1
|
Bai X, Duan T, Shao J, Zhang Y, Xing G, Wang J, Liu X, Wang M, He Y, Wang H, Zhang ZY, Ni M, Zhou JY, Pan J. CBX3 promotes multidrug resistance by suppressing ferroptosis in colorectal carcinoma via the CUL3/NRF2/GPX2 axis. Oncogene 2025; 44:1678-1693. [PMID: 40089640 PMCID: PMC12122363 DOI: 10.1038/s41388-025-03337-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 02/01/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Chemoresistance poses a significant challenge in colorectal cancer (CRC) treatment. However, the mechanisms underlying chemoresistance remain unclear. CBX3 promoted proliferation and metastasis in CRC. However, the role and mechanism of CBX3 in chemoresistance remain unknown. Therefore, we aimed to investigate the effects and mechanisms of CBX3 on multidrug resistance in CRC. Our studies showed that higher levels of CBX3 expression were associated with poor survival, especially in groups with progression following chemotherapy. CBX3 overexpression increased Irinotecan and Oxaliplatin resistance, whereas CBX3 knockdown suppressed multidrug resistance in CRC cells. Additionally, CBX3 inhibited ferroptosis associated with multidrug resistance, and the ferroptosis activators prevented CBX3 overexpression-mediated cell survival. RNA sequencing revealed that the NRF2-signaling pathway was involved in this process. CBX3-upregulated NRF2 protein expression by directly binding to the promoter of Cullin3 (CUL3) to suppress CUL3 transcription and CUL3-mediated NRF2 degradation. Moreover, Glutathione Peroxidase 2 (GPX2) was downstream of the CBX3-NRF2 pathway in CRC chemoresistance. ML385, an NRF2 inhibitor, suppressed GPX2 expression, and increased ferroptosis in PDX models. Our study identified CBX3/NRF2/GPX2 axis may be a novel signaling pathway that mediates multidrug resistance in CRC. This study proposes developing novel strategies for cancer treatment to overcome drug resistance in the future.
Collapse
Affiliation(s)
- Xiaoming Bai
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Tinghong Duan
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
- Department of Pathology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, PR China
| | - Jiaofang Shao
- Department of Bioinformatics, Nanjing Medical University, Nanjing, PR China
| | - Yutong Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Guangyuan Xing
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Jie Wang
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Xue Liu
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Min Wang
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Yuanqiao He
- Center of Laboratory Animal Science, Nanchang University, Nanchang, PR China
- Key Laboratory of New Drug Evaluation and Transformation of Jiangxi Province, Nanchang Royo Biotech Co., Ltd, Nanchang, PR China
| | - Hai Wang
- Department of Pathology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, PR China
| | - Zhi-Yuan Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Min Ni
- Department of Colorectum, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, PR China.
| | - Jin-Yong Zhou
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, PR China.
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, PR China.
| | - Jinshun Pan
- Department of Biotherapy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
2
|
Zhang L, Zhu LJ, Zhao Y, Lei XY, Wu DH, He KY, Liu MJ, Yang JY, Guo JR, Jiang ZH, Yan ZH, Xu ZX, Jian YP. STING inhibits the progression of esophageal squamous cell carcinoma by suppressing CPT1A-mediated fatty acid β-oxidation. Acta Pharmacol Sin 2025:10.1038/s41401-025-01581-z. [PMID: 40394235 DOI: 10.1038/s41401-025-01581-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is characterized by high aggressiveness and poor prognosis. Metabolic reprogramming is a hallmark of ESCC, with lipid metabolism frequently upregulated. It has been shown that lipid metabolism, particularly fatty acid β-oxidation (FAO), plays an essential role in energy homeostasis, membrane biosynthesis, and tumor progression. Stimulator of interferon genes (STING), a key innate immune signaling molecule, also acts as a metabolic checkpoint by inhibiting hexokinase 2, thereby limiting aerobic glycolysis and enhancing anti-tumor immune responses. In this study, we investigated the impact of STING on FAO and tumorigenesis in ESCC. We showed that the expression levels of STING were significantly reduced in ESCC compared to adjacent normal tissue. In the ESCC cell line KYSE-510, knockdown of STING significantly elevated lipid metabolites, decreased intracellular lipid droplets, and increased FAO products, whereas overexpression of STING inhibited ESCC cell proliferation and tumor progression by suppressing FAO. Targeted lipid metabolomic analyses revealed that STING interacted with carnitine palmitoyltransferase 1A (CPT1A), a key enzyme in FAO. STING promoted the ubiquitination and degradation of CPT1A by disrupting its interaction with USP15, a deubiquitinating enzyme. Treatment with the CPT1A inhibitor etomoxir (50 μM) reversed the increased FAO induced by STING depletion in KYSE-30 cells. In both in vitro and in vivo models, supplementation with palmitic acid rescued STING-induced growth inhibition, restoring tumor cell growth. In addition, STING knockout in 4-NQO-induced ESCC mice led to accelerated tumor progression, which could be mitigated by CPT1A inhibition. Our results suggest that reduced STING expression enhances FAO and promotes ESCC cell proliferation, implicating FAO suppression as a potential therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Ling-Jun Zhu
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Zhi-Hao Jiang
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Zhen-Hua Yan
- School of Life Sciences, Henan University, Kaifeng, 475001, China
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, 475001, China.
