1
|
Zhu W, Zhang Y, Yang L, Chen L, Chen C, Shi Q, Xu Z. Construction of a lung adenocarcinoma prognostic model based on KEAP1/NRF2/HO‑1 mutation‑mediated upregulated genes and bioinformatic analysis. Oncol Lett 2025; 29:155. [PMID: 39911153 PMCID: PMC11795234 DOI: 10.3892/ol.2025.14902] [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: 08/27/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a prevalent malignant tumor of the respiratory tract. The Kelch like ECH associated protein 1 (KEAP1)/nuclear factor erythroid 2-related factor 2 (NRF2)/heme oxygenase 1 (HO-1) axis serves a pivotal role in the occurrence and progression of LUAD. The present study aimed to identify specific genes regulated by mutations of the KEAP1/NRF2/HO-1 axis and to investigate their prognostic potential in LUAD, as well as their association with the tumor microenvironment. Immunohistochemistry was performed to assess the expression levels of KEAP1, NRF2 and HO-1 in LUAD tissues and to evaluate their association with clinical pathology. Sequencing data and clinical information were obtained from The Cancer Genome Atlas (TCGA)-LUAD and Gene Expression Omnibus (GSE68465) databases, whilst mutation information was sourced from the cBio Cancer Genomics Portal website. The R package 'limma' and Venn diagram were utilized to identify upregulated differentially expressed genes. Subsequently, a prognostic model was constructed using univariate Cox regression analysis and 101 machine learning methods. A nomogram of the prognostic model was generated to assess its efficacy in evaluating survival among patients with LUAD. The 'ImmuCellAI' and 'oncoPredict' R packages were used to compare and evaluate differences in immune cell infiltration and immunotherapy between high- and low-risk groups, as well as the sensitivity of LUAD to chemotherapy drugs. Compared with the group with negative expression, the results revealed that the group with positive expression of NRF2 and HO-1 exhibited advanced tumor, lymph node and clinical stages and a worse prognosis. A predictive model incorporating four genes (kynureninase, serpin family B member 5, insulin like 4 and γ-aminobutyric acid type A receptor subunit α3) was constructed based on KEAP1/NRF2/HO-1 mutation-mediated upregulated genes (KNHMUGs). Risk score was an independent prognostic factor for patients with LUAD (hazard ratio, 1.038; 95% confidence interval, 1.034-1.043; P<0.001). A nomogram was developed to predict the prognosis of patients with LUAD, which was validated as a reliable prognostic tool. The low-risk group exhibited higher immune cell infiltration, including CD4+ T, CD8+ T, natural killer (NK) and NKT cells, compared with the high-risk group. In addition, it demonstrated increased expression levels of immune checkpoint inhibitory genes such as cytotoxic T-lymphocyte associated protein 4, T cell immunoreceptor with Ig and ITIM domains, hepatitis A virus cellular receptor 2 and B and T lymphocyte associated protein. Moreover, it displayed enhanced sensitivity to immunotherapy. Drug sensitivity analysis revealed that the high-risk group exhibited increased sensitivity towards vinblastine, docetaxel and cisplatin, whereas the low-risk group showed increased sensitivity to BMS_754807, SB505124_1194 and JQ1_2172. In conclusion, a KNHMUGs-based gene signature was constructed in the present study, which holds promise as a biomarker for evaluating patient prognosis and guiding treatment by effectively assessing immunotherapy response and chemotherapy sensitivity in patients with LUAD.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Pathology, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| | - Ye Zhang
- Department of Pathology, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| | - Lingyun Yang
- Department of Renal and Rheumatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, Jiangsu 214000, P.R. China
| | - Lu Chen
- Department of Pathology, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| | - Chaobo Chen
- Department of General Surgery, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
- Department of Hepatobiliary and Transplantation Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210000, P.R. China
| | - Qifeng Shi
- Department of Pathology, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| | - Zipeng Xu
- Department of General Surgery, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| |
Collapse
|
2
|
Rostami Ravari N, Sadri F, Mahdiabadi MA, Mohammadi Y, Ourang Z, Rezaei Z. Ferroptosis and noncoding RNAs: exploring mechanisms in lung cancer treatment. Front Cell Dev Biol 2025; 13:1522873. [PMID: 40078365 PMCID: PMC11897296 DOI: 10.3389/fcell.2025.1522873] [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: 11/05/2024] [Accepted: 01/31/2025] [Indexed: 03/14/2025] Open
Abstract
Lung cancer (LC) is a highly prevalent and deadly type of cancer characterized by intricate molecular pathways that drive tumor development, metastasis, and resistance to conventional treatments. Recently, ferroptosis, a controlled mechanism of cell death instigated by iron-dependent lipid peroxidation, has gained attention for its role in LC progression and treatment. Noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are emerging as key modulators of ferroptosis, significantly influencing LC biology. This review explores how ncRNAs control ferroptotic pathways and affect tumor growth, metastasis, and therapy resistance in LC. By understanding the dual functions of ncRNAs in both activating and inhibiting ferroptosis, we aim to uncover new therapeutic targets and strategies for LC. These insights provide a promising direction for the development of ncRNA-based treatments designed to induce ferroptosis, potentially improving therapeutic outcomes for patients with LC.
Collapse
Affiliation(s)
- Nadi Rostami Ravari
- Department of Animal Science Researches, Agriculture and Natural Resources Education and Research Center of Kerman, Agriculture and Natural Resources Education and Research Organization (AREEO), Kerman, Iran
| | - Farzad Sadri
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Geriatric Health Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Ali Mahdiabadi
- Department of Internal Medicine, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Yaser Mohammadi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Ourang
- Department of Biochemistry, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Zohreh Rezaei
- Geriatric Health Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran
| |
Collapse
|
3
|
Zhang X, Wang R, Zhang X, Yang Y, Tian R. Ferroptosis related CPT1A and GDF15 gene polymorphisms are risk factors for lung adenocarcinoma: A case-control study. Gene 2025; 933:149002. [PMID: 39401734 DOI: 10.1016/j.gene.2024.149002] [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: 05/28/2024] [Revised: 09/28/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Ferroptosis is not only a consequence of inflammation, but also a dynamic process. Recent bioinformatics analysis suggests that ferroptosis related genes might be associated with lung adenocarcinoma (LUAD). CPT1A and GDF15 are critical for the process of ferroptosis and development of inflammation; however, little study focused on the mutation level of these genes in patients with LUAD. METHODS The candidate SNPs in CPT1A and GDF15 were genotyped in 320 pairs of LUAD patients and controls using Mass ARRAY platform. Moreover, the different expression of CPT1A and GDF15 in LUAD cases and healthy controls were validated by qRT-PCR and ELISA. RESULTS The rs80356779 G > A, rs3019594 C > T, rs888663 T > G and rs4808793 G > C all exhibited an increased risk of the disease (p < 0.05). Moreover, the rs80356779-GA, rs3019594-TT, rs888663-TG and rs4808793-CC genotypes were all related to different levels of increase in LUAD risk (p < 0.05). Genetic model results showed that rs80356779 G > A, rs888663 T > G and rs4808793 G > C were associated with LUAD susceptibility under dominant and additive models (p < 0.05), while rs3019594 C > T was correlated with an elevated risk of the disease in all three models (p < 0.05). Additionally, patients with rs80356779 G > A and rs3019594 C > T exhibited lower expression and serum concentration of CPT1A compared with wile types, and patients with rs888663 T > G and rs4808793 G > C exhibited higher serum and expression level of GDF15. CONCLUSION The results provided new clues for the role of ferroptosis in LUAD and new potential targets for screening of susceptible population.
Collapse
Affiliation(s)
- Xing Zhang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Rong Wang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xia Zhang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Yanli Yang
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Ruifen Tian
- Respiratory department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Science/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
4
|
Chen S, Jiang Y, Xie G, Wu P, Zhu J. Comprehensive analysis of ferroptosis-related genes reveals potential therapeutic targets in osteoporosis patients: a computational analysis and in vitro experiments. Front Genet 2025; 15:1522809. [PMID: 39867575 PMCID: PMC11757248 DOI: 10.3389/fgene.2024.1522809] [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: 11/05/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Background Ferroptosis-related genes have been reported to play important roles in many diseases, but their molecular mechanisms in osteoporosis have not been elucidated. Methods Based on two independent GEO datasets (GSE35956 and GSE35958), and GSE35959 as the validation dataset, we comprehensively elucidated the pathological mechanism of ferroptosis-related genes in osteoporosis by GO analyses, KEGG analyses and a PPI network. Then, We used Western Blot (WB) and Quantitative real-time polymerase chain reaction (qPCR) to verify the expression level of KMT2D, a ferroptosis-related hub gene, in clinical samples. Subsequently, we predicted the upstream miRNA of KMT2D gene and analyzed the mechanism of KMT2D in osteoporosis, the potential prognostic value and its immune invasion of KMT2D in pan-cancer. Results This study identified KMT2D and MYCN, TP63, RELA, SOX2, and CDKN1A as key ferroptosis-related genes in osteoporotic cell aging. The independent dataset validated that the expression level of KMT2D was significantly upregulated in osteoporosis samples. The experimental verification results of qPCR and WB indicate that KMT2D is highly expressed in patients with osteoporosis. Further analysis revealed that the hsa-miR-204-5p-KMT2D axis may play an important role in the aging of osteoporotic cells. The analysis of KMT2D reveals that KMT2D may mainly play a role in the aging of osteoporotic cells through epigenetics and the value in pan-cancer. Conclusion The study provides a theoretical basis for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Sihui Chen
- Department of Orthopedics, First Hospital of Jiaxing, Jiaxing, China
- College of Medicine, Jiaxing University, Jiaxing, China
| | - Yi Jiang
- Department of Orthopedics, First Hospital of Jiaxing, Jiaxing, China
- College of Medicine, Jiaxing University, Jiaxing, China
| | - Guoqin Xie
- Department of Orthopedics, First Hospital of Jiaxing, Jiaxing, China
- College of Medicine, Jiaxing University, Jiaxing, China
| | - Peng Wu
- Department of Orthopedics, First Hospital of Jiaxing, Jiaxing, China
- College of Medicine, Jiaxing University, Jiaxing, China
| | - Jinyu Zhu
- Department of Orthopedics, First Hospital of Jiaxing, Jiaxing, China
- College of Medicine, Jiaxing University, Jiaxing, China
| |
Collapse
|
5
|
Yang Y, Yu S, Liu W, Zhuo Y, Qu C, Zeng Y. Ferroptosis-related signaling pathways in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:1. [PMID: 39935430 PMCID: PMC11813627 DOI: 10.20517/cdr.2024.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
Ferroptosis is an iron-dependent form of programmed cell death induced by lipid peroxidation. This process is regulated by signaling pathways associated with redox balance, iron metabolism, and lipid metabolism. Cancer cells' increased iron demand makes them especially susceptible to ferroptosis, significantly influencing cancer development, therapeutic response, and metastasis. Recent findings indicate that cancer cells can evade ferroptosis by downregulating key signaling pathways related to this process, contributing to drug resistance. This underscores the possibility of modulating ferroptosis as an approach to counteract drug resistance and enhance therapeutic efficacy. This review outlines the signaling pathways involved in ferroptosis and their interactions with cancer-related signaling pathways. We also highlight the current understanding of ferroptosis in cancer drug resistance, offering insights into how targeting ferroptosis can provide novel therapeutic approaches for drug-resistant cancers. Finally, we explore the potential of ferroptosis-inducing compounds and examine the challenges and opportunities for drug development in this evolving field.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Simin Yu
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
- Department of Urology, Innovation Institute for Integration of Medicine and Engineering, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wanyao Liu
- XiangYa School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Yi Zhuo
- First Clinical Department of Changsha Medical University, Changsha 410219, Hunan, China
| | - Chunrun Qu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
6
|
Li X, Zhang F, Ke Y, Liu H, Hu Z, Wei L. Differentiation of Acute Leukemia Cells Including Cells with MLL-AF4 Rearrangements Induced by Jiyuan Oridonin A. Recent Pat Anticancer Drug Discov 2025; 20:158-167. [PMID: 38204268 DOI: 10.2174/0115748928263141231204112640] [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: 06/03/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Chromosomal rearrangements involving the Mixed lineage leukemia (MLL) gene are observed in acute leukemia (AL) patients, which have poor prognosis, especially in infants. Hence, there is still a challenge to develop other effective agents to treat AL with MLL rearrangements (MLLr). MLL has been shown to rearrange with partner genes, of which the most frequently observed are AF4 and AF9. Moreover, AL is characterized by a differentiation blockage resulting in the accumulation of immature cells. An ent-kaurene diterpenoid compound, Jiyuan Oridonin A (JOA), has been shown to reduce the viability of AML cells by differentiation. METHODS We aimed to evaluate the effect of JOA on the growth and differentiation of AL cells (SEM, JURKAT and MV4-11) including cells with MLLr-AF4 by cell proliferation assay, colony formation assay, cell cycle analysis, cell apoptosis analysis, measurement of cell surface antigens and cell morphology, mRNA-sequencing analysis, quantitative Real-time PCR and Western blotting analysis. RESULTS Our findings demonstrated that the proliferation of AL cells including cells with MLLr-AF4 was significantly suppressed by JOA, which induced cell differentiation followed by G0/G1 cell cycle withdrawal. Moreover, JOA-mediated cell differentiation was likely due to activation of G-CSFR in MV4-11 cells. CONCLUSION Our results suggest that JOA may be considered a promising anti-leukemia compound to develop to surmount the differentiation block in AL patients.
