p, which regulates the BAP1/SLC7A11/IP3R axis and inhibits mPTP opening.
Collapse
Affiliation(s)
- Zhiyuan Xiao
- Department of Medical Intensive Care Unit, the First People′s Hospital of Yunnan Province, No.157 Jinbi Road, Kunming, Yunnan 650032, China
| | - Si Li
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, P.R. China
| | - Xinxin Wu
- Yunnan University of Traditional Chinese Medicine, No.1076 Yuhua Road, Kunming, Yunnan 650500, China
| | - Xinhao Chen
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650500, P.R. China
| | - Dan Yan
- Department of Medical Intensive Care Unit, the First People′s Hospital of Yunnan Province, No.157 Jinbi Road, Kunming, Yunnan 650032, China
| | - Jigang He
- Department of Cardiovascular Surgery, the First People′s Hospital of Yunnan Province, No.157 Jinbi Road, Kunming, Yunnan 650032, China
| |
Collapse
|
37
|
Zheng H, Chen H, Cai Y, Shen M, Li X, Han Y, Deng X, Cao H, Liu J, Li H, Liu B, Li G, Wang X, Chen H, Hou J, Lin SH, Zong L, Zhang Y. Hydrogen sulfide-mediated persulfidation regulates homocysteine metabolism and enhances ferroptosis in non-small cell lung cancer. Mol Cell 2024; 84:4016-4030.e6. [PMID: 39321805 DOI: 10.1016/j.molcel.2024.08.035] [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: 11/15/2023] [Revised: 06/15/2024] [Accepted: 08/30/2024] [Indexed: 09/27/2024]
Abstract
Hydrogen sulfide (H₂S), a metabolite of the transsulfuration pathway, has been implicated in ferroptosis, a unique form of cell death caused by lipid peroxidation. While the exact mechanisms controlling ferroptosis remain unclear, our study reveals that H₂S sensitizes human non-small cell lung cancer (NSCLC) cells to this process, particularly when cysteine levels are low. Combining H₂S with cystine depletion significantly enhances the effectiveness of ferroptosis-based cancer therapy. Mechanistically, H₂S persulfidates the 195th cysteine on S-adenosyl homocysteine hydrolase (SAHH), reducing its enzymatic activity. This leads to decreased homocysteine levels, subsequently lowering cysteine and glutathione concentrations under cystine depletion conditions. These changes ultimately increase the vulnerability of NSCLC cells to ferroptosis. Our findings establish H₂S as a key regulator of homocysteine metabolism and a critical factor in determining NSCLC cell susceptibility to ferroptosis. These results highlight the potential of H₂S-based therapies to improve the efficacy of ferroptosis-targeted cancer treatments for NSCLC.
Collapse
Affiliation(s)
- Hualei Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Huidi Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yunjie Cai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Min Shen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xilin Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yi Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xusheng Deng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hongjie Cao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Junjia Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hao Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Benchao Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Ganlin Li
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xindong Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hui Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Jingjing Hou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Shu-Hai Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute for Data Science in Health and Medicine Engineering, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, Fujian, China
| | - Lili Zong
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yongyou Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute for Data Science in Health and Medicine Engineering, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, Fujian, China.
| |
Collapse
|
38
|
Li T, Song Y, Wei L, Song X, Duan R. Disulfidptosis: a novel cell death modality induced by actin cytoskeleton collapse and a promising target for cancer therapeutics. Cell Commun Signal 2024; 22:491. [PMID: 39394612 PMCID: PMC11470700 DOI: 10.1186/s12964-024-01871-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024] Open
Abstract
Disulfidptosis is a novel discovered form of programmed cell death (PCD) that diverges from apoptosis, necroptosis, ferroptosis, and cuproptosis, stemming from disulfide stress-induced cytoskeletal collapse. In cancer cells exhibiting heightened expression of the solute carrier family 7 member 11 (SLC7A11), excessive cystine importation and reduction will deplete nicotinamide adenine dinucleotide phosphate (NADPH) under glucose deprivation, followed by an increase in intracellular disulfide stress and aberrant disulfide bond formation within actin networks, ultimately culminating in cytoskeletal collapse and disulfidptosis. Disulfidptosis involves crucial physiological processes in eukaryotic cells, such as cystine and glucose uptake, NADPH metabolism, and actin dynamics. The Rac1-WRC pathway-mediated actin polymerization is also implicated in this cell death due to its contribution to disulfide bond formation. However, the precise mechanisms underlying disulfidptosis and its role in tumors are not well understood. This is probably due to the multifaceted functionalities of SLC7A11 within cells and the complexities of the downstream pathways driving disulfidptosis. This review describes the critical roles of SLC7A11 in cells and summarizes recent research advancements in the potential pathways of disulfidptosis. Moreover, the less-studied aspects of this newly discovered cell death process are highlighted to stimulate further investigations in this field.