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, 475001, China.
| |
Collapse
|
3
|
Chen C, Zhang Y, Zang Y, Fan Z, Han Y, Bai X, Wang A, Zhang J, Wang J, Zhang K. SIRT3 Functions as an Eraser of Histone H3K9 Lactylation to Modulate Transcription for Inhibiting the Progression of Esophageal Cancer. Mol Cell Proteomics 2025; 24:100973. [PMID: 40252727 PMCID: PMC12144510 DOI: 10.1016/j.mcpro.2025.100973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
Lysine lactylation (Kla) links lactate metabolism to epigenetic regulation, playing a key role in modulation of gene expression in tumor and immune microenvironment. Our recent study shows that HBO1-mediated histone H3K9la activates the transcription of genes encoding tumorigenesis, suggesting the potential significance of intervening in this Kla site for tumor therapy. Evidence so far indicates that traditional deacetylases can catalyze the removal of Kla; however, the precise demodifying enzyme to histone H3K9la in vivo and functional consequence remain elusive. Herein, we combined an antibody-based proximity labeling approach with mass spectrometry analysis to identify SIRT3 as a major binder to histone H3K9la and showed the specific catalysis of SIRT3 for the removal of lactylation. Molecular docking further revealed the molecular mechanism of the binding of histone H3K9la to SIRT3. More importantly, SIRT3 can specifically modulate gene transcription by regulating H3K9la, inhibiting the progression of esophageal squamous cancer cells. Together, our work identifies the specific delactylase of H3K9la and reveals an H3K9la-mediated molecular mechanism catalyzed by SIRT3 for gene transcription regulation in esophageal squamous cancer cells, and our findings provide an opportunity to investigate the physiological significance of Kla controlled by SIRT3 in cancer.
Collapse
Affiliation(s)
- Chen Chen
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| | - Yingao Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yong Zang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China; School of Biomedical Engineer, Tianjin Medical University, Tianjin, China
| | - Zilong Fan
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China; Department of Bioinformatics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Yanpu Han
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xue Bai
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Aiyuan Wang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China; Department of Bioinformatics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Jianji Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ju Wang
- School of Biomedical Engineer, Tianjin Medical University, Tianjin, China
| | - Kai Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China; Department of Bioinformatics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China; State Key Laboratory of Experimental Hematology, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
4
|
Yang Y, Deng X, Li W, Leng Y, Xiong Y, Wang B, Gong S, Wang Y, Yang B, Li W. Targeting the epigenetic regulation of ferroptosis: a potential therapeutic approach for sepsis-associated acute kidney injury. Clin Epigenetics 2025; 17:57. [PMID: 40189571 PMCID: PMC11974148 DOI: 10.1186/s13148-025-01861-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
Sepsis is a syndrome of organ dysfunction caused by the invasion of pathogenic microorganisms. In clinical practice, patients with sepsis are prone to concurrent acute kidney injury, which has high morbidity and mortality rates. Thus, understanding the pathogenesis of sepsis-associated acute kidney injury is of significant clinical importance. Ferroptosis is an iron-dependent programmed cell death pathway, which is proved to play a critical role in the process of sepsis-associated acute kidney injury through various mechanisms. Epigenetic regulation modulates the content and function of nucleic acids and proteins within cells through various modifications. Its impact on ferroptosis has garnered increasing attention; however, the role of epigenetic regulation targeting ferroptosis in sepsis-associated acute kidney injury has not been fully elucidated. Growing evidence suggests that epigenetic regulation can modulate ferroptosis through complex pathway networks, thereby affecting the development and prognosis of sepsis-associated acute kidney injury. This paper summarizes the impact of ferroptosis on sepsis-associated acute kidney injury and the regulatory mechanisms of epigenetic regulation on ferroptosis, providing new insights for the targeted therapy of sepsis-associated acute kidney injury.