Collapse
Affiliation(s)
- Xueming Li
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Fenglian Zhang
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Yu Ke
- School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Hongmin Liu
- School of Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Zhenbo Hu
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Shandong Weifang Medical University, China
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
7
|
Du Z, Shi Y, Tan J. Advances in integrating single-cell sequencing data to unravel the mechanism of ferroptosis in cancer. Brief Funct Genomics 2024; 23:713-725. [PMID: 38874174 DOI: 10.1093/bfgp/elae025] [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: 03/18/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
Ferroptosis, a commonly observed type of programmed cell death caused by abnormal metabolic and biochemical mechanisms, is frequently triggered by cellular stress. The occurrence of ferroptosis is predominantly linked to pathophysiological conditions due to the substantial impact of various metabolic pathways, including fatty acid metabolism and iron regulation, on cellular reactions to lipid peroxidation and ferroptosis. This mode of cell death serves as a fundamental factor in the development of numerous diseases, thereby presenting a range of therapeutic targets. Single-cell sequencing technology provides insights into the cellular and molecular characteristics of individual cells, as opposed to bulk sequencing, which provides data in a more generalized manner. Single-cell sequencing has found extensive application in the field of cancer research. This paper reviews the progress made in ferroptosis-associated cancer research using single-cell sequencing, including ferroptosis-associated pathways, immune checkpoints, biomarkers, and the identification of cell clusters associated with ferroptosis in tumors. In general, the utilization of single-cell sequencing technology has the potential to contribute significantly to the investigation of the mechanistic regulatory pathways linked to ferroptosis. Moreover, it can shed light on the intricate connection between ferroptosis and cancer. This technology holds great promise in advancing tumor-wide diagnosis, targeted therapy, and prognosis prediction.
Collapse
Affiliation(s)
- Zhaolan Du
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Yi Shi
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Jianjun Tan
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
- Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
8
|
Zhao P, Ren X, Zhang Z, Duan Z, Yang X, Jin J, Hu J. Blocking METTL3-mediated lncRNA FENDRR silence reverses cisplatin resistance of lung adenocarcinoma through activating TFRC-mediated ferroptosis pathway. J Mol Histol 2024; 56:21. [PMID: 39627631 DOI: 10.1007/s10735-024-10276-4] [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: 08/22/2024] [Accepted: 11/09/2024] [Indexed: 02/07/2025]
Abstract
Targeting ferroptosis pathway becomes a new solution for cisplatin (DDP) resistance in lung adenocarcinoma (LUAD), and further research is required to explore the molecular mechanisms underlying ferroptosis and DDP resistance, providing biotargets for LUAD treatment. In this study, DDP-sensitive A549 cells and DDP-resistant A549/DDP cells were treated with DDP, DDP sensitivity was detected through using CCK-8 method and colony formation assay, ferroptosis-related markers were determined through commercial kits, and the molecular regulatory mechanism was analyzed through methylated RNA immunoprecipitation, RNA pull-down, dual luciferase assay, quantitative real-time polymerase chain reaction and western blotting assay. Results showed that compared to A549 cells, FENDRR was downregulated in A549/DDP cells, and FENDRR increased iron content, labile iron pool, lipid peroxidation, LDH release and ROS levels, accelerating ferroptosis to promote DDP sensitivity. Interestingly, we found that METTL3-mediated N6-methyladenosine modification YTHDF2 dependently resulted in FENDRR degradation, and FENDRR overexpression elevated TFRC expression through sponging miR-761. Mechanistically, METTL3 inhibited the FENDRR/TFRC axis to alleviate DDP-induced ferroptosis, promoting DDP resistance in LUAD cells. Collectively, our findings identify a novel molecular regulatory mechanism in DDP resistance of LUAD, and suggest that FENDRR might be an attractive target for addressing DDP resistance.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Thoracic Surgery, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Congtai District, Handan, 056000, Hebei, China
| | - Xiaoguo Ren
- Oncology Department, Shexian Hospital, Handan, 056400, China
| | - Zhenchao Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Congtai District, Handan, 056000, Hebei, China
| | - Zhentao Duan
- Department of Thoracic Surgery, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Congtai District, Handan, 056000, Hebei, China
| | - Xiaogang Yang
- Department of Cardiac Surgery, Affiliated Hospital of Hebei Engineering University, Handan, 056000, China
| | - Jiatai Jin
- Department of Thoracic Surgery, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Congtai District, Handan, 056000, Hebei, China
| | - Jigang Hu
- Department of Thoracic Surgery, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Congtai District, Handan, 056000, Hebei, China.
| |
Collapse
|
9
|
Chen Y, Jiang Z, Li X. New insights into crosstalk between Nrf2 pathway and ferroptosis in lung disease. Cell Death Dis 2024; 15:841. [PMID: 39557840 PMCID: PMC11574213 DOI: 10.1038/s41419-024-07224-1] [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: 07/18/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
Ferroptosis is a distinctive process of cellular demise that is linked to amino acid metabolism, lipid oxidation, and iron oxidation. The ferroptosis cascade genes, which are closely associated with the onset of lung diseases, are among the regulatory targets of nuclear factor erythroid 2-related factor 2 (Nrf2). Although the regulation of ferroptosis is mostly mediated by Nrf2, the precise roles and underlying regulatory mechanisms of ferroptosis and Nrf2 in lung illness remain unclear. This review provides new insights from recent discoveries involving the modulation of Nrf2 and ferroptosis in a range of lung diseases. It also systematically describes regulatory mechanisms involving lipid peroxidation, intracellular antioxidant levels, ubiquitination of Nrf2, and expression of FSP1 and GPX4. Finally, it summarises active ingredients and drugs with potential for the treatment of lung diseases. With the overarching aim of expediting improvements in treatment, this review provides a reference for novel therapeutic mechanisms and offers suggestions for the development of new medications for a variety of lung disorders.
Collapse
Affiliation(s)
- Yonghu Chen
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China
| | - Zhe Jiang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China.
| | - Xuezheng Li
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, 133002, P. R. China.
| |
Collapse
|
10
|
Chen Z, Wang W, Hou J, Gao C, Song M, Zhao Z, Guan R, Chen J, Wu H, Abdul Razak SR, Han T, Zhang J, Wang L, Ahmad NH, Li X. NEDD4L contributes to ferroptosis and cell growth inhibition in esophageal squamous cell carcinoma by facilitating xCT ubiquitination. Cell Death Discov 2024; 10:473. [PMID: 39557844 PMCID: PMC11574128 DOI: 10.1038/s41420-024-02243-5] [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: 02/21/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
The oncogene xCT plays an indispensable role in tumor growth by protecting cancer cells from oxidative stress and ferroptosis. Emerging evidence indicated xCT function is tightly controlled by posttranslational modifications, especially ubiquitination. However, it still remains unclear what specific regulatory mechanism of xCT by ubiquitin ligases in human cancers. Here, we reported that NEDD4L, an E3 ubiquitin ligases, inhibited esophageal squamous cell carcinoma (ESCC) tumor growth and facilitated ferroptosis by ubiquitination of xCT. NEDD4L expression was declined in ESCC and was associated with tumor invasion, lymph node metastasis and distant metastasis. Silencing NEDD4L triggered ESCC tumor growth. Meanwhile, knock down of NEDD4L prevented the accumulation of ROS, elevated the level of GSH, reduced the content of MDA in ESCC cells, thereby inhibiting ferroptosis. Mechanistically, NEDD4L directly bound to the ∆CT domain of xCT through its WW and HECT domain. More importantly, NEDD4L promoted xCT degradation by facilitating its polyubiquitination in ESCC cells. Collectively, these findings suggest that NEDD4L is crucial in governing the stability of xCT and mediating ferroptosis in ESCC.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Weilong Wang
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Jinghan Hou
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Can Gao
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Meili Song
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Zijun Zhao
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Ruirui Guan
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Jingsheng Chen
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Huicheng Wu
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Siti Razila Abdul Razak
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia
| | - Tao Han
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Junbo Zhang
- Department of Surgery, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China
| | - Lidong Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, Henan Province, PR China
| | - Nor Hazwani Ahmad
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200 Kepala Batas, Bertam, Pulau Pinang, Malaysia.
| | - Xiumin Li
- Department of Gastroenterology, the First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, PR China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453000, Henan Province, PR China.
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, PR China.
| |
Collapse
|
11
|
Razi S, Khojini JY, Norioun H, Hayati MJ, Naseri N, Tajbaksh A, Gheibihayat SM. MicroRNA-mediated regulation of Ferroptosis: Implications for disease pathogenesis and therapeutic interventions. Cell Signal 2024; 125:111503. [PMID: 39510403 DOI: 10.1016/j.cellsig.2024.111503] [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: 07/03/2024] [Revised: 10/05/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Ferroptosis, a form of iron-dependent regulated cell death, is characterized by the accumulation of lipid peroxides and distinctive morphological features. Moreover, the reduction of intracellular antioxidant enzyme expression or activity, specifically glutathione peroxidase 4 (GPX4) results in activation of the endogenous pathway of ferroptosis. In this review, we aimed to explore the intricate interplay between microRNAs (miRNAs) and ferroptosis, shedding light on its implications in various disease pathologies. This review delves into the role of miRNAs in modulating key regulators of ferroptosis, including genes involved in iron metabolism, lipid peroxidation, and antioxidant defenses. Furthermore, the potential of targeting miRNAs for therapeutic interventions in ferroptosis-related diseases, such as cancer, neurodegenerative disorders, and ischemia/reperfusion injury, is highlighted.
Collapse
Affiliation(s)
- Shokufeh Razi
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamid Norioun
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Mohammad Javad Hayati
- Department of Medical Biotechnology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nasim Naseri
- Department of Animal Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Amir Tajbaksh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
12
|
Li Z, Xing J. Nuclear factor erythroid 2-related factor-mediated signaling alleviates ferroptosis during cerebral ischemia-reperfusion injury. Biomed Pharmacother 2024; 180:117513. [PMID: 39341075 DOI: 10.1016/j.biopha.2024.117513] [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: 07/25/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiac arrest (CA) is a significant challenge for emergency physicians worldwide and leads to increased morbidity and mortality rates. The poor prognosis of CA primarily stems from the complexity and irreversibility of cerebral ischemia-reperfusion injury (CIRI). Ferroptosis, a form of programmed cell death characterized by iron overload and lipid peroxidation, plays a crucial role in the progression and treatment of CIRI. In this review, we highlight the mechanisms of ferroptosis within the context of CIRI, focusing on its role as a key contributor to neuronal damage and dysfunction post-CA. We explore the crucial involvement of the nuclear factor erythroid 2-related factor (Nrf2)-mediated signaling pathway in modulating ferroptosis-associated processes during CIRI. Through comprehensive analysis of the regulatory role of Nrf2 in the cellular responses to oxidative stress, we highlight its potential as a therapeutic target for mitigating ferroptotic cell death and improving the neurological prognosis of patients experiencing CA. Furthermore, we discuss interventions targeting the Kelch-like ECH-associated protein 1/Nrf2/antioxidant response element pathway, including the use of traditional Chinese medicine and Western medicine, which demonstrate potential for attenuating ferroptosis and preserving neuronal function in CIRI. Owing to the limitations in the safety, specificity, and effectiveness of Nrf2-targeted drugs, as well as the technical difficulties and ethical constraints in obtaining the results related to the brain pathological examination of patients, most of the studies focusing on Nrf2-related regulation of ferroptosis in CIRI are still in the basic research stage. Overall, this review aims to provide a comprehensive understanding of the mechanisms underlying ferroptosis in CIRI, offering insights into novel therapeutics aimed at enhancing the clinical outcomes of patients with CA.
Collapse
Affiliation(s)
- Zheng Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
13
|
Shan G, Bian Y, Yao G, Liang J, Shi H, Hu Z, Zheng Z, Bi G, Fan H, Zhan C. Targeting ALDH2 to augment platinum-based chemosensitivity through ferroptosis in lung adenocarcinoma. Free Radic Biol Med 2024; 224:310-324. [PMID: 39216560 DOI: 10.1016/j.freeradbiomed.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Ferroptosis is a regulated cell death driven by iron-dependent lipid peroxidation and associated with drug resistance in lung adenocarcinoma (LUAD). It's found that aldehyde dehydrogenase 2 (ALDH2), which is highly mutated in East Asian populations, is correlated with response to chemotherapy in LUAD patients. The rs671 variant knock-in, downregulation, and pharmacological inhibition of ALDH2 render LUAD cells more vulnerable to ferroptosis inducers and platinum-based chemotherapy. ALDH2 inhibits ferroptosis through the detoxification of 4-hydroxynonenal and malondialdehyde, the product of lipid peroxidation, as well as the production of NADH at the same time. Besides, ALDH2 deficiency leads to elevated intracellular pH (pHi), thus inhibiting the ERK/CREB1/GPX4 axis. Interestingly, ALDH2 is also regulated by CREB1, and the ALDH2 enzyme activity was decreased with elevated pHi. What's more, the elevated pHi caused by impaired ALDH2 activity promotes the biosynthesis of lipid droplets to counteract ferroptosis. At last, the effect of ALDH2 on ferroptosis and chemosensitivity is confirmed in patient-derived organoids and xenograft models. Collectively, this study demonstrates that ALDH2 deficiency confers sensitivity to platinum through ferroptosis in LUAD, and targeting ALDH2 is a promising new strategy to enhance the sensitivity of platinum-based chemotherapy for the treatment of LUAD patients.