Collapse
Affiliation(s)
- Tianyi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Lijuan Wei
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Xiangyi Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Ruifeng Duan
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China.
| |
Collapse
|
39
|
Li W, Moretti L, Su X, Yeh CR, Torres MP, Barker TH. Strain-dependent glutathionylation of fibronectin fibers impacts mechano-chemical behavior and primes an integrin switch. Nat Commun 2024; 15:8751. [PMID: 39384749 PMCID: PMC11479631 DOI: 10.1038/s41467-024-52742-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/18/2024] [Indexed: 10/11/2024] Open
Abstract
The extracellular matrix (ECM) is a protein polymer network that physically supports cells within a tissue. It acts as an important physical and biochemical stimulus directing cell behaviors. For fibronectin (Fn), a predominant component of the ECM, these physical and biochemical activities are inextricably linked as physical forces trigger conformational changes that impact its biochemical activity. Here, we analyze whether oxidative post-translational modifications, specifically glutathionylation, alter Fn's mechano-chemical characteristics through stretch-dependent protein modification. ECM post-translational modifications represent a potential for time- or stimulus-dependent changes in ECM structure-function relationships that could persist over time with potentially significant impacts on cell and tissue behaviors. In this study, we show evidence that glutathionylation of Fn ECM fibers is stretch-dependent and alters Fn fiber mechanical properties with implications on the selectivity of engaging integrin receptors. These data demonstrate the existence of multimodal post-translational modification mechanisms within the ECM with high relevance to the microenvironmental regulation of downstream cell behaviors.
Collapse
Affiliation(s)
- Wei Li
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA, USA
| | - Leandro Moretti
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA, USA
| | - Xinya Su
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Chiuan-Ren Yeh
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA, USA
| | - Matthew P Torres
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, Schools of Engineering and Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
40
|
Xue L, Luo K, Hou K, Huo W, Ruan P, Xue Y, Yao X, Meng C, Xia D, Tang Y, Zhao W, Yuan H, Zhao L, Gao L, Yuan Q, Gao X, Cao K. Targeted Gold Nanoclusters for Synergistic High-Risk Neuroblastoma Therapy through Noncanonical Ferroptosis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53555-53566. [PMID: 39327976 DOI: 10.1021/acsami.4c11979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Children with extracranial high-risk neuroblastoma (NB) have a poor prognosis due to resistance against apoptosis. Recently, ferroptosis, another form of programmed cell death, has been tested in clinical trials for high-risk NB; however, drug resistance and side effects have also been observed. Here, we find that the gold element in gold nanoclusters can significantly affect iron metabolism and sensitize high-risk NB cells to ferroptosis. Accordingly, we developed a gold nanocluster conjugated with a modified NB-targeting peptide. This gold nanocluster, namely, NANT, shows excellent NB targeting efficiency and dramatically promotes ferroptosis. Surprisingly, this effect is exerted by elevating the noncanonical ferroptosis pathway, which is dependent on heme oxygenase-1-regulated Fe(II) accumulation. Furthermore, NANT dramatically inhibits the growth of high-risk NB in both tumor spheroid and xenograft models by promoting noncanonical ferroptosis evidenced by enhanced intratumoral Fe(II) and heme oxygenase-1. Importantly, this strategy shows excellent cardiosafety, offering a promising strategy to overcome ferroptosis resistance for the efficient and safe treatment of children with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Liyuan Xue
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Kaidi Luo
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Kaixiao Hou
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Wendi Huo
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Panpan Ruan
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Yilin Xue
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Xiuxiu Yao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Cong Meng
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Dongfang Xia
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Yuhua Tang
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Wencong Zhao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Hui Yuan
- Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Liang Zhao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Liang Gao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Qing Yuan
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Xueyun Gao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Kai Cao
- Department of Chemistry, College of Chemistry and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
41
|