Collapse
Affiliation(s)
- Yuhang Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xinqi Deng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wenyuan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yan Leng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yonghong Xiong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Bihan Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Siyuan Gong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yunhao Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Baichuan Yang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
5
|
Zhang Q, Chen D, Liu X, Deng Z, Li J, Zhu S, Ma B, Liu R, Zhu H. High Photocytotoxicity Iridium(III) Complex Photosensitizer for Photodynamic Therapy Induces Antitumor Effect Through GPX4-Dependent Ferroptosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2403165. [PMID: 39246173 DOI: 10.1002/smll.202403165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/21/2024] [Indexed: 09/10/2024]
Abstract
The development of small molecule photosensitizers based on iridium complex is limited by the mismatch between therapeutic effect and systemic toxicity, as well as the incomplete understanding of the molecular mechanism underlying cell death induction. Herein, a small molecule iridium complex IrC with high photocytotoxicity is synthesized, with half maximal inhibitory concentration as low as 91 nm, demonstrating excellent anti-tumor, relief of splenomegaly, and negligible side effects. Starting from the factors of effective photosensitizers, the in-depth theoretical analysis on photon absorption efficiency, energy transfer level matching, and properties of the triplet excited state of IrC is conducted. This also elucidates the feasibility of generating the high singlet oxygen quantum yield. In addition, the death mechanism induced by IrC is focused, innovatively utilizing GPX4-overexpression and GPX4-knockout cells via CRISPR/Cas9 technique to comprehensively verify ferroptosis and its further molecular mechanism. The generation of ROS mediated by IrC, along with the direct inhibition of GPX4 and glutathione, synergistically increased cellular oxidative stress and the level of lipid peroxidation. This study provides an effective approach for small molecule complexes to induce GPX4-dependent ferroptosis at low-dose photodynamic therapy.
Collapse
Affiliation(s)
- Qing Zhang
- School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Dezhi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Xiaomeng Liu
- School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Zhewen Deng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Jiaqi Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Senqiang Zhu
- School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816, China
| | - Rui Liu
- School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Hongjun Zhu
- School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, 211816, China
| |
Collapse
|
6
|
Yang W, Gu M, Zhang Y, Zhang Y, Liu T, Wu D, Deng J, Liu M, Zhang Y. A method for the identification of lactate metabolism-related prognostic biomarkers and its validations in non-small cell lung cancer. Sci Rep 2025; 15:5812. [PMID: 39962075 PMCID: PMC11832748 DOI: 10.1038/s41598-025-85620-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Lactate metabolism (LM) plays a crucial role in tumor progression and therapy resistance in non-small cell lung cancer (NSCLC). Several methods had been developed for NSCLC prognosis prediction based on lactate metabolism-related information. The existing methods for the construction of prognosis prediction models are mostly based on single models such as linear models, SVM, and decision trees. Prognosis biomarkers and prognosis prediction models based on this kind of methods often have limited prognostic performance. In this study, we proposed a novel methodology for constructing prognosis prediction model and identifying lactate-related prognostic biomarkers in NSCLC. We first screened for lactate metabolism-related malignant genes from the scRNA-Seq data of NSCLC malignant cells. We proposed a Cox elastic-net regression combined with genetic algorithm (GA-EnCox) to predict prognosis and optimize the selection of key biomarkers. We identified five key LM-related genes (LYPD3, KRT8, CCT6A, PSMB7, and HMGA1) that significantly correlated with patient prognosis in LUAD cohorts. The prognostic model constructed with these genes outperformed other currently popular models across multiple datasets, demonstrating stable predictive capability. Survival analysis based on bulk RNA-Seq data demonstrated that the low-risk group had significantly better overall survival compared to the high-risk group. Further analysis revealed that lactate metabolism-related prognosis risk might be associated with monocyte lineages such as macrophages and DC's infiltration and these prognosis biomarkers may indicate the therapeutic responses of immune checkpoint inhibitors for NSCLC patients. More importantly, we validated HMGA1 and KRT8 at protein level and their association with histologic grades, stages, and clinical outcomes in consistently treated in-house NSCLC cohorts. Finally, we experimentally validated one of the biomarkers, HMGA1, confirming its role in promoting malignant phenotypes of NSCLC. This study provides valuable insights into the role of lactate metabolism-related biomarkers and their impact on patient outcomes, it was expected to provide important reference value for prognosis assessment and personalized treatment decision of NSCLC patients.