Collapse
Affiliation(s)
- Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangyu Yao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haochun Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaolin Zheng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hong Fan
- 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
|
14
|
Ding D, Shang W, Shi K, Ying J, Wang L, Chen Z, Zhang C. FTO/m6A mediates miR-138-5p maturation and regulates gefitinib resistance of lung adenocarcinoma cells by miR-138-5p/LCN2 axis. BMC Cancer 2024; 24:1270. [PMID: 39394098 PMCID: PMC11470737 DOI: 10.1186/s12885-024-13036-5] [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: 11/22/2023] [Accepted: 10/07/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Lung cancer (LC) occupies an important position in the lethality of cancer patients. Acquired resistance to gefitinib in lung adenocarcinoma (LUAD) seriously affects the therapeutic efficacy of LC. Thus, it is of major scientific and clinical significance to probe the mechanism of gefitinib resistance in LUAD for ameliorating the prognosis of patients. METHODS The expression of miRNAs in gefitinib-resistant LUAD cells was validated using qRT-PCR. Cell viability was assessed through CCK-8, whereas cell death was examined through PI staining. Changes in the ferroptosis process were evaluated by detecting the intracellular Glutathione (GSH), Malondialdehyde (MDA), and Reactive Oxygen Species (ROS) levels. Downstream targets of miR-138-5p were verified via luciferase reporter and RNA pull-down assays. RIP and qRT-PCR were employed to evaluate pri-miR-138-5p binding to DiGeorge critical region 8 (DGCR8) and the pri-miR-138-5p m6A modification level. Additionally, the impact of fat mass and obesity-associated protein (FTO) on LUAD gefitinib sensitivity was assessed in vivo by constructing a xenograft model. RESULTS We observed that miR-138-5p was notably diminished in gefitinib-resistant cells. Overexpression of miR-138-5p suppressed viability while facilitated cell death and intracellular ferroptosis in gefitinib-resistant cells. Moreover, lipocalin 2 (LCN2) was the downstream target of miR-138-5p. The biological functions of miR-138-5p on gefitinib-resistant cells was reversed by introduction of LCN2. FTO suppressed the binding of DGCR8 to pri-miR-138-5p through m6A modification, thereby restraining the processing of miR-138-5p. Meanwhile, silencing of FTO enhanced the sensitivity of LUAD to gefitinib treatment. CONCLUSION FTO suppressed the processing of miR-138-5p and then modulated the proliferation, death, and ferroptosis of gefitinib-resistant cells through the miR-138-5p/LCN2 pathway, which may put forward novel insights for clinically ameliorating the therapeutic effect of gefitinib in LUAD.
Collapse
Affiliation(s)
- Dongxiao Ding
- Department of Thoracic Surgery, People's Hospital of Beilun District, No.1288 Lushan East Road, Beilun District, Ningbo, Zhejiang, 3158000, China.
| | - Wenjun Shang
- Department of Thoracic Surgery, People's Hospital of Beilun District, No.1288 Lushan East Road, Beilun District, Ningbo, Zhejiang, 3158000, China
| | - Ke Shi
- Department of Thoracic Surgery, People's Hospital of Beilun District, No.1288 Lushan East Road, Beilun District, Ningbo, Zhejiang, 3158000, China
| | - Junjie Ying
- Department of Thoracic Surgery, People's Hospital of Beilun District, No.1288 Lushan East Road, Beilun District, Ningbo, Zhejiang, 3158000, China
| | - Li Wang
- Department of Thoracic Surgery, People's Hospital of Beilun District, No.1288 Lushan East Road, Beilun District, Ningbo, Zhejiang, 3158000, China
| | - Zhongjie Chen
- Department of Thoracic Surgery, People's Hospital of Beilun District, No.1288 Lushan East Road, Beilun District, Ningbo, Zhejiang, 3158000, China
| | - Chong Zhang
- Department of Thoracic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, No.79, Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
15
|
Elsakka EGE, Midan HM, Abulsoud AI, Fathi D, Abdelmaksoud NM, Abdel Mageed SS, Zaki MB, Abd-Elmawla MA, Rizk NI, Elrebehy MA, Abdelghany TM, Elesawy AE, Shahin RK, El Tabaa MM, Mohammed OA, Abdel-Reheim MA, Elballal MS, Doghish AS. Emerging insights: miRNA modulation of ferroptosis pathways in lung cancer. Exp Cell Res 2024; 442:114272. [PMID: 39362302 DOI: 10.1016/j.yexcr.2024.114272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/26/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
The newly discovered programmed iron-dependent necrosis, ferroptosis, is a novel pathway that is controlled by iron-dependent lipid peroxidation and cellular redox changes. It can be triggered intrinsically by low antioxidant enzyme activity or extrinsically by blocking amino acid transporters or activating iron transporters. The induction of ferroptosis involves the activation of specific proteins, suppression of transporters, and increased endoplasmic reticulum (ER) stress (a condition in which the ER, a crucial organelle involved in protein folding and processing, becomes overwhelmed by an accumulation of misfolded or unfolded proteins. This situation disrupts the normal functioning of the ER, leading to a cellular stress response known as the unfolded protein response), leading to lipid peroxidation byproduct accumulation and toxic reactive oxygen species (ROS), which are highly reactive molecules derived from diatomic oxygen and include various forms such as superoxide (O₂⁻), hydroxyl radicals (•OH), and hydrogen peroxide (H₂O₂). Ferroptosis is closely associated with signaling molecules in lung cancer, including epidermal growth factor receptor (EGFR), mitogen-activated protein kinase (MAPK), hypoxia-inducible factor 1-alpha (HIF-1α), and P53, and is regulated by epigenetic factors such as microRNAs (miRNAs). miRNAs are small non-coding RNA molecules that regulate gene expression by binding to target messenger RNAs (mRNAs), leading to translational repression or degradation. Several miRNAs have been found to modulate ferroptosis by targeting key genes involved in iron metabolism, lipid peroxidation, and antioxidant defense pathways. The research on ferroptosis has expanded to target its role in lung cancer treatment and resistance prevention. This review encapsulates the significance of ferroptosis in lung cancer. Understanding the mechanisms and implications of ferroptosis in lung cancer cells may lead to targeted therapies exploiting cancer cell vulnerabilities to ferroptosis Also, improving treatment outcomes, and overcoming resistance.
Collapse
Affiliation(s)
- Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| | - Heba M Midan
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Doaa Fathi
- Biochemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Nourhan M Abdelmaksoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Mohamed Bakr Zaki
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia, 32897, Egypt
| | - Mai A Abd-Elmawla
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nehal I Rizk
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Tamer M Abdelghany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, 3 Cairo-Belbeis Desert Road, P.O. Box 3020, El Salam, 11785, Cairo, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Reem K Shahin
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | | | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt; BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang, Republic of Korea
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
16
|
Ji T, Ye L, Xi E, Liu Y, Wang X, Wang S. Sinensetin Inhibits Angiogenesis in Lung Adenocarcinoma via the miR-374c-5p/VEGF-A/VEGFR-2/AKT Axis. Cell Biochem Biophys 2024; 82:2413-2425. [PMID: 39030333 DOI: 10.1007/s12013-024-01352-3] [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] [Accepted: 06/04/2024] [Indexed: 07/21/2024]
Abstract
Sinensetin is a product isolated from Orthosiphon aristatus, and its antitumor activities have been well established. This study focused on the role and mechanism of sinensetin in lung adenocarcinoma (LUAD). LUAD cells were treated with various concentrations of sinensetin. The proliferation, migration, invasion, and angiogenesis of LUAD cells were detected using colony formation, transwell, and tube formation assays, respectively. The protein levels of VEGF-A, VEGFR-2, and phosphorylated AKT (ser473) were measured by western blotting. The targeted relationship between VEGF-A and miR-374c-5p was verified by luciferase reporter assay. BALB/c nude mice inoculated with A549 cells were treated with sinensetin (40 mg/kg/day) by gavage for 21 days to investigate the effect of sinensetin on tumor growth and angiogenesis in vivo. We found that sinensetin reduced proliferation, migration, invasion, angiogenesis, and cancer stem characteristics of LUAD cells. Sinensetin also suppressed LUAD tumor growth and angiogenesis in vivo. Sinensetin downregulated VEGF-A expression in LUAD cells by enhancing miR-374c-5p expression. MiR-374c-5p inhibited the VEGF-A/VEGFR-2/AKT pathway in LUAD cells. The antitumor effect of sinensetin was reversed by overexpression of VEGF-A or inhibition of miR-374c-5p. Overall, sinensetin upregulates miR-374c-5p to inhibit the VEGF-A/VEGFR-2/AKT pathway, thereby exerting antitumor effect on LUAD.
Collapse
Affiliation(s)
- Tao Ji
- Department of Cardiothoracic Surgery, General Hospital of Central Theater Command, Wuhan, 430070, China
| | - Lin Ye
- Department of Cardiothoracic Surgery, General Hospital of Central Theater Command, Wuhan, 430070, China
| | - Erping Xi
- Department of Cardiothoracic Surgery, General Hospital of Central Theater Command, Wuhan, 430070, China
| | - Ying Liu
- Department of Cardiothoracic Surgery, General Hospital of Central Theater Command, Wuhan, 430070, China
| | - Xiumei Wang
- Department of Cardiothoracic Surgery, General Hospital of Central Theater Command, Wuhan, 430070, China
| | - Sha Wang
- Department of Dermatology, General Hospital of Central Theater Command, Wuhan, 430070, China.
| |
Collapse
|
17
|
Yong X, Zhang Y, Tang H, Hu H, Song R, Wu Q. CDKN2A inhibited ferroptosis through activating JAK2/STAT3 pathway to modulate cisplatin resistance in cervical squamous cell carcinoma. Anticancer Drugs 2024; 35:698-708. [PMID: 38748610 DOI: 10.1097/cad.0000000000001620] [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] [Indexed: 07/10/2024]
Abstract
Cervical squamous cell carcinoma (CESC) is a significant threat to women's health. Resistance to cisplatin (DDP), a common treatment, hinders the therapeutic efficacy. Understanding the molecular basis of DDP resistance in CESC is imperative. Cyclin-dependent kinase inhibitor 2A (CDKN2A) expression was evaluated through quantitative real-time-PCR and western blot in clinical samples from 30 CESC patients and human cervical epithelial cells and CESC cell lines (SiHa, C33A, and Caski). It was also evaluated through bioinformatics analysis in Timer, Ualcan, and GEPIA database. Cell viability was detected by CCK-8. Apoptosis was detected by Calcein AM/PI assay. Lipid reactive oxygen species (ROS), malondialdehyde, glutathione, Fe 2+ , and iron level were detected by kits. Protein level of JAK2, STAT3, p-JAK2, p-STAT3, ACSL4, GPX4, SLC7A11, and FTL were detected by western blot. In CESC, elevated CDKN2A expression was observed. Cisplatin exhibited a dual effect, inhibiting cell proliferation and inducing ferroptosis in CESC. CDKN2A knockdown in a cisplatin-resistant cell line suppressed proliferation and induced ferroptosis. Moreover, CDKN2A was identified as an inhibitor of erastin-induced ferroptosis. Additionally, targeting the JAK2/STAT3 pathway enhanced ferroptosis in cisplatin-resistant cells. CDKN2A could inhibit ferroptosis in CESC through activating JAK2/STAT3 pathway to modulate cisplatin resistance.
Collapse
Affiliation(s)
- Xiang Yong
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei City
- Department of Pathology, Anhui Wanbei Coal-Electricity Group General Hospital
- Department of Tumor Pathology, Suzhou City Key Laboratory of Tumor Pathology
| | - Yanling Zhang
- Department of Oncology, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou City, Anhui Province, China
| | - Heng Tang
- Department of Pathology, Anhui Wanbei Coal-Electricity Group General Hospital
- Department of Tumor Pathology, Suzhou City Key Laboratory of Tumor Pathology
| | - Huaiyuan Hu
- Department of Pathology, Anhui Wanbei Coal-Electricity Group General Hospital
- Department of Tumor Pathology, Suzhou City Key Laboratory of Tumor Pathology
| | - Rui Song
- Department of Pathology, Anhui Wanbei Coal-Electricity Group General Hospital
- Department of Tumor Pathology, Suzhou City Key Laboratory of Tumor Pathology
| | - Qiang Wu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei City
| |
Collapse
|
18
|
Wang H, Fleishman JS, Cheng S, Wang W, Wu F, Wang Y, Wang Y. Epigenetic modification of ferroptosis by non-coding RNAs in cancer drug resistance. Mol Cancer 2024; 23:177. [PMID: 39192329 DOI: 10.1186/s12943-024-02088-7] [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: 07/09/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
The development of drug resistance remains a major challenge in cancer treatment. Ferroptosis, a unique type of regulated cell death, plays a pivotal role in inhibiting tumour growth, presenting new opportunities in treating chemotherapeutic resistance. Accumulating studies indicate that epigenetic modifications by non-coding RNAs (ncRNA) can determine cancer cell vulnerability to ferroptosis. In this review, we first summarize the role of chemotherapeutic resistance in cancer growth/development. Then, we summarize the core molecular mechanisms of ferroptosis, its upstream epigenetic regulation, and its downstream effects on chemotherapeutic resistance. Finally, we review recent advances in understanding how ncRNAs regulate ferroptosis and from such modulate chemotherapeutic resistance. This review aims to enhance general understanding of the ncRNA-mediated epigenetic regulatory mechanisms which modulate ferroptosis, highlighting the ncRNA-ferroptosis axis as a key druggable target in overcoming chemotherapeutic resistance.