Chen B, Fan P, Song X, Duan M. The role and possible mechanism of the ferroptosis-related SLC7A11/GSH/GPX4 pathway in myocardial ischemia-reperfusion injury. BMC Cardiovasc Disord 2024; 24:531. [PMID: 39354361 PMCID: PMC11445876 DOI: 10.1186/s12872-024-04220-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MI/RI) is an unavoidable risk event for acute myocardial infarction, with ferroptosis showing close involvement. We investigated the mechanism of MI/RI inducing myocardial injury by inhibiting the ferroptosis-related SLC7A11/glutathione (GSH)/glutathione peroxidase 4 (GPX4) pathway and activating mitophagy. METHODS A rat MI/RI model was established, with myocardial infarction area and injury assessed by TTC and H&E staining. Rat cardiomyocytes H9C2 were cultured in vitro, followed by hypoxia/reoxygenation (H/R) modeling and the ferroptosis inhibitor lipoxstatin-1 (Lip-1) treatment, or 3-Methyladenine or rapamycin treatment and overexpression plasmid (oe-SLC7A11) transfection during modeling. Cell viability and death were evaluated by CCK-8 and LDH assays. Mitochondrial morphology was observed by transmission electron microscopy. Mitochondrial membrane potential was detected by fluorescence dye JC-1. Levels of inflammatory factors, reactive oxygen species (ROS), Fe2+, malondialdehyde, lipid peroxidation, GPX4 enzyme activity, glutathione reductase, GSH and glutathione disulfide, and SLC7A11, GPX4, LC3II/I and p62 proteins were determined by ELISA kit, related indicator detection kits and Western blot. RESULTS The ferroptosis-related SLC7A11/GSH/GPX4 pathway was repressed in MI/RI rat myocardial tissues, inducing myocardial injury. H/R affected GSH synthesis and inhibited GPX4 enzyme activity by down-regulating SLC7A11, thus promoting ferroptosis in cardiomyocytes, which was averted by Lip-1. SLC7A11 overexpression improved H/R-induced cardiomyocyte ferroptosis via the GSH/GPX4 pathway. H/R activated mitophagy in cardiomyocytes. Mitophagy inhibition reversed H/R-induced cellular ferroptosis. Mitophagy activation partially averted SLC7A11 overexpression-improved H/R-induced cardiomyocyte ferroptosis. H/R suppressed the ferroptosis-related SLC7A11/GSH/GPX4 pathway by inducing mitophagy, leading to cardiomyocyte injury. CONCLUSIONS Increased ROS under H/R conditions triggered cardiomyocyte injury by inducing mitophagy to suppress the ferroptosis-related SLC7A11/GSH/GPX4 signaling pathway activation.
Collapse
Affiliation(s)
- Bingxin Chen
- Department of Cardiac Function, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, High-tech District, Urumqi, Xinjiang Uygur Autonomous Region, 830054, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Ping Fan
- Department of Cardiac Function, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, High-tech District, Urumqi, Xinjiang Uygur Autonomous Region, 830054, China
| | - Xue Song
- Department of Cardiac Function, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, High-tech District, Urumqi, Xinjiang Uygur Autonomous Region, 830054, China
| | - Mingjun Duan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Animal Experimental Center of Xinjiang Medical University, No. 137 Liyushan South Road, High-tech District, Urumqi, Xinjiang Uygur Autonomous Region, 830000, China.
| |
Collapse
|
42
|
Long Y, Xu Z, Yu J, Hu X, Xie Y, Duan X, Li N, Yan Y, Wang Y, Qin J. Targeting xCT with sulfasalazine suppresses triple-negative breast cancer growth via inducing autophagy and coordinating cell cycle and proliferation. Anticancer Drugs 2024; 35:830-843. [PMID: 39016262 DOI: 10.1097/cad.0000000000001630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
There is a substantial unmet need for effective treatment strategies in triple-negative breast cancer (TNBC). Recently, renewed attention has been directed towards targeting glutamine (Gln) metabolism to enhance the efficacy of cancer treatment. Nonetheless, a comprehensive exploration into the mechanistic implications of targeting Gln metabolism in TNBC is lacking. In this study, our objective was to probe the sensitivity of TNBC to alterations in Gln metabolism, using representative TNBC cell lines: MDA-MB-231, MDA-MB-468, and 4T1. Through an integration of bioinformatics, in-vitro, and in-vivo investigations, we demonstrated that sulfasalazine (SAS), like erastin (a known xCT inhibitor), effectively suppressed the expression and transport function of xCT, resulting in a depletion of glutathione levels in MDA-MB-231 and MDA-MB-468 cells. Furthermore, both xCT knockdown and SAS treatment demonstrated the promotion of cellular autophagy. We unveiled a positive correlation between xCT and the autophagy-related molecule p62, their co-expression indicating poor survival outcomes in breast cancer patients. In addition, our research revealed the influence of SAS and xCT on the expression of proteins regulating cell cycle and proliferation. Treatment with SAS or xCT knockdown led to the inhibition of MYC, CDK1, and CD44 expression. Significantly, the combined administration of SAS and rapamycin exhibited a synergistic inhibitory effect on the growth of transplanted breast tumor in mouse models constructed from murine-derived 4T1 cells. Taken together, our findings suggested the potential and clinical relevance of the SAS and rapamycin combination in the treatment of TNBC.