Collapse
Affiliation(s)
- Weiyang Yang
- Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Miao Gu
- Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Yabin Zhang
- Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Yunfan Zhang
- Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Tao Liu
- Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Di Wu
- Department of Medical Oncology, Xuzhou Central Hospital, Xuzhou, 221009, China
| | - Juntao Deng
- Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Min Liu
- Department of Automation, Tsinghua University, Beijing, 100084, China.
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Xuzhou, 221009, China.
| |
Collapse
|
7
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2025; 480:759-784. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [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: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
8
|
Desterke C, Fu Y, Bonifacio-Mundaca J, Monge C, Pineau P, Mata-Garrido J, Francés R. Ferroptosis Transcriptional Regulation and Prognostic Impact in Medulloblastoma Subtypes Revealed by RNA-Seq. Antioxidants (Basel) 2025; 14:96. [PMID: 39857430 PMCID: PMC11761645 DOI: 10.3390/antiox14010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children, typically arising during infancy and childhood. Despite multimodal therapies achieving a response rate of 70% in children older than 3 years, treatment remains challenging. Ferroptosis, a form of regulated cell death, can be induced in medulloblastoma cells in vitro using erastin or RSL3. Using two independent medulloblastoma RNA-sequencing cohorts (MB-PBTA and MTAB-10767), we investigated the expression of ferroptosis-related molecules through multiple approaches, including Weighted Gene Co-Expression Network Analysis (WGCNA), molecular subtype stratification, protein-protein interaction (PPI) networks, and univariable and multivariable overall survival analyses. A prognostic expression score was computed based on a cross-validated ferroptosis signature. In training and validation cohorts, the regulation of the ferroptosis transcriptional program distinguished the four molecular subtypes of medulloblastoma. WGCNA identified nine gene modules in the MB tumor transcriptome; five correlated with molecular subtypes, implicating pathways related to oxidative stress, hypoxia, and trans-synaptic signaling. One module, associated with disease recurrence, included epigenetic regulators and nucleosome organizers. Univariable survival analyses identified a 45-gene ferroptosis prognostic signature associated with nutrient sensing, cysteine and methionine metabolism, and trans-sulfuration within a one-carbon metabolism. The top ten unfavorable ferroptosis genes included CCT3, SNX5, SQOR, G3BP1, CARS1, SLC39A14, FAM98A, FXR1, TFAP2C, and ATF4. Patients with a high ferroptosis score showed a worse prognosis, particularly in the G3 and SHH subtypes. The PPI network highlighted IL6 and CBS as unfavorable hub genes. In a multivariable overall survival model, which included gender, age, and the molecular subtype classification, the ferroptosis expression score was validated as an independent adverse prognostic marker (hazard ratio: 5.8; p-value = 1.04 × 10-9). This study demonstrates that the regulation of the ferroptosis transcriptional program is linked to medulloblastoma molecular subtypes and patient prognosis. A cross-validated ferroptosis signature was identified in two independent RNA-sequencing cohorts, and the ferroptosis score was confirmed as an independent and adverse prognostic factor in medulloblastoma.
Collapse
Affiliation(s)
- Christophe Desterke
- INSERM UMRS-1310, Faculté de Médecine du Kremlin Bicêtre, Université Paris-Saclay, F-94270 Le Kremlin-Bicêtre, France;
| | - Yuanji Fu
- INSERM, CNRS, Institut Necker Enfants Malades, Université Paris Cité, F-75015 Paris, France;
| | - Jenny Bonifacio-Mundaca
- National Tumor Bank, Department of Pathology, National Institute of Neoplastic Diseases, Surquillo 15038, Peru;
| | - Claudia Monge
- Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, INSERM U993, F-75015 Paris, France; (C.M.); (P.P.)
| | - Pascal Pineau
- Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, INSERM U993, F-75015 Paris, France; (C.M.); (P.P.)
| | - Jorge Mata-Garrido
- Unité Organisation Nucléaire et Oncogenèse, Institut Pasteur, Université Paris Cité, INSERM U993, F-75015 Paris, France; (C.M.); (P.P.)