Collapse
Affiliation(s)
- Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sihang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Weixue Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China
| | - Fan Wu
- Department of Hepatobiliary Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| | - Yu Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, China.
| |
Collapse
|
19
|
Papatsirou M, Kontos CK, Ntanasis‐Stathopoulos I, Malandrakis P, Theodorakakou F, Liacos C, Mavrianou‐Koutsoukou N, Fotiou D, Migkou M, Gavriatopoulou M, Kastritis E, Dimopoulos MA, Scorilas A, Terpos E. ciRS-7 circular RNA overexpression in plasma cells is a promising molecular biomarker of unfavorable prognosis in multiple myeloma. EJHAEM 2024; 5:677-689. [PMID: 39157602 PMCID: PMC11327729 DOI: 10.1002/jha2.903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 08/20/2024]
Abstract
Several non-coding RNAs are known to be associated with the pathobiology and progression of multiple myeloma (MM). ciRS-7 (also known as CDR1-AS), a key oncogenic circular RNA (circRNA) that sponges miR-7-5p and other cancer-related microRNAs, was recently found to be downregulated in malignant plasma cells resistant to immunomodulatory drugs. Considering that various circRNAs have a strong potential as molecular biomarkers, we aimed to investigate the expression of ciRS-7 in plasma cell disorders, assess its prognostic importance in MM, and compare these findings with those of individuals with smoldering MM (SMM) and monoclonal gammopathy of unknown significance (MGUS). This study included 171 patients (110 newly diagnosed MM, 34 SMM, and 27 MGUS cases), from which bone marrow aspirate samples were collected for CD138+ plasma cell selection. Total RNA was reversely transcribed using random hexamer primers, and the expression levels of ciRS-7 were quantified using an in-house-developed protocol that includes pre-amplification and real-time quantitative polymerase chain reaction. ciRS-7 levels were found to significantly differ among CD138+ plasma cells of MM, SMM, and MGUS patients. ROC analysis indicated that ciRS-7 expression effectively distinguishes between MM and SMM patients. Moreover, high levels of ciRS-7 were associated with unfavorable prognosis in MM, independently of MM patients' age and Revised International Staging System stage. Additionally, in silico analysis predicted the binding of 85 microRNAs to ciRS-7. In conclusion, this study provides novel insights into the role of ciRS-7 as a promising molecular marker able to distinguish MM from SMM and predict prognosis in MM.
Collapse
Affiliation(s)
- Maria Papatsirou
- Department of Biochemistry and Molecular BiologyFaculty of BiologyNational and Kapodistrian University of AthensAthensGreece
| | - Christos K. Kontos
- Department of Biochemistry and Molecular BiologyFaculty of BiologyNational and Kapodistrian University of AthensAthensGreece
| | | | - Panagiotis Malandrakis
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Foteini Theodorakakou
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Christine‐Ivy Liacos
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Nefeli Mavrianou‐Koutsoukou
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Despina Fotiou
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Magdalini Migkou
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Maria Gavriatopoulou
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Efstathios Kastritis
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Meletios A. Dimopoulos
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular BiologyFaculty of BiologyNational and Kapodistrian University of AthensAthensGreece
| | - Evangelos Terpos
- Department of Clinical TherapeuticsSchool of MedicineNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
20
|
Gu X, Mu C, Zheng R, Zhang Z, Zhang Q, Liang T. The Cancer Antioxidant Regulation System in Therapeutic Resistance. Antioxidants (Basel) 2024; 13:778. [PMID: 39061847 PMCID: PMC11274344 DOI: 10.3390/antiox13070778] [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: 05/10/2024] [Revised: 06/15/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Antioxidants play a pivotal role in neutralizing reactive oxygen species (ROS), which are known to induce oxidative stress. In the context of cancer development, cancer cells adeptly maintain elevated levels of both ROS and antioxidants through a process termed "redox reprogramming". This balance optimizes the proliferative influence of ROS while simultaneously reducing the potential for ROS to cause damage to the cell. In some cases, the adapted antioxidant machinery can hamper the efficacy of treatments for neoplastic diseases, representing a significant facet of the resistance mechanisms observed in cancer therapy. In this review, we outline the contribution of antioxidant systems to therapeutic resistance. We detail the fundamental constituents of these systems, encompassing the central regulatory mechanisms involving transcription factors (of particular importance is the KEAP1/NRF2 signaling axis), the molecular effectors of antioxidants, and the auxiliary systems responsible for NADPH generation. Furthermore, we present recent clinical trials based on targeted antioxidant systems for the treatment of cancer, assessing the potential as well as challenges of this strategy in cancer therapy. Additionally, we summarize the pressing issues in the field, with the aim of illuminating a path toward the emergence of novel anticancer therapeutic approaches by orchestrating redox signaling.
Collapse
Affiliation(s)
- Xuanhao Gu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Chunyang Mu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Rujia Zheng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Zhe Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.G.); (C.M.); (Z.Z.)
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou 310003, China
- Zhejiang University Cancer Center, Hangzhou 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
21
|
Bian Y, Shan G, Liang J, Hu Z, Sui Q, Shi H, Wang Q, Bi G, Zhan C. Retinoic acid receptor alpha inhibits ferroptosis by promoting thioredoxin and protein phosphatase 1F in lung adenocarcinoma. Commun Biol 2024; 7:751. [PMID: 38902322 PMCID: PMC11190241 DOI: 10.1038/s42003-024-06452-7] [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: 08/01/2023] [Accepted: 06/14/2024] [Indexed: 06/22/2024] Open
Abstract
Ferroptosis is a recently discovered form of cell death that plays an important role in tumor growth and holds promise as a target for antitumor therapy. However, evidence in the regulation of ferroptosis in lung adenocarcinoma (LUAD) remains elusive. Here, we show that retinoic acid receptor alpha (RARA) is upregulated with the treatment of ferroptosis inducers (FINs). Pharmacological activation of RARA increases the resistance of LUAD to ferroptosis according to cell viability and lipid peroxidation assays, while RARA inhibitor or knockdown (KD) does the opposite. Through transcriptome sequencing in RARA-KD cells and chromatin immunoprecipitation (CHIP)-Seq data, we identify thioredoxin (TXN) and protein phosphatase 1 F (PPM1F) as downstream targets of RARA, both of which inhibit ferroptosis. We confirm that RARA binds to the promotor region of TXN and PPM1F and promotes their transcription by CHIP-qPCR and dual-luciferase assays. Overexpression of TXN and PPM1F reverses the effects of RARA knockdown on ferroptosis in vitro and vivo. Clinically, RARA knockdown or inhibitor increases cells' sensitivity to pemetrexed and cisplatin (CDDP). Immunohistochemistry (IHC) of LUAD from our cohort shows the same expression tendency of RARA and the downstream targets. Our study uncovers that RARA inhibits ferroptosis in LUAD by promoting TXN and PPM1F, and inhibiting RARA-TXN/PPM1F axis represents a promising strategy for improving the efficacy of FINs or chemotherapy in the treatment of LUAD patients.
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
| | - 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
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
22
|
Zhao G, Liang J, Zhang Y, Shan G, Bian Y, Gu J, Zhan C, Ge D. MNT inhibits lung adenocarcinoma ferroptosis and chemosensitivity by suppressing SAT1. Commun Biol 2024; 7:680. [PMID: 38831092 PMCID: PMC11148173 DOI: 10.1038/s42003-024-06373-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Ferroptosis, a type of iron-dependent non-apoptotic cell death, plays a vital role in both tumor proliferation and resistance to chemotherapy. Here, our study demonstrates that MAX's Next Tango (MNT), by involving itself in the spermidine/spermine N1-acetyltransferase 1 (SAT1)-related ferroptosis pathway, promotes the proliferation of lung adenocarcinoma (LUAD) cells and diminishes their sensitivity to chemotherapy. Initially, an RNA-sequence screen of LUAD cells treated with ferroptosis inducers (FINs) reveals a significant increase in MNT expression, suggesting a potential link between MNT and ferroptosis. Overexpression of MNT in LUAD cells hinders changes associated with ferroptosis. Moreover, the upregulation of MNT promotes cell proliferation and suppresses chemotherapy sensitivity, while the knockdown of MNT has the opposite effect. Through the intersection of ChIP-Seq and ferroptosis-associated gene sets, and validation by qPCR and western blot, SAT1 is identified as a potential target of MNT. Subsequently, we demonstrate that MNT binds to the promoter sequence of SAT1 and suppresses its transcription by ChIP-qPCR and dual luciferase assays. Restoration of SAT1 levels antagonizes the efficacy of MNT to inhibit ferroptosis and chemosensitivity and promote cell growth in vitro as well as in vivo. In the clinical context, MNT expression is elevated in LUAD and is inversely connected with SAT1 expression. High MNT expression is also associated with poor patient survival. Our research reveals that MNT inhibits ferroptosis, and impairing chemotherapy effectiveness of LUAD.
Collapse
Affiliation(s)
- Guangyin Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuchen Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Nejadi Orang F, Abdoli Shadbad M. Competing endogenous RNA networks and ferroptosis in cancer: novel therapeutic targets. Cell Death Dis 2024; 15:357. [PMID: 38778030 PMCID: PMC11111666 DOI: 10.1038/s41419-024-06732-4] [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: 12/12/2023] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
As a newly identified regulated cell death, ferroptosis is a metabolically driven process that relies on iron and is associated with polyunsaturated fatty acyl peroxidation, elevated levels of reactive oxygen species (ROS), and mitochondrial damage. This distinct regulated cell death is dysregulated in various cancers; activating ferroptosis in malignant cells increases cancer immunotherapy and chemoradiotherapy responses across different malignancies. Over the last decade, accumulating research has provided evidence of cross-talk between non-coding RNAs (ncRNAs) and competing endogenous RNA (ceRNA) networks and highlighted their significance in developing and progressing malignancies. Aside from pharmaceutical agents to regulate ferroptosis, recent studies have shed light on the potential of restoring dysregulated ferroptosis-related ceRNA networks in cancer treatment. The present study provides a comprehensive and up-to-date review of the ferroptosis significance, ferroptosis pathways, the role of ferroptosis in cancer immunotherapy and chemoradiotherapy, ceRNA biogenesis, and ferroptosis-regulating ceRNA networks in different cancers. The provided insights can offer the authorship with state-of-the-art findings and future perspectives regarding the ferroptosis and ferroptosis-related ceRNA networks and their implication in the treatment and determining the prognosis of affected patients.
Collapse
Affiliation(s)
| | - Mahdi Abdoli Shadbad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
24
|
Gong G, Wan Y, Liu Y, Zhang Z, Zheng Y. Ononin triggers ferroptosis-mediated disruption in the triple negative breast cancer both in vitro and in vivo. Int Immunopharmacol 2024; 132:111959. [PMID: 38554442 DOI: 10.1016/j.intimp.2024.111959] [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: 02/09/2024] [Revised: 03/16/2024] [Accepted: 03/26/2024] [Indexed: 04/01/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is difficult to treat due to a lack of targeted therapies. In this study, we aimed to investigate whether a natural flavonoid compound called ononin could be effective in treating TNBC by triggering ferroptosis in MDA-MB-231 and 4 T1 cell lines, and MDA-MB-231-xenograft nude mice model. Ononin inhibited TNBC through ferroptosis, which was determined by MTT assay, flow cytometry, RT-PCR, immunofluorescence, transmission electron microscopy, histological analysis, western blot and bioluminescence assay. Our results showed that treatment with ononin led to increased levels of malondialdehyde and reactive oxygen species and decreased activity of superoxide dismutase, which are indicatives of ferroptosis. We also found that ononin downregulated two key markers of ferroptosis, SLC7A11 and Nrf2, at both the transcriptional and translational level. Additionally, the administration of ononin resulted in a notable decrease in tumor size and weight in the mouse model. Furthermore, it was observed to enhance the rate of apoptosis in TNBC cells. Importantly, ononin did not induce any histological changes in the kidney, liver, and heart. Taken together, our findings suggest that ononin could be a promising therapeutic strategy for TNBC, and that it works by disrupting the Nrf2/SLC7A11 axis through ferroptosis. These results are encouraging and may lead to the development of new treatments for this challenging cancer subtype.
Collapse
Affiliation(s)
- Guowei Gong
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, Guangdong 519041, China; Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China.
| | - Yukai Wan
- Second Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Yaqun Liu
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Zhenxia Zhang
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China
| | - Yuzhong Zheng
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, Guangdong 521041, China.
| |
Collapse
|
25
|
Wu J, Li Z, Wu Y, Cui N. The crosstalk between exosomes and ferroptosis: a review. Cell Death Discov 2024; 10:170. [PMID: 38594265 PMCID: PMC11004161 DOI: 10.1038/s41420-024-01938-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/17/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024] Open
Abstract
Exosomes are a subtype of extracellular vesicles composed of bioactive molecules, including nucleic acids, proteins, and lipids. Exosomes are generated by the fusion of intracellular multivesicular bodies (MVBs) with the cell membrane and subsequently released into the extracellular space to participate in intercellular communication and diverse biological processes within target cells. As a crucial mediator, exosomes have been implicated in regulating ferroptosis-an iron-dependent programmed cell death characterized by lipid peroxide accumulation induced by reactive oxygen species. The involvement of exosomes in iron, lipid, and amino acid metabolism contributes to their regulatory role in specific mechanisms underlying how exosomes modulate ferroptosis, which remains incompletely understood, and some related studies are still preliminary. Therefore, targeting the regulation of ferroptosis by exosomes holds promise for future clinical treatment strategies across various diseases. This review aims to provide insights into the pathophysiology and mechanisms governing the interaction between exosomes and ferroptosis and their implications in disease development and treatment to serve as a reference for further research.