Collapse
Affiliation(s)
- Yaping Long
- Department of Immunology, School of Medicine, Nankai University
| | - Zizheng Xu
- Department of Immunology, School of Medicine, Nankai University
| | - Jing Yu
- Department of Immunology, School of Medicine, Nankai University
| | - Xiao Hu
- Department of Immunology, School of Medicine, Nankai University
| | - Yu Xie
- Department of Immunology, School of Medicine, Nankai University
| | - Xianxian Duan
- Department of Immunology, School of Medicine, Nankai University
| | - Ning Li
- School of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University
| | - Yan Yan
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Hospital of Stomatology, Nankai University, Tianjin, China
| | - Yue Wang
- Department of Immunology, School of Medicine, Nankai University
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Hospital of Stomatology, Nankai University, Tianjin, China
| | - Junfang Qin
- Department of Immunology, School of Medicine, Nankai University
| |
Collapse
|
43
|
Shi JX, Zhang ZC, Yin HZ, Piao XJ, Liu CH, Liu QJ, Zhang JC, Zhou WX, Liu FC, Yang F, Wang YF, Liu H. RNA m6A modification in ferroptosis: implications for advancing tumor immunotherapy. Mol Cancer 2024; 23:213. [PMID: 39342168 PMCID: PMC11437708 DOI: 10.1186/s12943-024-02132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
The pursuit of innovative therapeutic strategies in oncology remains imperative, given the persistent global impact of cancer as a leading cause of mortality. Immunotherapy is regarded as one of the most promising techniques for systemic cancer therapies among the several therapeutic options available. Nevertheless, limited immune response rates and immune resistance urge us on an augmentation for therapeutic efficacy rather than sticking to conventional approaches. Ferroptosis, a novel reprogrammed cell death, is tightly correlated with the tumor immune environment and interferes with cancer progression. Highly mutant or metastasis-prone tumor cells are more susceptible to iron-dependent nonapoptotic cell death. Consequently, ferroptosis-induction therapies hold the promise of overcoming resistance to conventional treatments. The most prevalent post-transcriptional modification, RNA m6A modification, regulates the metabolic processes of targeted RNAs and is involved in numerous physiological and pathological processes. Aberrant m6A modification influences cell susceptibility to ferroptosis, as well as the expression of immune checkpoints. Clarifying the regulation of m6A modification on ferroptosis and its significance in tumor cell response will provide a distinct method for finding potential targets to enhance the effectiveness of immunotherapy. In this review, we comprehensively summarized regulatory characteristics of RNA m6A modification on ferroptosis and discussed the role of RNA m6A-mediated ferroptosis on immunotherapy, aiming to enhance the effectiveness of ferroptosis-sensitive immunotherapy as a treatment for immune-resistant malignancies.
Collapse
Affiliation(s)
- Jun-Xiao Shi
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Zhi-Chao Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Hao-Zan Yin
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Xian-Jie Piao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Cheng-Hu Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Qian-Jia Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Jia-Cheng Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Wen-Xuan Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu-Chen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu Yang
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China.
- Key Laboratory of Biosafety Defense, Ministry of Education, Shanghai, 200433, China.
- Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China.
| | - Yue-Fan Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| |
Collapse
|
44
|
Xu K, Wei G, Qi W, Ye C, Liu Y, Wang S, Yang F, Tang J. CircPOLA2 sensitizes non-small cell lung cancer cells to ferroptosis and suppresses tumorigenesis via the Merlin-YAP signaling pathway. iScience 2024; 27:110832. [PMID: 39310771 PMCID: PMC11416675 DOI: 10.1016/j.isci.2024.110832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 07/14/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
Circular RNAs (circRNAs) have been implicated in the tumorigenesis of non-small cell lung cancer (NSCLC). Ferroptosis is considered a mechanism to suppress tumorigenesis. Herein, we identified a downregulated circRNA, circPOLA2 (hsa_circ_0004291), in NSCLC tissues and found that it was correlated with advanced clinical stage in patients. Nuclear-cytoplasmic fractionation assays and FISH assays confirmed that circPOLA2 was predominantly localized in the cytoplasm. Overexpression of circPOLA2 promoted lipid peroxidation and ferroptosis in NSCLC cells, thereby inhibiting cell proliferation and migration, while knockdown of circPOLA2 exerted the opposite effects. Mechanistically, circPOLA2 interacted with Merlin, a critical regulator of the Hippo pathway, and restricted Merlin phosphorylation at S518, leading to the activation of the Hippo pathway. In addition, circPOLA2 enhanced ferroptosis in NSCLC cells by activating the Hippo pathway. Together, circPOLA2 sensitizes cells to ferroptosis and suppresses tumorigenesis in NSCLC by facilitating Merlin-mediated activation of the Hippo signaling pathway.