| | - Raquel Francés
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, F-75006 Paris, France
| |
Collapse
|
9
|
Wu DH, Zhao ZL, Yin WT, Liu H, Xiang XY, Zhu LJ, Li JQ, Yan ZH, Li YJ, Jian YP, Xu ZX. STING exerts antiviral innate immune response by activating pentose phosphate pathway. Cell Commun Signal 2024; 22:599. [PMID: 39695767 DOI: 10.1186/s12964-024-01983-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The innate immune system serves as the host's first line of defense against invading pathogens. Stimulator of interferon genes (STING) is a key component of this system, yet its relationship with glucose metabolism, particularly in antiviral immunity, remains underexplored. METHODS Metabolomics analysis was used for detecting metabolic alterations in spleens from STING knockout (KO) and wild-type (WT) mice. Co-immunoprecipitation was employed for determining ubiquitination of TKT. Mass spectrometry was used for detecting interaction proteins of STING. Enzyme activity kits were used for detecting the activities of TKT and G6PD. RESULTS In this study, we demonstrate that herpes simplex virus (HSV) infection activates the pentose phosphate pathway (PPP) in host cells, thereby initiating an antiviral immune response. Using STING-manipulated cells and systemic knockout mice, we show that STING positively regulates PPP, which, in turn, limits HSV infection. Inhibition of the PPP significantly reduced the production of antiviral immune factors and dampened STING-induced innate immune responses. Mechanistically, we discovered that STING interacts with transketolase (TKT), a key enzyme in the non-oxidative branch of the PPP, and reduces its ubiquitination via the E3 ubiquitin ligase UBE3A, stabilizing TKT. Silencing TKT or inhibiting its activity with oxythiamine diminished antiviral immune factor production. CONCLUSION Our findings reveal that the PPP plays a synergistic role in generating antiviral immune factors during viral infection and suggest that PPP activation could serve as an adjunct strategy for antiviral therapy.
Collapse
Affiliation(s)
- Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Zi-Long Zhao
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Wei-Tao Yin
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Huai Liu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Xiong-Yan Xiang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Ling-Jun Zhu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Zhen-Hua Yan
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yu-Jia Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| |
Collapse
|
10
|
Huang Fu ZM, Xiao M, Xie H, Zhang S, Yi T, Li Q, Li M, Wang Y. Suppressing GDF15 enhances the chemotherapeutic effect of 5 FU on MSI-H CRC by regulating the ferroptosis pathway SLC7A11/GSH/GPX4. J Cancer Res Clin Oncol 2024; 151:6. [PMID: 39636345 PMCID: PMC11621149 DOI: 10.1007/s00432-024-06036-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
Growth differentiation factor 15 (GDF15) is a member of the transforming growth factor beta (TGF-β) superfamily and is related to metabolism, injury, and aging. GDF15 has both tumor-promoting and tumor-suppressing effects. However, its role in colorectal cancer (CRC) with high microsatellite instability (MSI-H) must be further clarified. In our study, we found that GDF15 is generally elevated in pancarcinoma, particularly in colorectal cancer, and serves as an early indicator of the development of colorectal cancer. IHC and WB confirmed that GDF15 was elevated in MSI-H CRC clinical tissues and MSI-H CRC cell lines (HCT-116 and LoVo). Suppressing GDF15 by siRNA resulted in a substantial decrease in cell viability and proliferation. Furthermore, suppressing GDF15 can increase the sensitivity of MSI-H CRC cells to 5-fluorouracil (5-FU), which decreases cell viability and increases the apoptosis rate. In vivo experiments also demonstrated that mouse xenografts with suppressed GDF15 expression were more susceptible to 5-FU chemotherapy. We examined alterations in mitochondria via electron microscopy and changes in the mitochondrial membrane potential, ferroptosis-related signals (MDA, Fe2+), and SLC7A11/GSH/GPX4 protein pathway. Our research indicates that inhibiting GDF15 affects ferroptosis-related pathways, leading to ferroptosis and improving the MSI-H CRC response to 5-FU therapy. As a result, GDF15 could be a promising target for diagnosing and treating MSI-H CRC, potentially enhancing the overall effectiveness of therapy for patients with MSI-H CRC.
Collapse
Affiliation(s)
- Zhi Min Huang Fu
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
- Department of Pathology, The Second Hospital Affiliated to Third Military Medical University, XinQiao Hospital, Chongqing, China
| | - Ming Xiao
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Hailun Xie
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Shuxian Zhang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Tang Yi
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Qingshu Li
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China
| | - Ming Li
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China.