Collapse
Affiliation(s)
- Jiao Wu
- Oncology Department of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhongyu Li
- Department of Internal Medicine, Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yu Wu
- Oncology Department of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ning Cui
- Oncology Department of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
26
|
GUO X, WANG T, XIA J, ZENG H, SHI W. [Role of Ferroptosis in Non-small Cell Lung Cancer and Progress
of Traditional Chinese Medicine Intervention]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:216-230. [PMID: 38590196 PMCID: PMC11002191 DOI: 10.3779/j.issn.1009-3419.2024.101.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 04/10/2024]
Abstract
Non-small cell lung cancer (NSCLC) is one of the malignant tumors with high morbidity and mortality worldwide. Ferroptosis is a new type of programmed cell death caused by abnormal accumulation of iron-dependent reactive oxygen species (ROS) leading to lipid peroxidation. It involves the balance between iron metabolism, lipid metabolism, oxygen free radical reaction and lipid peroxidation. Recent studies have found that ferroptosis is closely related to the occurrence and development of NSCLC. Due to the emergence of chemotherapy resistance and radiotherapy resistance in the treatment of NSCLC, there is an urgent need to develop new effective drugs and treatment strategies. Traditional Chinese medicine has unique advantages in the prevention and treatment of NSCLC due to its multi-targets and minimal side effects. In this review, we summarize the mechanism of ferroptosis in NSCLC, and discuss the research status of active ingredients of traditional Chinese medicine, single-herb traditional Chinese medicine and Chinese herbal compounds in the intervention of NSCLC through ferroptosis, in order to provide a new theoretical basis for the research of ferroptosis pathway and the prevention and treatment of NSCLC by targeted ferroptosis of traditional Chinese medicine.
.
Collapse
|
27
|
Bi G, Liang J, Bian Y, Shan G, Huang Y, Lu T, Zhang H, Jin X, Chen Z, Zhao M, Fan H, Wang Q, Gan B, Zhan C. Polyamine-mediated ferroptosis amplification acts as a targetable vulnerability in cancer. Nat Commun 2024; 15:2461. [PMID: 38504107 PMCID: PMC10951362 DOI: 10.1038/s41467-024-46776-w] [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: 08/10/2023] [Accepted: 03/11/2024] [Indexed: 03/21/2024] Open
Abstract
Targeting ferroptosis, an iron-dependent form of regulated cell death triggered by the lethal overload of lipid peroxides, in cancer therapy is impeded by our limited understanding of the intersection of tumour's metabolic feature and ferroptosis vulnerability. In the present study, arginine is identified as a ferroptotic promoter using a metabolites library. This effect is mainly achieved through arginine's conversion to polyamines, which exerts their potent ferroptosis-promoting property in an H2O2-dependent manner. Notably, the expression of ornithine decarboxylase 1 (ODC1), the critical enzyme catalysing polyamine synthesis, is significantly activated by the ferroptosis signal--iron overload--through WNT/MYC signalling, as well as the subsequent elevated polyamine synthesis, thus forming a ferroptosis-iron overload-WNT/MYC-ODC1-polyamine-H2O2 positive feedback loop that amplifies ferroptosis. Meanwhile, we notice that ferroptotic cells release enhanced polyamine-containing extracellular vesicles into the microenvironment, thereby further sensitizing neighbouring cells to ferroptosis and accelerating the "spread" of ferroptosis in the tumour region. Besides, polyamine supplementation also sensitizes cancer cells or xenograft tumours to radiotherapy or chemotherapy through inducing ferroptosis. Considering that cancer cells are often characterized by elevated intracellular polyamine pools, our results indicate that polyamine metabolism exposes a targetable vulnerability to ferroptosis and represents an exciting opportunity for therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xing Jin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
28
|
Liang J, Bi G, Huang Y, Zhao G, Sui Q, Zhang H, Bian Y, Yin J, Wang Q, Chen Z, Zhan C. MAFF confers vulnerability to cisplatin-based and ionizing radiation treatments by modulating ferroptosis and cell cycle progression in lung adenocarcinoma. Drug Resist Updat 2024; 73:101057. [PMID: 38266355 DOI: 10.1016/j.drup.2024.101057] [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: 06/04/2023] [Revised: 12/22/2023] [Accepted: 01/16/2024] [Indexed: 01/26/2024]
Abstract
AIMS Lung cancer is the leading cause of cancer mortality and lung adenocarcinoma (LUAD) accounts for more than half of all lung cancer cases. Tumor elimination is mostly hindered by drug resistance and the mechanisms remain to be explored in LUAD. METHODS CRISPR screens in cell and murine models and single-cell RNA sequencing were conducted, which identified MAF bZIP transcription factor F (MAFF) as a critical factor regulating tumor growth and treatment resistance in LUAD. RNA and ChIP sequencing analyses were performed for transcriptional target expression and specific binding sites of MAFF. Functions of MAFF in inhibiting tumor growth and promoting cisplatin or irradiation efficacy were investigated using cellular and xenograft models. RESULTS Patients with lung adenocarcinoma and reduced MAFF expression had worse clinical outcomes. MAFF inhibited tumor cell proliferation by regulating the expression of SLC7A11, CDK6, and CDKN2C, promoting ferroptosis and preventing cell cycle progression from G1 to S. MAFF also conferred tumor cells vulnerable to cisplatin-based or ionizing radiation treatments. MAFF reduction was a final event in the acquisition of cisplatin resistance of LUAD cells. The intracellular cAMP/PKA/CREB1 pathway upregulated MAFF in response to cisplatin-based or ionizing radiation treatments. CONCLUSIONS MAFF suppresses tumor growth, and pharmacological agonists targeting MAFF may improve cisplatin or irradiation therapies for lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Guangyin Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Qihai Sui
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Huan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Jiacheng Yin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China.
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, China.
| |
Collapse
|
29
|
Hu T, Zeng C, Song Z, Liu Q, Chen S, Huang W, Ma Q, Li H. HNRNPA2B1 and HNRNPR stabilize ASCL1 in an m6A-dependent manner to promote neuroblastoma progression. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167050. [PMID: 38331110 DOI: 10.1016/j.bbadis.2024.167050] [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: 11/09/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
HNRNPA2B1 and HNRNPR stabilize ASCL1 mRNA in neuroblastoma, but whether their regulatory effects depend on m6A modification and whether their function involves ASCL1 remain unknown. This study investigated the m6A-dependent binding of HNRNPA2B1 and HNRNPR to ASCL1 and subsequent regulation, as well as the expression, clinical significance, and function of HNRNPA2B1 and HNRNPR in neuroblastoma. We revealed that METTL14 mediated ASCL1 m6A modification to stabilize ASCL1. HNRNPA2B1 and HNRNPR significantly enriched ASCL1 mRNA by binding to the 5' and 3' untranslated regions, respectively, and METTL14 knockdown reduced this enrichment. Mutations in m6A sites in the untranslated regions of ASCL1 mRNA considerably decreased probe capacity to engage HNRNPA2B1 and HNRNPR. HNRNPR interacts with IGF2BP1, and knocking down either impaired binding to ASCL1 mRNA. HNRNPA2B1 and HNRNPR knockdown suppressed neuroblastoma cell growth and invasion, while ASCL1 overexpression restored these effects. The high HNRNPA2B1 and HNRNPR expression in neuroblastoma correlated with ASCL1 expression. Thus, HNRNPA2B1 and HNRNPR bind and stabilize ASCL1 mRNA in an m6A-dependent manner to promote neuroblastoma progression. This study not only discovered a new mechanism underlying the high ASCL1 expression in neuroblastoma but also identified the HNRNPA2B1/HNRNPR/ASCL1 axis as a promising target for inhibiting neuroblastoma progression.
Collapse
Affiliation(s)
- Ting Hu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chong Zeng
- Department of Medicine, The Seventh Affiliated Hospital, Hengyang Medical School, University of South China, Changsha 410119, China
| | - Zhihao Song
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Si Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, China; Hunan Key Laboratory of Ophthalmology, Changsha 410008, China
| | - Wei Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Qianquan Ma
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing 100191, China.
| | - Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
30
|
Li Z, Li J, Wu Z, Zhu Y, Zhuo T, Nong J, Qian J, Peng H, Dai L, Wang Y, Chen M, Zeng X. Upregulation of POC1A in lung adenocarcinoma promotes tumour progression and predicts poor prognosis. J Cell Mol Med 2024; 28:e18135. [PMID: 38429900 PMCID: PMC10907829 DOI: 10.1111/jcmm.18135] [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: 09/29/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 03/03/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is characterized by a high incidence rate and mortality. Recently, POC1 centriolar protein A (POC1A) has emerged as a potential biomarker for various cancers, contributing to cancer onset and development. However, the association between POC1A and LUAD remains unexplored. We extracted The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) data sets to analyse the differential expression of POC1A and its relationship with clinical stage. Additionally, we performed diagnostic receiver operator characteristic (ROC) curve analysis and Kaplan-Meier (KM) survival analysis to assess the diagnostic and prognostic value of POC1A in LUAD. Furthermore, we investigated the correlation between POC1A expression and immune infiltration, tumour mutation burden (TMB), immune checkpoint expression and drug sensitivity. Finally, we verified POC1A expression using real-time quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC). Cell experiments were conducted to validate the effect of POC1A expression on the proliferation, migration and invasion of lung cancer cells. POC1A exhibited overexpression in most tumour tissues, and its overexpression in LUAD was significantly correlated with late-stage presentation and poor prognosis. The high POC1A expression group showed lower levels of immune infiltration but higher levels of immune checkpoint expression and TMB. Moreover, the high POC1A expression group demonstrated sensitivity to multiple drugs. In vitro experiments confirmed that POC1A knockdown led to decreased proliferation, migration, and invasion of lung cancer cells. Our findings suggest that POC1A may contribute to tumour development by modulating the cell cycle and immune cell infiltration. It also represents a potential therapeutic target and marker for the diagnosis and prognosis of LUAD.
Collapse
Affiliation(s)
- Zi‐Hao Li
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Jia‐Yi Li
- Geriatrics Department of Endocrinology and MetabolismThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Zuo‐Tao Wu
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Yong‐Jie Zhu
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Ting Zhuo
- Department of Respiratory MedicineThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Ju‐Sen Nong
- Department of Pediatric SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Jing Qian
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Hua‐Jian Peng
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Lei Dai
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Yong‐Yong Wang
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Ming‐Wu Chen
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Xiao‐Chun Zeng
- Department of Cardio‐Thoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| |
Collapse
|
31
|
Han Z, Luo W, Shen J, Xie F, Luo J, Yang X, Pang T, Lv Y, Li Y, Tang X, He J. Non-coding RNAs are involved in tumor cell death and affect tumorigenesis, progression, and treatment: a systematic review. Front Cell Dev Biol 2024; 12:1284934. [PMID: 38481525 PMCID: PMC10936223 DOI: 10.3389/fcell.2024.1284934] [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: 08/29/2023] [Accepted: 01/08/2024] [Indexed: 11/02/2024] Open
Abstract
Cell death is ubiquitous during development and throughout life and is a genetically determined active and ordered process that plays a crucial role in regulating homeostasis. Cell death includes regulated cell death and non-programmed cell death, and the common types of regulatory cell death are necrosis, apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis. Apoptosis, Necrosis and necroptosis are more common than autophagy, ferroptosis and pyroptosis among cell death. Non-coding RNAs are regulatory RNA molecules that do not encode proteins and include mainly microRNAs, long non-coding RNAs, and circular RNAs. Non-coding RNAs can act as oncogenes and tumor suppressor genes, with significant effects on tumor occurrence and development, and they can also regulate tumor cell autophagy, ferroptosis, and pyroptosis at the transcriptional or post-transcriptional level. This paper reviews the recent research progress on the effects of the non-coding RNAs involved in autophagy, ferroptosis, and pyroptosis on tumorigenesis, tumor development, and treatment, and looks forward to the future direction of this field, which will help to elucidate the molecular mechanisms of tumorigenesis and tumor development, as well as provide a new vision for the treatment of tumors.