Collapse
Affiliation(s)
- Kaiying Xu
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, People's Republic of China
| | - Guangxia Wei
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, People's Republic of China
| | - Wanghong Qi
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, People's Republic of China
| | - Chunlin Ye
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, People's Republic of China
| | - Yangyang Liu
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, People's Republic of China
| | - Shijiang Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, People's Republic of China
| | - Feng Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, People's Republic of China
| | - Jian Tang
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, People's Republic of China
- National Regional Center for Respiratory Medicine, China Japan Friendship Jiangxi Hospital, Nanchang 330000, People's Republic of China
| |
Collapse
|
45
|
Zhang C, Liu H, Li X, Xiao N, Chen H, Feng H, Li Y, Yang Y, Zhang R, Zhao X, Du Y, Bai L, Ma R, Wan J. Cold atmospheric plasma enhances SLC7A11-mediated ferroptosis in non-small cell lung cancer by regulating PCAF mediated HOXB9 acetylation. Redox Biol 2024; 75:103299. [PMID: 39127016 PMCID: PMC11363999 DOI: 10.1016/j.redox.2024.103299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/24/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Lung cancer is a leading cause of cancer death worldwide, with high incidence and poor survival rates. Cold atmospheric plasma (CAP) technology has emerged as a promising therapeutic approach for cancer treatment, inducing oxidative stress in malignant tissues without causing thermal damage. However, the role of CAP in regulating lung cancer cell ferroptosis remains unclear. Here, we observed that CAP effectively suppressed the growth and migration abilities of lung cancer cells, with significantly increased ferroptotic cell death, lipid peroxidation, and decreased mitochondrial membrane potential. Mechanistically, CAP regulates SLC7A11-mediated cell ferroptosis by modulating HOXB9. SLC7A11, a potent ferroptosis suppressor, was markedly reduced by HOXB9 knockdown, while it was enhanced by overexpressing HOXB9. The luciferase and ChIP assays confirmed that HOXB9 can directly target SLC7A11 and regulate its gene transcription. Additionally, CAP enhanced the acetylation modification level of HOXB9 by promoting its interaction with acetyltransferase p300/CBP-associated factor (PCAF). Acetylated HOXB9 affects its protein ubiquitination modification level, which in turn affects its protein stability. Notably, the upregulation of SLC7A11 and HOXB9 mitigated the suppressive effects of CAP on ferroptosis status, cell proliferation, invasion, and migration in lung cancer cells. Furthermore, animal models have also confirmed that CAP can inhibit the progression of lung cancer in vivo. Overall, this study highlights the significance of the downregulation of the HOXB9/SLC7A11 axis by CAP treatment in inhibiting lung cancer, offering novel insights into the potential mechanisms and therapeutic strategies of CAP for lung cancer.
Collapse
Affiliation(s)
- Chenxing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaohu Li
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Nan Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huanxiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haoran Feng
- Henan Key Laboratory of Ion-beam Bioengineering, Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruike Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangzhuan Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanmin Du
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lu Bai
- Department of General Surgery, Zhecheng People's Hospital, Shangqiu, Henan, China
| | - Ruonan Ma
- Henan Key Laboratory of Ion-beam Bioengineering, Zhengzhou University, Zhengzhou, Henan, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
46
|
Berndt C, Alborzinia H, Amen VS, Ayton S, Barayeu U, Bartelt A, Bayir H, Bebber CM, Birsoy K, Böttcher JP, Brabletz S, Brabletz T, Brown AR, Brüne B, Bulli G, Bruneau A, Chen Q, DeNicola GM, Dick TP, Distéfano A, Dixon SJ, Engler JB, Esser-von Bieren J, Fedorova M, Friedmann Angeli JP, Friese MA, Fuhrmann DC, García-Sáez AJ, Garbowicz K, Götz M, Gu W, Hammerich L, Hassannia B, Jiang X, Jeridi A, Kang YP, Kagan VE, Konrad DB, Kotschi S, Lei P, Le Tertre M, Lev S, Liang D, Linkermann A, Lohr C, Lorenz S, Luedde T, Methner A, Michalke B, Milton AV, Min J, Mishima E, Müller S, Motohashi H, Muckenthaler MU, Murakami S, Olzmann JA, Pagnussat G, Pan Z, Papagiannakopoulos T, Pedrera Puentes L, Pratt DA, Proneth B, Ramsauer L, Rodriguez R, Saito Y, Schmidt F, Schmitt C, Schulze A, Schwab A, Schwantes A, Soula M, Spitzlberger B, Stockwell BR, Thewes L, Thorn-Seshold O, Toyokuni S, Tonnus W, Trumpp A, Vandenabeele P, Vanden Berghe T, Venkataramani V, Vogel FCE, von Karstedt S, Wang F, Westermann F, Wientjens C, Wilhelm C, Wölk M, Wu K, Yang X, Yu F, Zou Y, Conrad M. Ferroptosis in health and disease. Redox Biol 2024; 75:103211. [PMID: 38908072 PMCID: PMC11253697 DOI: 10.1016/j.redox.2024.103211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