| | - Yalan Wang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Department of Clinical Pathololgy, Laboratory of Pathology Diagnostic Center, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
11
|
Lei XY, He KY, Li QT, Zhang L, Wu DH, Yang JY, Guo JR, Liu MJ, Zhao ZL, Li JQ, Liu H, Zhao Y, Li YJ, Sun QH, Wu CG, Wang YF, Cao GS, Wang G, Jian YP, Xu ZX. PARylation of HMGA1 desensitizes esophageal squamous cell carcinoma to olaparib. Clin Transl Med 2024; 14:e70111. [PMID: 39690136 DOI: 10.1002/ctm2.70111] [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: 04/11/2024] [Revised: 11/01/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
As a chromatin remodelling factor, high mobility group A1 (HMGA1) plays various roles in both physiological and pathological conditions. However, its role in DNA damage response and DNA damage-based chemotherapy remains largely unexplored. In this study, we report the poly ADP-ribosylation (PARylation) of HMGA1 during DNA damage, leading to desensitization of esophageal squamous cell carcinoma (ESCC) cells to the poly(ADP-ribose) polymerase 1 (PARP1) inhibitor, olaparib. We found that HMGA1 accumulates at sites of DNA damage, where it interacts with PARP1 and undergoes PARylation at residues E47 and E50 in its conserved AT-hook domain. This modification enhances the accumulation of Ku70/Ku80 at the site of DNA damage and activates the DNA-dependent protein kinase catalytic subunit, facilitating nonhomologous end-joining repair. In both subcutaneous tumour models and genetically engineered mouse models of in situ esophageal cancer, HMGA1 interference increased tumour sensitivity to olaparib. Moreover, HMGA1 was highly expressed in ESCC tissues and positively correlated with PARP1 levels as well as poor prognosis in ESCC patients. Taken together, these findings reveal a mechanistic link between HMGA1 and PARP1 in regulating cell responses to DNA damage and suggest that targeting HMGA1 could be a promising strategy to increase cancer cell sensitivity to olaparib.
Collapse
Affiliation(s)
- Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qiu-Tong Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, China
| | - Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Zi-Long Zhao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Huai Liu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yu-Jia Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qian-Hui Sun
- School of Life Sciences, Henan University, Kaifeng, China
| | - Chen-Guang Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yun-Fan Wang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Geng-Sheng Cao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Gang Wang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, China
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, China
| |
Collapse
|
12
|
Veglia Tranchese R, Battista S, Cerchia L, Fedele M. Ferroptosis in Cancer: Epigenetic Control and Therapeutic Opportunities. Biomolecules 2024; 14:1443. [PMID: 39595619 PMCID: PMC11592303 DOI: 10.3390/biom14111443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical pathway in cancer biology. This review delves into the epigenetic mechanisms that modulate ferroptosis in cancer cells, focusing on how DNA methylation, histone modifications, and non-coding RNAs influence the expression and function of essential genes involved in this process. By unraveling the complex interplay between these epigenetic mechanisms and ferroptosis, the article sheds light on novel gene targets and functional insights that could pave the way for innovative cancer treatments to enhance therapeutic efficacy and overcome resistance in cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council—CNR, 80131 Naples, Italy; (R.V.T.); (S.B.); (L.C.)
| |
Collapse
|
13
|
Wang P, Ye Y, Chen Z, Li R, Hou G, Liu Z. PFKL promotes cell viability and glycolysis and inhibits cisplatin chemosensitivity of laryngeal squamous cell carcinoma. Biochem Biophys Res Commun 2024; 730:150366. [PMID: 38991254 DOI: 10.1016/j.bbrc.2024.150366] [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: 04/24/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024]
Abstract
Laryngeal squamous cell carcinoma (LSCC) with a high incidence and mortality rate, has a serious impact worldwide. Phosphofructokinase-1 liver type (PFKL) is a major enzyme in glycolysis progress, but its role in modulating tumorigenesis and cisplatin (DDP) chemosensitivity of LSCC was still unclear. The mRNA and protein levels of PFKL were detected by qRT-PCR and immunohistochemical assay. Cell Counting Kit-8 assay and flow cytometry were carried out to observe cell viability, as well as apoptosis and mitochondrial reactive oxygen species (mito-ROS). Extracellular acidification rate and lactate content were measured using extracellular flux analysis and lactate assay kit. Immunofluorescent staining was used to evaluate the expression of γ-H2AX foci. DNA damage was detected via single-cell gel electrophoresis. Western blotting was introduced to evaluate the protein level of PFKL, LDHA, γ-H2AX, cleaved PARP, H3K27ac, and H3K9ac. Mice xenograft model of LSCC was built for in vivo validation. The PFKL expression was significantly increased in LSCC and associated with poor survival of LSCC patients. Knockdown of PFKL in LSCC cells significantly inhibited cell viability, ECAR, lactate content, and LDHA expression, but promoted mito-ROS level. Furthermore, knockdown of PFKL enhanced response of LSCC cells to DDP by increasing DDP-induced apoptosis, promoting DDP-induced mito-ROS level, γ-H2AX foci, tail DNA, and the expression of γ-H2AX and cleaved PARP. However, the overexpression of PFKL in LSCC cells had opposite experimental results. Nude mice tumor formation experiment proved that downregulation of PFKL significantly enhanced response of cells to DDP, demonstrated by reduced tumor volume, weight and increased TUNEL-positive cells. Suppression of CBP/EP300-mediated PFKL transcription inhibited cell viability and glycolysis and promoted mito-ROS in LSCC. PFKL promotes cell viability and DNA damage repair in DDP-treated LSCC through regulation of glycolysis pathway.