Collapse
Affiliation(s)
- Zeping Han
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
- Rehabilitation Medicine Institute of Panyu District, Guangzhou, China
| | - Wenfeng Luo
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Jian Shen
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Fangmei Xie
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Jinggen Luo
- Department of General Surgery, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Xiang Yang
- Department of Gynaecology and Obstetrics, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Ting Pang
- Clinical Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Yubing Lv
- Clinical Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Yuguang Li
- He Xian Memorial Hospital, Southern Medical University, Guangzhou, China
| | - Xingkui Tang
- Department of General Surgery, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Jinhua He
- Central Laboratory, Guangzhou Panyu Central Hospital, Guangzhou, China
- Rehabilitation Medicine Institute of Panyu District, Guangzhou, China
| |
Collapse
|
32
|
Ma J, Yu P, Ma S, Li J, Wang Z, Hu K, Su X, Zhang B, Cheng S, Wang S. Bioinformatics and Integrative Experimental Method to Identifying and Validating Co-Expressed Ferroptosis-Related Genes in OA Articular Cartilage and Synovium. J Inflamm Res 2024; 17:957-980. [PMID: 38370466 PMCID: PMC10871044 DOI: 10.2147/jir.s434226] [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: 09/14/2023] [Accepted: 01/13/2024] [Indexed: 02/20/2024] Open
Abstract
Purpose Osteoarthritis (OA) is the most common joint disease worldwide and is the primary cause of disability and chronic pain in older adults.Ferroptosis is a type of programmed cell death characterized by aberrant iron metabolism and reactive oxygen species accumulation; however, its role in OA is not known. Methods To identify ferroptosis markers co-expressed in articular cartilage and synovium samples from patients with OA, in silico analysis was performed.Signature genes were analyzed and the results were evaluated using a ROC curve prediction model.The biological function, correlation between Signature genes, immune cell infiltration, and ceRNA network analyses were performed. Signature genes and ferroptosis phenotypes were verified through in vivo animal experiments and clinical samples. The expression levels of non-coding RNAs in samples from patients with OA were determined using qRT-PCR. ceRNA network analysis results were confirmed using dual-luciferase assays. Results JUN, ATF3, and CDKN1A were identified as OA- and ferroptosis-associated signature genes. GSEA analysis demonstrated an enrichment of these genes in immune and inflammatory responses, and amino acid metabolism. The CIBERSORT algorithm showed a negative correlation between T cells and these signature genes in the cartilage, and a positive correlation in the synovium. Moreover, RP5-894D12.5 and FAM95B1 regulated the expression of JUN, ATF3, and CDKN1A by competitively binding to miR-1972, miR-665, and miR-181a-2-3p. In vivo, GPX4 was downregulated in both OA cartilage and synovium; however, GPX4 and GSH were downregulated, while ferrous ions were upregulated in patient OA cartilage and synovium samples, indicating that ferroptosis was involved in the pathogenesis of OA. Furthermore, JUN, ATF3, and CDKN1A expression was downregulated in both mouse and human OA synovial and cartilage tissues. qRT-PCR demonstrated that miR-1972, RP5-894D12.5, and FAM95B1 were differentially expressed in OA tissues. Targeted interactions between miR-1972 and JUN, and a ceRNA regulatory mechanism between RP5-894D12.5, miR-1972, and JUN were confirmed by dual-luciferase assays. Conclusion This study identified JUN, ATF3, and CDKN1A as possible diagnostic biomarkers and therapeutic targets for joint synovitis and OA. Furthermore, our finding indicated that RP5-894D12.5/miR-1972/JUN was a potential ceRNA regulatory axis in OA, providing an insight into the connection between ferroptosis and OA.
Collapse
Affiliation(s)
- Jinxin Ma
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Peng Yu
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Shang Ma
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Jinjin Li
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Zhen Wang
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Kunpeng Hu
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Xinzhe Su
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Bei Zhang
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Shao Cheng
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Department of Arthropathy, Henan Province Hospital of Chinese Medicine (The Second Affiliated Hospital of Henan University of Chinese Medicine), Zhengzhou, People’s Republic of China
- School of Osteopathy, Henan Province Engineering Research Center of Basic and Clinical Research of Bone and Joint Repair in Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Shangzeng Wang
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Department of Arthropathy, Henan Province Hospital of Chinese Medicine (The Second Affiliated Hospital of Henan University of Chinese Medicine), Zhengzhou, People’s Republic of China
- School of Osteopathy, Henan Province Engineering Research Center of Basic and Clinical Research of Bone and Joint Repair in Chinese Medicine, Zhengzhou, People’s Republic of China
| |
Collapse
|
33
|
Nisar H, Labonté FM, Roggan MD, Schmitz C, Chevalier F, Konda B, Diegeler S, Baumstark-Khan C, Hellweg CE. Hypoxia Modulates Radiosensitivity and Response to Different Radiation Qualities in A549 Non-Small Cell Lung Cancer (NSCLC) Cells. Int J Mol Sci 2024; 25:1010. [PMID: 38256084 PMCID: PMC10816011 DOI: 10.3390/ijms25021010] [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: 11/23/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Hypoxia-induced radioresistance reduces the efficacy of radiotherapy for solid malignancies, including non-small cell lung cancer (NSCLC). Cellular hypoxia can confer radioresistance through cellular and tumor micro-environment adaptations. Until recently, studies evaluating radioresistance secondary to hypoxia were designed to maintain cellular hypoxia only before and during irradiation, while any handling of post-irradiated cells was carried out in standard oxic conditions due to the unavailability of hypoxia workstations. This limited the possibility of simulating in vivo or clinical conditions in vitro. The presence of molecular oxygen is more important for the radiotoxicity of low-linear energy transfer (LET) radiation (e.g., X-rays) than that of high-LET carbon (12C) ions. The mechanisms responsible for 12C ions' potential to overcome hypoxia-induced radioresistance are currently not fully understood. Therefore, the radioresistance of hypoxic A549 NSCLC cells following exposure to X-rays or 12C ions was investigated along with cell cycle progression and gene expression by maintaining hypoxia before, during and after irradiation. A549 cells were incubated under normoxia (20% O2) or hypoxia (1% O2) for 48 h and then irradiated with X-rays (200 kV) or 12C ions (35 MeV/n, LET ~75 keV/µm). Cell survival was evaluated using colony-forming ability (CFA) assays immediately or 24 h after irradiation (late plating). DNA double-strand breaks (DSBs) were analyzed using γH2AX immunofluorescence microscopy. Cell cycle progression was determined by flow cytometry of 4',6-diamidino-2-phenylindole-stained cells. The global transcription profile post-irradiation was evaluated by RNA sequencing. When hypoxia was maintained before, during and after irradiation, hypoxia-induced radioresistance was observed only in late plating CFA experiments. The killing efficiency of 12C ions was much higher than that of X-rays. Cell survival under hypoxia was affected more strongly by the timepoint of plating in the case of X-rays compared to 12C ions. Cell cycle arrest following irradiation under hypoxia was less pronounced but more prolonged. DSB induction and resolution following irradiation were not significantly different under normoxia and hypoxia. Gene expression response to irradiation primarily comprised cell cycle regulation for both radiation qualities and oxygen conditions. Several PI3K target genes involved in cell migration and cell motility were differentially upregulated in hypoxic cells. Hypoxia-induced radioresistance may be linked to altered cell cycle response to irradiation and PI3K-mediated changes in cell motility and migration in A549 cells rather than less DNA damage or faster repair.
Collapse
Affiliation(s)
- Hasan Nisar
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Department of Medical Sciences, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad 44000, Pakistan
| | - Frederik M. Labonté
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Marie Denise Roggan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Claudia Schmitz
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - François Chevalier
- UMR6252 CIMAP, CEA-CNRS-ENSICAEN-University of Caen Normandy, 14000 Caen, France;
| | - Bikash Konda
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - Sebastian Diegeler
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christa Baumstark-Khan
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| | - Christine E. Hellweg
- Department of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147 Cologne, Germany; (H.N.); (F.M.L.); (M.D.R.); (C.S.); (B.K.); (S.D.); (C.B.-K.)
| |
Collapse
|
34
|
Zou Q, Tang B, Chen X, Zhang C, Huang Y. STK11 (LKB1) mutation suppresses ferroptosis in lung adenocarcinoma by facilitating monounsaturated fatty acid synthesis. Open Med (Wars) 2024; 19:20230845. [PMID: 38205151 PMCID: PMC10775415 DOI: 10.1515/med-2023-0845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/22/2023] [Accepted: 10/22/2023] [Indexed: 01/12/2024] Open
Abstract
Serine/threonine kinase 11 (STK11), a tumor suppressor gene, exhibits frequent mutations in lung adenocarcinoma (LUAD). However, the specific molecular mechanisms by which STK11 mutations exert an influence on the biosynthesis of monounsaturated fatty acids (MUFAs) and subsequently affect ferroptosis in LUAD remain indistinct. In this study, bioinformatic analysis was employed to probe into the linkage between STK11 and key inhibitory genes of ferroptosis, namely SLC7A11 and SCD1, in LUAD tissues. Quantitative reverse transcription polymerase chain reaction was employed to assess the expression of STK11 in both wild-type and mutant STK11 LUAD cells, cell counting kit-8 to assess cell viability, and flow cytometry to detect apoptosis. A transmission electron microscope was utilized to observe mitochondrial morphology, and Western blot to ascertain the protein expression of STK11, ferroptosis-related proteins, and the enzyme SCD1 involved in MUFA synthesis. Oil red O staining was employed to test the distribution of lipid droplets in cancer cells, and a lipid quantification method to measure the content of MUFAs. Commercial kits were employed to assess the levels of lipid reactive oxygen species, malondialdehyde, glutathione, and Fe2+ in cells. The result revealed a negative correlation between STK11 and SLC7A11 as well as SCD1, with STK11 expression downregulated in mutant STK11 LUAD cells. Furthermore, STK11 mutations were found to suppress ferroptosis in LUAD cells by affecting MUFA synthesis. Subsequent rescue assays demonstrated that STK11 mutations hindered ferroptosis by impacting the synthesis of MUFAs in LUAD cells. This study provided evidence that STK11 mutations suppressed ferroptosis in LUAD cells by promoting MUFA synthesis, thus offering a novel research direction in the management of LUAD.
Collapse
Affiliation(s)
- Qiang Zou
- Department of Cardio-Thoracic Surgery, Zigong Fourth People’s Hospital, Zigong, Sichuan, 643000, China
| | - Bo Tang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People’s Hospital, Zigong, Sichuan, 643000, China
| | - Xianchao Chen
- Department of Cardio-Thoracic Surgery, Zigong Fourth People’s Hospital, Zigong, Sichuan, 643000, China
| | - Chuang Zhang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People’s Hospital, Zigong, Sichuan, 643000, China
| | - Yun Huang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People’s Hospital, Zigong, Sichuan, 643000, China
| |
Collapse
|
35
|
Zhang Z, Wang Z, Fan H, Li J, Ding J, Zhou G, Yuan C. The Indispensable Roles of GMDS and GMDS-AS1 in the Advancement of Cancer: Fucosylation, Signal Pathway and Molecular Pathogenesis. Mini Rev Med Chem 2024; 24:1712-1722. [PMID: 38591197 DOI: 10.2174/0113895575285276240324080234] [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: 12/01/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/10/2024]
Abstract
Fucosylation is facilitated by converting GDP-mannose to GDP-4-keto-6-deoxymannose, which GDP-mannose 4,6-dehydratase, a crucial enzyme in the route, carries out. One of the most prevalent glycosylation alterations linked to cancer has reportedly been identified as fucosylation. There is mounting evidence that GMDS is intimately linked to the onset and spread of cancer. Furthermore, the significance of long-chain non-coding RNAs in the development and metastasis of cancer is becoming more well-recognized, and the regulatory mechanism of lncRNAs has emerged as a prominent area of study in the biological sciences. GMDS-AS1, an antisense RNA of GMDS, was discovered to have the potential to be an oncogene. We have acquired and analyzed relevant data to understand better how GMDS-AS1 and its lncRNA work physiologically and in tumorigenesis and progression. Additionally, we have looked into the possible effects of these molecules on cancer treatment approaches and patient outcomes. The physiological roles and putative processes of GMDS and lncRNA GMDS-AS1 throughout the development and progression of tumors have been assembled and examined. We also examined how these chemicals might affect patient prognosis and cancer therapy approaches. GMDS and GMDS-AS1 were determined to be research subjects by searching and gathering pertinent studies using the PubMed system. The analysis of these research articles demonstrated the close relationship between GMDS and GMDS-AS1 and tumorigenesis and the factors that influence them. GMDS plays a vital role in regulating fucosylation. The related antisense gene GMDS-AS1 affects the biological behaviors of cancer cells through multiple pathways, including the key processes of proliferation, migration, invasion, and apoptosis, providing potential biomarkers and therapeutic targets for cancer treatment and prognosis assessment.
Collapse
Affiliation(s)
- Ziyan Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Zhuowei Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Hong Fan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Jiayi Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Jiaqi Ding
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| | - Gang Zhou
- College of Traditional Chinese Medicine, China Three Gorges University, Yichang 443002, China
- Yichang Hospital of Traditional Chinese Medicine, Yichang 443002, China
| | - Chengfu Yuan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China
- College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang 443005, China
| |
Collapse
|
36
|
Zou J. Site-specific delivery of cisplatin and paclitaxel mediated by liposomes: A promising approach in cancer chemotherapy. ENVIRONMENTAL RESEARCH 2023; 238:117111. [PMID: 37734579 DOI: 10.1016/j.envres.2023.117111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/25/2023] [Accepted: 09/09/2023] [Indexed: 09/23/2023]
Abstract
The site-specific delivery of drugs, especially anti-cancer drugs has been an interesting field for researchers and the reason is low accumulation of cytotoxic drugs in cancer cells. Although combination cancer therapy has been beneficial in providing cancer drug sensitivity, targeted delivery of drugs appears to be more efficient. One of the safe, biocompatible and efficient nano-scale delivery systems in anti-cancer drug delivery is liposomes. Their particle size is small and they have other properties such as adjustable physico-chemical properties, ease of functionalization and high entrapment efficiency. Cisplatin is a chemotherapy drug with clinical approval in patients, but its accumulation in cancer cells is low due to lack of targeted delivery and repeated administration results in resistance development. Gene and drug co-administration along with cisplatin/paclitaxel have resulted in increased sensitivity in tumor cells, but there is still space for more progress in cancer therapy. The delivery of cisplatin/paclitaxel by liposomes increases accumulation of drug in tumor cells and impairs activity of efflux pumps in promoting cytotoxicity. Moreover, phototherapy along with cisplatin/paclitaxel delivery can increase potential in tumor suppression. Smart nanoparticles including pH-sensitive nanoparticles provide site-specific delivery of cisplatin/paclitaxel. The functionalization of liposomes can be performed by ligands to increase targetability towards tumor cells in mediating site-specific delivery of cisplatin/paclitaxel. Finally, liposomes can mediate co-delivery of cisplatin/paclitaxel with drugs or genes in potentiating tumor suppression. Since drug resistance has caused therapy failure in cancer patients, and cisplatin/paclitaxel are among popular chemotherapy drugs, delivery of these drugs mediates targeted suppression of cancers and prevents development of drug resistance. Because of biocompatibility and safety of liposomes, they are currently used in clinical trials for treatment of cancer patients. In future, the optimal dose of using liposomes and optimal concentration of loading cisplatin/paclitaxel on liposomal nanocarriers in clinical trials should be determined.