Ferroptosis is a pervasive non-apoptotic form of cell death highly relevant in various degenerative diseases and malignancies. The hallmark of ferroptosis is uncontrolled and overwhelming peroxidation of polyunsaturated fatty acids contained in membrane phospholipids, which eventually leads to rupture of the plasma membrane. Ferroptosis is unique in that it is essentially a spontaneous, uncatalyzed chemical process based on perturbed iron and redox homeostasis contributing to the cell death process, but that it is nonetheless modulated by many metabolic nodes that impinge on the cells' susceptibility to ferroptosis. Among the various nodes affecting ferroptosis sensitivity, several have emerged as promising candidates for pharmacological intervention, rendering ferroptosis-related proteins attractive targets for the treatment of numerous currently incurable diseases. Herein, the current members of a Germany-wide research consortium focusing on ferroptosis research, as well as key external experts in ferroptosis who have made seminal contributions to this rapidly growing and exciting field of research, have gathered to provide a comprehensive, state-of-the-art review on ferroptosis. Specific topics include: basic mechanisms, in vivo relevance, specialized methodologies, chemical and pharmacological tools, and the potential contribution of ferroptosis to disease etiopathology and progression. We hope that this article will not only provide established scientists and newcomers to the field with an overview of the multiple facets of ferroptosis, but also encourage additional efforts to characterize further molecular pathways modulating ferroptosis, with the ultimate goal to develop novel pharmacotherapies to tackle the various diseases associated with - or caused by - ferroptosis.
Collapse
Affiliation(s)
- Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Vera Skafar Amen
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - Uladzimir Barayeu
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany; Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany; German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York City, NY, USA
| | - Christina M Bebber
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Kivanc Birsoy
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Ashley R Brown
- Department of Biological Sciences, Columbia University, New York City, NY, USA
| | - Bernhard Brüne
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Giorgia Bulli
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany
| | - Alix Bruneau
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Gina M DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Ayelén Distéfano
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jan B Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | | | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | - Dominic C Fuhrmann
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Ana J García-Sáez
- Institute for Genetics, CECAD, University of Cologne, Germany; Max Planck Institute of Biophysics, Frankfurt/Main, Germany
| | | | - Magdalena Götz
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, Germany
| | - Wei Gu
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | | | - Xuejun Jiang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Aicha Jeridi
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), Germany, Member of the German Center for Lung Research (DZL)
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Republic of Korea
| | | | - David B Konrad
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Peng Lei
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Marlène Le Tertre
- Center for Translational Biomedical Iron Research, Heidelberg University, Germany
| | - Sima Lev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Deguang Liang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany; Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Carolin Lohr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Svenja Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Axel Methner
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Bernhard Michalke
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Germany
| | - Anna V Milton
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Junxia Min
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | | | - Shohei Murakami
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Gabriela Pagnussat
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Zijan Pan
- School of Life Sciences, Westlake University, Hangzhou, China
| | | | | | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Canada
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Lukas Ramsauer
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | | | - Yoshiro Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Felix Schmidt
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Carina Schmitt
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Almut Schulze
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Annemarie Schwab
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Anna Schwantes
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Mariluz Soula
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Benedikt Spitzlberger
- Department of Immunobiology, Université de Lausanne, Switzerland; Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York City, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Chemistry, Columbia University, New York, NY, USA
| | - Leonie Thewes
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Nagoya, Japan
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Department of Biomedical Sciences, University of Antwerp, Belgium; VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Vivek Venkataramani
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Germany
| | - Felix C E Vogel
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silvia von Karstedt
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Germany
| | - Fudi Wang
- School of Medicine, Zhejiang University, Hangzhou, China
| | | | - Chantal Wientjens
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - Katherine Wu
- Department of Pathology, Grossman School of Medicine, New York University, NY, USA
| | - Xin Yang
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Fan Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yilong Zou
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany.