Collapse
Affiliation(s)
- Peng Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361000, China.
| | - Yixian Ye
- Department of Otorhinolaryngology, Zhongshan Hospital Xiamen Branch, Fudan University, Xiamen, 361000, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361000, China
| | - Zhaoyue Chen
- Department of Otorhinolaryngology, Zhongshan Hospital Xiamen Branch, Fudan University, Xiamen, 361000, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361000, China
| | - Ruilong Li
- Department of Otorhinolaryngology, Zhongshan Hospital Xiamen Branch, Fudan University, Xiamen, 361000, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361000, China
| | - Guanghui Hou
- Department of Otorhinolaryngology, Zhongshan Hospital Xiamen Branch, Fudan University, Xiamen, 361000, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361000, China
| | - Zheng Liu
- Department of Otorhinolaryngology, Zhongshan Hospital Xiamen Branch, Fudan University, Xiamen, 361000, China; Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361000, China
| |
Collapse
|
14
|
Song K, Ma C, Maswikiti EP, Gu B, Wang B, Wang N, Jiang P, Chen H. Downregulation of ALDH5A1 suppresses cisplatin resistance in esophageal squamous cell carcinoma by regulating ferroptosis signaling pathways. Mol Carcinog 2024; 63:1892-1906. [PMID: 38923019 DOI: 10.1002/mc.23778] [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: 02/22/2024] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
This study explores the specific role and underlying mechanisms of ALDH5A1 in the chemoresistance of esophageal squamous cell carcinoma (ESCC). The levels of cleaved caspase-3, 4-hydroxynonenal (4-HNE), intracellular Fe2+, and lipid reactive oxygen species (ROS) were evaluated via immunofluorescence. Cell viability and migration were quantified using cell counting kit-8 assays and wound healing assays, respectively. Flow cytometry was utilized to analyze cell apoptosis and ROS production. The concentrations of malondialdehyde (MDA) and reduced glutathione were determined by enzyme-linked immunosorbent assay. Proteome profiling was performed using data-independent acquisition. Additionally, a xenograft mouse model of ESCC was established to investigate the relationship between ALDH5A1 expression and the cisplatin (DDP)-resistance mechanism in vivo. ALDH5A1 is overexpressed in both ESCC patients and ESCC/DDP cells. Silencing of ALDH5A1 significantly enhances the inhibitory effects of DDP treatment on the viability and migration of KYSE30/DDP and KYSE150/DDP cells and promotes apoptosis. Furthermore, it intensifies DDP's suppressive effects on tumor volume and weight in nude mice. Gene ontology biological process analysis has shown that ferroptosis plays a crucial role in both KYSE30/DDP cells and KYSE30/DDP cells transfected with si-ALDH5A1. Our in vitro and in vivo experiments demonstrate that DDP treatment promotes the accumulation of ROS, lipid ROS, MDA, LPO, and intracellular Fe2+ content, increases the levels of proteins that promote ferroptosis (ACSL4 and FTH1), and decreases the expression of anti-ferroptosis proteins (SLC7A11, FTL, and GPX4). Silencing of ALDH5A1 further amplifies the regulatory effects of DDP both in vitro and in vivo. ALDH5A1 potentially acts as an oncogene in ESCC chemoresistance. Silencing of ALDH5A1 can reduce DDP resistance in ESCC through promoting ferroptosis signaling pathways. These findings suggest a promising strategy for the treatment of ESCC in clinical practice.