Collapse
Affiliation(s)
- Jianyong Zou
- Department of Thoracic Surgery, The first Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, PR China.
| |
Collapse
|
37
|
Zhang Y, Shen G, Meng T, Lv Z, Li X, Li J, Li K. Eicosapentaenoic acid enhances the sensitivity of osteosarcoma to cisplatin by inducing ferroptosis through the DNA-PKcs/AKT/NRF2 pathway and reducing PD-L1 expression to attenuate immune evasion. Int Immunopharmacol 2023; 125:111181. [PMID: 37951196 DOI: 10.1016/j.intimp.2023.111181] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/13/2023]
Abstract
Acquired drug resistance poses a significant challenge in osteosarcoma therapy. Therefore, it is necessary for us to discover and develop an alternative anti-cancer strategy. Previous studies have shown that eicosapentaenoic acid (EPA) significantly increases chemosensitivity in cancer cells. In this study, we discovered that EPA enhances the sensitivity of osteosarcoma to cisplatin (DDP). Interestingly, in addition to inhibiting growth and inducing apoptosis, EPA also enhances DDP-induced ferroptosis. Western blot analysis confirmed that EPA treatment significantly decreases the expression of DNA-dependent protein kinase catalytic subunit (DNA-PKcs), p-AKT, nuclear factor erythroid 2-related factor 2 (NRF2), and glutathione peroxidase 4 (GPX4) in cells. Knockdown of DNA-PKcs by siRNA further enhances the level of ferroptosis induced by EPA. Importantly, EPA can reverse the high expression level of programmed death ligand 1 (PD-L1) induced by DDP. ELISA and western blotting analysis revealed that EPA treatment decreases the levels of IL-6 and p-STAT3, which are increased by DDP treatment. Furthermore, a co-immunoprecipitation (co-IP) assay confirmed the interaction between DNA-PKcs and PD-L1, and knockdown of DNA-PKcs further reduces the expression of PD-L1. This data provides the first evidence that EPA suppresses the DNA-PKcs/AKT/NRF2/GPX4 pathway to enhance ferroptosis, and inhibits IL-6/STAT3 and DNA-PKcs to decrease PD-L1 expression, thereby sensitizing osteosarcoma to DDP. The combination of EPA and DDP presents an encouraging and promising anti-tumor strategy.
Collapse
Affiliation(s)
- Yining Zhang
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
| | - Guohong Shen
- Department of Pediatrics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Tingting Meng
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Zhaorui Lv
- The First Clinical College of Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China; Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
| | - Xin Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
| | - Jianmin Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China
| | - Ka Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, 250012 Jinan, Shandong, China.
| |
Collapse
|
38
|
Lee J, Roh JL. Epigenetic modulation of ferroptosis in cancer: Identifying epigenetic targets for novel anticancer therapy. Cell Oncol (Dordr) 2023; 46:1605-1623. [PMID: 37438601 DOI: 10.1007/s13402-023-00840-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
Ferroptosis is a newly recognized form of oxidative-regulated cell death resulting from iron-mediated lipid peroxidation accumulation. Radical-trapping antioxidant systems can eliminate these oxidized lipids and prevent disrupting the integrity of cell membranes. Epigenetic modifications can regulate ferroptosis by altering gene expression or cell phenotype without permanent sequence changes. These mechanisms include DNA methylation, histone modifications, RNA modifications, and noncoding RNAs. Epigenetic alterations in cancer can control the expression of ferroptosis regulators or related pathways, leading to changes in cell sensitivity to ferroptosis inducers or cancer progression. Epigenetic alterations in cancer are influenced by a wide range of cancer hallmarks, contributing to therapeutic resistance. Targeting epigenetic alterations is a promising approach to overcoming cancer resilience. However, the exact mechanisms involved in different types of cancer remain unresolved. Discovering more ferroptosis-associated epigenetic targets and interventions can help overcome current barriers in anticancer therapy. Many papers on epigenetic modifications of ferroptosis have been continuously published, making it essential to summarize the current state-of-the-art in the epigenetic regulation of ferroptosis in human cancer.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, 13496, Republic of Korea
- Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-do, 13496, Republic of Korea.
- Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
39
|
Liao J, Qing X, Deng G, Xiao Y, Fu Y, Han S, Li X, Gan Y, Li W. Gastrodin destabilizes survivin and overcomes pemetrexed resistance. Cell Signal 2023; 110:110851. [PMID: 37586466 DOI: 10.1016/j.cellsig.2023.110851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/01/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Survivin is a bifunctional protein that plays crucial roles in tumorigenesis. In the present study, we discovered that the natural product gastrodin suppressed the cell viability and colony formation of non-small cell lung cancer (NSCLC) cell lines A549, HCC827, and H460 in a dose-dependent manner. In addition, gastrodin enhanced the protein levels of cleaved-caspase 3 by activating the endogenous mitochondrial apoptosis pathway. Gastrodin inhibits protein kinase B (Akt)/WEE1/cyclin-dependent kinase 1 (CDK1) signaling to downregulate survivin Thr34 phosphorylation. Survivin Thr34 dephosphorylation caused by gastrodin interfered with the binding of ubiquitin-specific protease 19 (USP19), which eventually destabilized survivin. We revealed that the growth of NSCLC xenograft tumors was markedly suppressed by gastrodin in vivo. Furthermore, gastrodin overcomes pemetrexed resistance in vivo or in vitro. Our results suggest that gastrodin is a potential antitumor agent by reducing survivin in NSCLC.
Collapse
Affiliation(s)
- Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiang Qing
- Department of Otolaryngology Head and Neck Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Gaoyan Deng
- Department of Thoracic Surgery, Hunan Chest Hospital, Changsha, Hunan, China
| | - Yeqing Xiao
- Department of Ultrasonography, Hunan Chest Hospital, Changsha, Hunan, China
| | - Yaqian Fu
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
40
|
Wei X, Li X, Hu S, Cheng J, Cai R. Regulation of Ferroptosis in Lung Adenocarcinoma. Int J Mol Sci 2023; 24:14614. [PMID: 37834062 PMCID: PMC10572737 DOI: 10.3390/ijms241914614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common lung cancer, which accounts for about 35-40% of all lung cancer patients. Despite therapeutic advancements in recent years, the overall survival time of LUAD patients still remains poor, especially KRAS mutant LUAD. Therefore, it is necessary to further explore novel targets and drugs to improve the prognos is for LUAD. Ferroptosis, an iron-dependent regulated cell death (RCD) caused by lipid peroxidation, has attracted much attention recently as an alternative target for apoptosis in LUAD therapy. Ferroptosis has been found to be closely related to LUAD at every stage, including initiation, proliferation, and progression. In this review, we will provide a comprehensive overview of ferroptosis mechanisms, its regulation in LUAD, and the application of targeting ferroptosis for LUAD therapy.
Collapse
Affiliation(s)
| | | | | | - Jinke Cheng
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (X.W.); (X.L.); (S.H.)
| | - Rong Cai
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (X.W.); (X.L.); (S.H.)
| |
Collapse
|
41
|
Zheng X, Zhang C. The Regulation of Ferroptosis by Noncoding RNAs. Int J Mol Sci 2023; 24:13336. [PMID: 37686142 PMCID: PMC10488123 DOI: 10.3390/ijms241713336] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
As a novel form of regulated cell death, ferroptosis is characterized by intracellular iron and lipid peroxide accumulation, which is different from other regulated cell death forms morphologically, biochemically, and immunologically. Ferroptosis is regulated by iron metabolism, lipid metabolism, and antioxidant defense systems as well as various transcription factors and related signal pathways. Emerging evidence has highlighted that ferroptosis is associated with many physiological and pathological processes, including cancer, neurodegeneration diseases, cardiovascular diseases, and ischemia/reperfusion injury. Noncoding RNAs are a group of functional RNA molecules that are not translated into proteins, which can regulate gene expression in various manners. An increasing number of studies have shown that noncoding RNAs, especially miRNAs, lncRNAs, and circRNAs, can interfere with the progression of ferroptosis by modulating ferroptosis-related genes or proteins directly or indirectly. In this review, we summarize the basic mechanisms and regulations of ferroptosis and focus on the recent studies on the mechanism for different types of ncRNAs to regulate ferroptosis in different physiological and pathological conditions, which will deepen our understanding of ferroptosis regulation by noncoding RNAs and provide new insights into employing noncoding RNAs in ferroptosis-associated therapeutic strategies.
Collapse
Affiliation(s)
| | - Cen Zhang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China;
| |
Collapse
|
42
|
Zhang H, Liang J, Lu T, Li M, Shan G, Bi G, Zhao M, Jin X, Wang Q, Chen Z, Zhan C. AGRN promotes lung adenocarcinoma progression by activating Notch signaling pathway and acts as a therapeutic target. Pharmacol Res 2023; 194:106819. [PMID: 37321467 DOI: 10.1016/j.phrs.2023.106819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/17/2023]
Abstract
Lung cancer is the main reason for cancer-associated death globally, and lung adenocarcinoma (LUAD) is the most prevalent subtype of lung cancer. Recently, AGRN is considered playing an vital role in the development of some cancers. However, the regulatory effects and mechanisms of AGRN in LUAD remain elusive. In this study, we clarified the significant upregulation of AGRN expression in LUAD by single-cell RNA sequencing combined with immunohistochemistry. Besides, we confirmed that LUAD patients with high AGRN expression are more susceptible to lymph node metastases and have a worse prognosis by a retrospective study of 120 LUAD patients. Next, we demonstrated that AGRN directly interact with NOTCH1, which results in the release of the intracellular structural domain of NOTCH1 and the subsequent activation of the NOTCH pathway. Moreover, we also found that AGRN promotes proliferation, migration, invasion, EMT and tumorigenesis of LUAD cells in vitro and in vivo, and that these effects are reversed by blocking the NOTCH pathway. Furthermore, we prepared several antibodies targeting AGRN, and clarify that Anti-AGRN antibody treatment could significantly inhibit proliferation and promote apoptosis of tumor cells. Our study highlights the important role and regulatory mechanism of AGRN in LUAD development and progression, and suggests that antibodies targeting AGRN have therapeutic potential for LUAD. We also provide theoretical and experimental evidence for further development of monoclonal antibodies targeting AGRN.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China; Division of Thoracic Surgery, Sichuan Cancer Hospital & Research Institute, School of Medicine, University of Electronic Science and Technology of China (UESTC), Chengdu, People's Republic of China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Tao Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xing Jin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zhengcong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China; Department of Thoracic Surgery, ShangHai Geriatric Medicine Center, Shanghai, People's Republic of China.
| |
Collapse
|
43
|
Zhang Q, Fan X, Zhang X, Ju S. Ferroptosis in tumors and its relationship to other programmed cell death: role of non-coding RNAs. J Transl Med 2023; 21:514. [PMID: 37516888 PMCID: PMC10387214 DOI: 10.1186/s12967-023-04370-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023] Open
Abstract
Programmed cell death (PCD) plays an important role in many aspects of individual development, maintenance of body homeostasis and pathological processes. Ferroptosis is a novel form of PCD characterized by the accumulation of iron-dependent lipid peroxides resulting in lethal cell damage. It contributes to tumor progression in an apoptosis-independent manner. In recent years, an increasing number of non-coding RNAs (ncRNAs) have been demonstrated to mediate the biological process of ferroptosis, hence impacting carcinogenesis, progression, drug resistance, and prognosis. However, the clear regulatory mechanism for this phenomenon remains poorly understood. Moreover, ferroptosis does not usually exist independently. Its interaction with PCD, like apoptosis, necroptosis, autophagy, pyroptosis, and cuproptosis, to destroy cells appears to exist. Furthermore, ncRNA seems to be involved. Here, we review the mechanisms by which ferroptosis occurs, dissect its relationship with other forms of death, summarize the key regulatory roles played by ncRNAs, raise relevant questions and predict possible barriers to its application in the clinic, offering new ideas for targeted tumour therapy.