| |
Collapse
|
47
|
Li K, Yang B, Du Y, Ding Y, Shen S, Sun Z, Liu Y, Wang Y, Cao S, Ren W, Wang X, Li M, Zhang Y, Wu J, Zheng W, Yan W, Li L. The HOXC10/NOD1/ERK axis drives osteolytic bone metastasis of pan-KRAS-mutant lung cancer. Bone Res 2024; 12:47. [PMID: 39191757 PMCID: PMC11349752 DOI: 10.1038/s41413-024-00350-8] [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: 12/07/2023] [Revised: 06/09/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024] Open
Abstract
While KRAS mutation is the leading cause of low survival rates in lung cancer bone metastasis patients, effective treatments are still lacking. Here, we identified homeobox C10 (HOXC10) as a lynchpin in pan-KRAS-mutant lung cancer bone metastasis. Through RNA-seq approach and patient tissue studies, we demonstrated that HOXC10 expression was dramatically increased. Genetic depletion of HOXC10 preferentially impeded cell proliferation and migration in vitro. The bioluminescence imaging and micro-CT results demonstrated that inhibition of HOXC10 significantly reduced bone metastasis of KRAS-mutant lung cancer in vivo. Mechanistically, the transcription factor HOXC10 activated NOD1/ERK signaling pathway to reprogram epithelial-mesenchymal transition (EMT) and bone microenvironment by activating the NOD1 promoter. Strikingly, inhibition of HOXC10 in combination with STAT3 inhibitor was effective against KRAS-mutant lung cancer bone metastasis by triggering ferroptosis. Taken together, these findings reveal that HOXC10 effectively alleviates pan-KRAS-mutant lung cancer with bone metastasis in the NOD1/ERK axis-dependent manner, and support further development of an effective combinatorial strategy for this kind of disease.
Collapse
Affiliation(s)
- Kun Li
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Health Science Center, East China Normal University, Shanghai, 200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China
| | - Bo Yang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yingying Du
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yi Ding
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Shihui Shen
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, 200240, China
| | - Zhengwang Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yun Liu
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yuhan Wang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Siyuan Cao
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wenjie Ren
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiangyu Wang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mengjuan Li
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yunpeng Zhang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Juan Wu
- Department of Pharmacy The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Wei Zheng
- Orthopaedic Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
- Department of Orthopedics, General Hospital of Western Theater Command, Chengdu, 610000, China.
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China.
| | - Wangjun Yan
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Lei Li
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China.
- School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, 200240, China.
| |
Collapse
|
48
|
Nokin MJ, Darbo E, Richard E, San José S, de Hita S, Prouzet-Mauleon V, Turcq B, Gerardelli L, Crake R, Velasco V, Koopmansch B, Lambert F, Xue JY, Sang B, Horne J, Ziemons E, Villanueva A, Blomme A, Herfs M, Cataldo D, Calvayrac O, Porporato P, Nadal E, Lito P, Jänne PA, Ricciuti B, Awad MM, Ambrogio C, Santamaría D. In vivo vulnerabilities to GPX4 and HDAC inhibitors in drug-persistent versus drug-resistant BRAF V600E lung adenocarcinoma. Cell Rep Med 2024; 5:101663. [PMID: 39094577 PMCID: PMC11384943 DOI: 10.1016/j.xcrm.2024.101663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 05/22/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024]
Abstract
The current targeted therapy for BRAFV600E-mutant lung cancer consists of a dual blockade of RAF/MEK kinases often combining dabrafenib/trametinib (D/T). This regimen extends survival when compared to single-agent treatments, but disease progression is unavoidable. By using whole-genome CRISPR screening and RNA sequencing, we characterize the vulnerabilities of both persister and D/T-resistant cellular models. Oxidative stress together with concomitant induction of antioxidant responses is boosted by D/T treatment. However, the nature of the oxidative damage, the choice of redox detoxification systems, and the resulting therapeutic vulnerabilities display stage-specific differences. Persister cells suffer from lipid peroxidation and are sensitive to ferroptosis upon GPX4 inhibition in vivo. Biomarkers of lipid peroxidation are detected in clinical samples following D/T treatment. Acquired alterations leading to mitogen-activated protein kinase (MAPK) reactivation enhance cystine transport to boost GPX4-independent antioxidant responses. Similarly to BRAFV600E-mutant melanoma, histone deacetylase (HDAC) inhibitors decrease D/T-resistant cell viability and extend therapeutic response in vivo.