Collapse
Affiliation(s)
- Kewei Song
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Public Health, Jining No. 1 People's Hospital, Jining, China
| | - Chenhui Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | | | - Baohong Gu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Bofang Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Na Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining No. 1 People's Hospital, Jining, China
| | - Hao Chen
- Department of Tumor Surgery, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, China
| |
Collapse
|
15
|
Pervushin NV, Yapryntseva MA, Panteleev MA, Zhivotovsky B, Kopeina GS. Cisplatin Resistance and Metabolism: Simplification of Complexity. Cancers (Basel) 2024; 16:3082. [PMID: 39272940 PMCID: PMC11394643 DOI: 10.3390/cancers16173082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Cisplatin is one of the most well-known anti-cancer drugs and has demonstrated efficacy against numerous tumor types for many decades. However, a key challenge with cisplatin, as with any chemotherapeutic agent, is the development of resistance with a resultant loss of efficacy. This resistance is often associated with metabolic alterations that allow insensitive cells to divide and survive under treatment. These adaptations could vary greatly among different tumor types and may seem questionable and incomprehensible at first glance. Here we discuss the disturbances in glucose, lipid, and amino acid metabolism in cisplatin-resistant cells as well as the roles of ferroptosis and autophagy in acquiring this type of drug intolerance.
Collapse
Affiliation(s)
- Nikolay V Pervushin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Maria A Yapryntseva
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Mikhail A Panteleev
- Department of Medical Physics, Physics Faculty, Lomonosov Moscow State University, 119991 Moscow, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia
| | - Boris Zhivotovsky
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, P.O. Box 210, 17177 Stockholm, Sweden
| | - Gelina S Kopeina
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
16
|
Lin Y, Lou X, Li S, Cai W, Che T. Identification and Validation of Immune Implication of R-Spondin 1 and an R-Spondin 1-Related Prognostic Signature in Esophagus Cancer. Int J Genomics 2024; 2024:7974277. [PMID: 38962149 PMCID: PMC11222003 DOI: 10.1155/2024/7974277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 07/05/2024] Open
Abstract
R-spondin 1 (RSPO1), which encodes a secretory-activating protein, is a promising therapeutic target for various tumors. The aim of this study was to establish a robust RSPO1-related signature specific to esophageal cancer (ESCA). Our comprehensive study involved meticulous analysis of RSPO1 expression in ESCA tissues and validation across ESCA cell lines and clinical samples using The Cancer Genome Atlas (TCGA) and GTEx databases. Using TCGA-ESCA dataset, we employed single-sample gene set enrichment analysis (ssGSEA) to elucidate the complex interaction between RSPO1 expression and the abundance of 22 specific immune cell types infiltrating ESCA. The biological significance of RSPO1 was further elucidated using KEGG, GO, and GSEA, demonstrating its relevance to pivotal tumor and immune pathways. This study culminated in the construction of prognostic nomograms enriched by calibration curves, facilitating the projection of individual survival probabilities at intervals of one, three, and five years. A substantial decrease in RSPO1 expression was observed within ESCA tissues and cell lines compared to their normal esophageal counterparts, and a significant decrease in the proportion of activated dendritic cells was evident within ESCA, accompanied by an augmented presence of macrophages and naive B cells relative to normal tissue. GSEA and KEGG analyses showed that RSPO1 was associated with tumor and immune pathways. Additionally, an independent prognostic risk score based on the RSPO1-related gene signature was developed and validated for patients with ESCA. Finally, RT-qPCR and western blotting were performed to confirm RSPO1 expression in normal and ESCA cell lines and tissue samples. In summary, our investigation underscores the pivotal role of RSPO1 in orchestrating tumor immunity and proposes RSPO1 as a prospective target for immunotherapeutic interventions in ESCA. Furthermore, the intricate profile of the two RSPO1-related genes has emerged as a promising predictive biomarker with notable potential for application in ESCA.
Collapse
Affiliation(s)
- Yuansheng Lin
- Department of Intensive Care UnitSuzhou HospitalAffiliated Hospital of Medical SchoolNanjing University, Suzhou 215000, China
| | - Xinqi Lou
- Institute of Clinical Medicine ResearchSuzhou HospitalAffiliated Hospital of Medical SchoolNanjing University, Suzhou 215000, China
| | - Shengjun Li
- Department of Emergency and Critical Care MedicineSuzhou HospitalAffiliated Hospital of Medical SchoolNanjing University, Suzhou 215000, China
| | - Wei Cai
- Department of Intensive Care UnitSuzhou HospitalAffiliated Hospital of Medical SchoolNanjing University, Suzhou 215000, China
| | - Tuanjie Che
- The Open Project of Key Laboratory of Functional Genomics and Molecular Diagnosis of Gansu Province, Lanzhou 730000, China
| |
Collapse
|