Collapse
Affiliation(s)
- Marie-Julie Nokin
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France; Laboratory of Biology of Tumor and Development (LBTD), GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Elodie Darbo
- Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France
| | - Elodie Richard
- Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France
| | - Sonia San José
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France; Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain
| | - Sergio de Hita
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France; Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France; Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain
| | - Valérie Prouzet-Mauleon
- Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France; CRISP'edit, TBMCore, University of Bordeaux, CNRS UAR 3427, INSERM US05, 33000 Bordeaux, France
| | - Béatrice Turcq
- Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France; CRISP'edit, TBMCore, University of Bordeaux, CNRS UAR 3427, INSERM US05, 33000 Bordeaux, France
| | - Laura Gerardelli
- Laboratory of Biology of Tumor and Development (LBTD), GIGA-Cancer, University of Liege, 4000 Liege, Belgium
| | - Rebekah Crake
- Laboratory of Biology of Tumor and Development (LBTD), GIGA-Cancer, University of Liege, 4000 Liege, Belgium
| | - Valérie Velasco
- Department of Biopathology, Institut Bergonié, 33076 Bordeaux, France
| | - Benjamin Koopmansch
- Department of Human Genetics, University Hospital Center of Liege, 4000 Liege, Belgium
| | - Frederic Lambert
- Department of Human Genetics, University Hospital Center of Liege, 4000 Liege, Belgium
| | - Jenny Y Xue
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ben Sang
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Julie Horne
- Laboratory of Pharmaceutical Analytical Chemistry, CIRM, University of Liege, 4000 Liege, Belgium
| | - Eric Ziemons
- Laboratory of Pharmaceutical Analytical Chemistry, CIRM, University of Liege, 4000 Liege, Belgium
| | - Alberto Villanueva
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO); Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet del Llobregat, 08908 Barcelona, Spain
| | - Arnaud Blomme
- Laboratory of Cancer Signaling, GIGA-Stem Cells, University of Liege, 4000 Liege, Belgium
| | - Michael Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, 4000 Liege, Belgium
| | - Didier Cataldo
- Laboratory of Biology of Tumor and Development (LBTD), GIGA-Cancer, University of Liege, 4000 Liege, Belgium
| | - Olivier Calvayrac
- Cancer Research Centre of Toulouse, INSERM UMR1037, CNRS UMR5071, 31100 Toulouse, France
| | - Paolo Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy
| | - Ernest Nadal
- Molecular Mechanisms of Cancer Program, Department of Medical Oncology, Catalan Institute of Oncology (ICO), Preclinical and Experimental Research in Thoracic Tumors (PReTT) Group, Oncobell Program, IDIBELL, L'Hospitalet del Llobregat, 08908 Barcelona, Spain
| | - Piro Lito
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Biagio Ricciuti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Mark M Awad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy.
| | - David Santamaría
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France; Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
49
|
Yao H, Jiang W, Liao X, Wang D, Zhu H. Regulatory mechanisms of amino acids in ferroptosis. Life Sci 2024; 351:122803. [PMID: 38857653 DOI: 10.1016/j.lfs.2024.122803] [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: 03/12/2024] [Revised: 05/19/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Ferroptosis, an iron-dependent non-apoptotic regulated cell death process, is associated with the pathogenesis of various diseases. Amino acids, which are indispensable substrates of vital activities, significantly regulate ferroptosis. Amino acid metabolism is involved in maintaining iron and lipid homeostasis and redox balance. The regulatory effects of amino acids on ferroptosis are complex. An amino acid may exert contrasting effects on ferroptosis depending on the context. This review systematically and comprehensively summarized the distinct roles of amino acids in regulating ferroptosis and highlighted the emerging opportunities to develop clinical therapeutic strategies targeting amino acid-mediated ferroptosis.
Collapse
Affiliation(s)
- Heying Yao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Wei Jiang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Xiang Liao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China
| | - Dongqing Wang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
50
|
Lin H, Wang L, Jiang X, Wang J. Glutathione dynamics in subcellular compartments and implications for drug development. Curr Opin Chem Biol 2024; 81:102505. [PMID: 39053236 PMCID: PMC11722958 DOI: 10.1016/j.cbpa.2024.102505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/30/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
Glutathione (GSH) is a pivotal tripeptide antioxidant essential for maintaining cellular redox homeostasis and regulating diverse cellular processes. Subcellular compartmentalization of GSH underscores its multifaceted roles across various organelles including the cytosol, mitochondria, endoplasmic reticulum, and nucleus, each exhibiting distinct regulatory mechanisms. Perturbations in GSH dynamics contribute to pathophysiological conditions, emphasizing the clinical significance of understanding its intricate regulation. This review consolidates current knowledge on subcellular GSH dynamics, highlighting its implications in drug development, particularly in covalent drug design and antitumor strategies targeting intracellular GSH levels. Challenges and future directions in deciphering subcellular GSH dynamics are discussed, advocating for innovative methodologies to advance our comprehension and facilitate the development of precise therapeutic interventions based on GSH modulation.
Collapse
Affiliation(s)
- Hanfeng Lin
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; Center for NextGen Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lingfei Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiqian Jiang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jin Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; Center for NextGen Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|