Collapse
Affiliation(s)
- Qi Zhang
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xinfeng Fan
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xinyu Zhang
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- Department of Medical School of Nantong University, No.19, Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Shaoqing Ju
- Medical School of Nantong University, Nantong University, Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, No.20, Xisi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
44
|
Bi G, Liang J, Shan G, Bian Y, Chen Z, Huang Y, Lu T, Li M, Besskaya V, Zhao M, Fan H, Wang Q, Gan B, Zhan C. Retinol Saturase Mediates Retinoid Metabolism to Impair a Ferroptosis Defense System in Cancer Cells. Cancer Res 2023; 83:2387-2404. [PMID: 37184371 DOI: 10.1158/0008-5472.can-22-3977] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/22/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
Ferroptosis is an iron-dependent form of regulated cell death induced by the lethal overload of lipid peroxides in cellular membranes. In recent years, modulating ferroptosis has gained attention as a potential therapeutic approach for tumor suppression. In the current study, retinol saturase (RETSAT) was identified as a significant ferroptosis mediator using a publicly accessible CRISPR/Cas9 screening dataset. RETSAT depletion protected tumor cells from lipid peroxidation and subsequent cell death triggered by various ferroptosis inducers. Furthermore, exogenous supplementation with retinoids, including retinol (the substrate of RETSAT) and its derivatives retinal and retinoic acid, also suppressed ferroptosis, whereas the product of RETSAT, 13, 14-dihydroretinol, failed to do so. As effective radical-trapping antioxidant, retinoids protected the lipid membrane from autoxidation and subsequent fragmentation, thus terminating the cascade of ferroptosis. Pseudotargeted lipidomic analysis identified an association between retinoid regulation of ferroptosis and lipid metabolism. Retinoic acid, but not 13, 14-dihydroretinoic acid, interacted with its nuclear receptor and activated transcription of stearoyl-CoA desaturase, which introduces the first double bond into saturated fatty acid and thus catalyzes the generation of monounsaturated fatty acid, a known ferroptosis suppressor. Therefore, RETSAT promotes ferroptosis by transforming retinol to 13, 14-dihydroretinol, thereby turning a strong anti-ferroptosis regulator into a relatively weak one. SIGNIFICANCE Retinoids have ferroptosis-protective properties and can be metabolized by RETSAT to promote ferroptosis, suggesting the possibility of targeting retinoid metabolism in cancer as a treatment strategy to trigger ferroptosis.
Collapse
Affiliation(s)
- Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Tao Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Valeria Besskaya
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
45
|
Zhao G, Liang J, Shan G, Gu J, Xu F, Lu C, Ma T, Bi G, Zhan C, Ge D. KLF11 regulates lung adenocarcinoma ferroptosis and chemosensitivity by suppressing GPX4. Commun Biol 2023; 6:570. [PMID: 37248295 DOI: 10.1038/s42003-023-04959-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 05/20/2023] [Indexed: 05/31/2023] Open
Abstract
Ferroptosis, an iron-dependent non-apoptotic cell death, has been shown to play a vital role in tumor proliferation and chemotherapy resistance. Here, we report that KLF11 inhibits lung adenocarcinoma (LUAD) cell proliferation and promotes chemotherapy sensitivity by participating in the GPX4-related ferroptosis pathway. Through an RNA-sequence screen from LUAD cells pretreatment with ferroptosis inducers (FINs), we discovered that KLF11 expression was significantly higher in FINs-treated cells, suggesting that KLF11 may be involved in ferroptosis. Overexpression of KLF11 promoted LUAD cells to undergo ferroptosis alterations. Meanwhile, upregulation of KLF11 expression also inhibited cell proliferation and increased chemosensitivity, whereas knockout of KLF11 did the opposite. With ChIP-Seq and RNA-Seq, we identified GPX4 as a downstream target of KLF11. Through ChIP-qPCR and dual luciferase assay, we clarified that KLF11 binds to the promoter region of GPX4 and represses its transcription. Restored GPX4 expression antagonized the ability of KLF11 to promote ferroptosis, increase chemotherapy sensitivity and inhibit cell proliferation in vitro and in vivo. Clinically, KLF11 declined in LUAD and its low expression was associated with reduced patient survival. Our findings established the function of KLF11 to promote ferroptosis in LUAD, thereby inhibiting cell proliferation and enhancing the efficacy of chemotherapy.
Collapse
Affiliation(s)
- Guangyin Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fengkai Xu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunlai Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Teng Ma
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
46
|
Guo S, Zhong A, Zhang D, Gao J, Ni Y, Zhao R, Ma W. ATP2B3 Inhibition Alleviates Erastin-Induced Ferroptosis in HT-22 Cells through the P62-KEAP1-NRF2-HO-1 Pathway. Int J Mol Sci 2023; 24:ijms24119199. [PMID: 37298147 DOI: 10.3390/ijms24119199] [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: 04/28/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Ferroptosis participates in the occurrence and development of neurological disorders. Modulating ferroptosis may have therapeutic potential in nervous system diseases. Therefore, TMTbased proteomic analysis in HT-22 cells was performed to identify erastin-induced differentially expressed proteins. The calcium-transporting ATP2B3 (ATP2B3) was screened as a target protein. ATP2B3 knockdown markedly alleviated the erastin-induced decrease in cell viability and elevated ROS (p < 0.01) and reversed the up-regulation of oxidative stress-related proteins polyubiquitin-binding protein p62 (P62), nuclear factor erythroid 2-related factor2 (NRF2), heme oxygenase-1 (HO-1), and NAD(P)H quinone oxidoreductase-1 (NQO1) protein expression (p < 0.05 or p < 0.01) and the down-regulation of Kelch-like ECH-associated protein 1(KEAP1) protein expression (p < 0.01). Moreover, NRF2 knockdown, P62 inhibition, or KEAP1 overexpression rescued the erastin-induced decrease in cell viability (p < 0.05) and increase in ROS production (p < 0.01) in HT-22 cells, while simultaneous overexpression of NRF2 and P62 and knockdown of KEAP1 partially offset the relief effect of ATP2B3 inhibition. In addition, knockdown of ATP2B3, NRF2, and P62 and overexpression of KEAP1 significantly down-regulated erastin-induced high expression of the HO-1 protein, while HO-1 overexpression reversed the alleviating effects of ATP2B3 inhibition on the erastin-induced decrease in cell viability (p < 0.01) and increase in ROS production (p < 0.01) in HT-22 cells. Taken together, ATP2B3 inhibition mediates the alleviation of erastin-induced ferroptosis in HT-22 cells through the P62-KEAP1-NRF2-HO-1 pathway.
Collapse
Affiliation(s)
- Shihui Guo
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Aiying Zhong
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Dongxu Zhang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiang Gao
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Yingdong Ni
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenqiang Ma
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
47
|
Zheng J, Zhang Q, Zhao Z, Qiu Y, Zhou Y, Wu Z, Jiang C, Wang X, Jiang X. Epigenetically silenced lncRNA SNAI3-AS1 promotes ferroptosis in glioma via perturbing the m 6A-dependent recognition of Nrf2 mRNA mediated by SND1. J Exp Clin Cancer Res 2023; 42:127. [PMID: 37202791 DOI: 10.1186/s13046-023-02684-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/24/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Ferroptosis has been linked to tumor progression and resistance to antineoplastic therapy. Long noncoding RNA (lncRNA) exerts a regulatory role in various biological processes of tumor cells, while the function and molecular mechanism of lncRNA in ferroptosis are yet to be clarified in glioma. METHODS Both gain-of-function and loss-of-function experiments were employed to investigate the effects of SNAI3-AS1 on the tumorigenesis and ferroptosis susceptibility of glioma in vitro and in vivo. Bioinformatics analysis, Bisulfite sequencing PCR, RNA pull-down, RIP, MeRIP and dual-luciferase reporter assay were performed to explore the low expression mechanism of SNAI3-AS1 and the downstream mechanism of SNAI3-AS1 in ferroptosis susceptibility of glioma. RESULTS We found that ferroptosis inducer erastin downregulates SNAI3-AS1 expression in glioma by increasing the DNA methylation level of SNAI3-AS1 promoter. SNAI3-AS1 functions as a tumor suppressor in glioma. Importantly, SNAI3-AS1 enhances the anti-tumor activity of erastin by promoting ferroptosis both in vitro and in vivo. Mechanistically, SNAI3-AS1 competitively binds to SND1 and perturbs the m6A-dependent recognition of Nrf2 mRNA 3'UTR by SND1, thereby reducing the mRNA stability of Nrf2. Rescue experiments confirmed that SND1 overexpression and silence can rescue the gain- and loss-of-function ferroptotic phenotypes of SNAI3-AS1, respectively. CONCLUSIONS Our findings elucidate the effect and detailed mechanism of SNAI3-AS1/SND1/Nrf2 signalling axis in ferroptosis, and provide a theoretical support for inducing ferroptosis to improve glioma treatment.
Collapse
Affiliation(s)
- Jianglin Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yue Qiu
- Department of Otolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yujie Zhou
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhipeng Wu
- Department of Neurosurgery, Weifang People's Hospital, Weifang, Shandong, China
| | - Cheng Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuan Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
48
|
Wang Z, Zhou L, Chen B, Li X, Zou Q, Xu W, Fang L, Wu A, Li Z, Chen Y. microRNA- 660 Enhances Cisplatin Sensitivity via Decreasing SATB2 Expression in Lung Adenocarcinoma. Genes (Basel) 2023; 14:genes14040911. [PMID: 37107669 PMCID: PMC10137726 DOI: 10.3390/genes14040911] [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: 02/02/2023] [Revised: 03/24/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Increasing evidence suggests that microRNAs' (miRNAs) abnormal expression is one of the main factors of chemotherapy resistance in various cancers. However, the role of miRNAs in lung adenocarcinoma (LUAD) resistance to cisplatin is still unclear. In this study, we analyzed a microarray dataset to investigate miRNAs related to cisplatin resistance in LUAD. The expression of miRNAs in LUAD tissues and cell lines was detected using real-time quantitative polymerase chain reaction (RT-qPCR). Special AT-Rich Sequence-Binding Protein 2 (SATB2) in LUAD cell lines was detected using RT-qPCR and Western blot. Cell proliferation was measured by CCK8 and colony formation assays, while cell cycle and apoptosis were measured by flow cytometry. A dual-luciferase reporter assay was performed to confirm that SATB2 is a target gene of microRNA-660 (miR-660). We showed that the expression of miR-660 was not only decreased in LUAD cells and tissues but also further decreased in the cisplatin-resistant A549 cell line. The overexpression of miR-660 increased cisplatin sensitivity in LUAD cells. In addition, we identified SATB2 as a direct target gene of miR-660. We also revealed that miR-660 increased cisplatin sensitivity in LUAD cells via targeting SATB2. In conclusion, miR-660/SATB2 axis is a key regulator of cisplatin resistance in LUAD.
Collapse
Affiliation(s)
- Ziyao Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Lingxuan Zhou
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Bisong Chen
- Department of Pathophysiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Xu Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Qiuyi Zou
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Wei Xu
- NHC Key Laboratory of Carcinogenesis, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Li Fang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Anbang Wu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Zheng Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
- NHC Key Laboratory of Carcinogenesis, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| | - Yuejun Chen
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410006, China
| |
Collapse
|
49
|
Xing N, Du Q, Guo S, Xiang G, Zhang Y, Meng X, Xiang L, Wang S. Ferroptosis in lung cancer: a novel pathway regulating cell death and a promising target for drug therapy. Cell Death Discov 2023; 9:110. [PMID: 37005430 PMCID: PMC10067943 DOI: 10.1038/s41420-023-01407-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/04/2023] Open
Abstract
Lung cancer is a common malignant tumor that occurs in the human body and poses a serious threat to human health and quality of life. The existing treatment methods mainly include surgical treatment, chemotherapy, and radiotherapy. However, due to the strong metastatic characteristics of lung cancer and the emergence of related drug resistance and radiation resistance, the overall survival rate of lung cancer patients is not ideal. There is an urgent need to develop new treatment strategies or new effective drugs to treat lung cancer. Ferroptosis, a novel type of programmed cell death, is different from the traditional cell death pathways such as apoptosis, necrosis, pyroptosis and so on. It is caused by the increase of iron-dependent reactive oxygen species due to intracellular iron overload, which leads to the accumulation of lipid peroxides, thus inducing cell membrane oxidative damage, affecting the normal life process of cells, and finally promoting the process of ferroptosis. The regulation of ferroptosis is closely related to the normal physiological process of cells, and it involves iron metabolism, lipid metabolism, and the balance between oxygen-free radical reaction and lipid peroxidation. A large number of studies have confirmed that ferroptosis is a result of the combined action of the cellular oxidation/antioxidant system and cell membrane damage/repair, which has great potential application in tumor therapy. Therefore, this review aims to explore potential therapeutic targets for ferroptosis in lung cancer by clarifying the regulatory pathway of ferroptosis. Based on the study of ferroptosis, the regulation mechanism of ferroptosis in lung cancer was understood and the existing chemical drugs and natural compounds targeting ferroptosis in lung cancer were summarized, with the aim of providing new ideas for the treatment of lung cancer. In addition, it also provides the basis for the discovery and clinical application of chemical drugs and natural compounds targeting ferroptosis to effectively treat lung cancer.
Collapse
Affiliation(s)
- Nan Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qinyun Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Sa Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Gelin Xiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, 620010, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Li Xiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Shaohui Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
- State Key Laboratory of Southwestern Chinese Medicine Resources, Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, 620010, China.
| |
Collapse
|
50
|
Hu C, Zeng X, Zhu Y, Huang Z, Liu J, Ji D, Zheng Z, Wang Q, Tan W. Regulation of ncRNAs involved with ferroptosis in various cancers. Front Genet 2023; 14:1136240. [PMID: 37065473 PMCID: PMC10090411 DOI: 10.3389/fgene.2023.1136240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/08/2023] [Indexed: 03/31/2023] Open
Abstract
As a special pattern of programmed cell death, ferroptosis is reported to participate in several processes of tumor progression, including regulating proliferation, suppressing apoptotic pathways, increasing metastasis, and acquiring drug resistance. The marked features of ferroptosis are an abnormal intracellular iron metabolism and lipid peroxidation that are pluralistically modulated by ferroptosis-related molecules and signals, such as iron metabolism, lipid peroxidation, system Xc−, GPX4, ROS production, and Nrf2 signals. Non-coding RNAs (ncRNAs) are a type of functional RNA molecules that are not translated into a protein. Increasing studies demonstrate that ncRNAs have a diversity of regulatory roles in ferroptosis, thus influencing the progression of cancers. In this study, we review the fundamental mechanisms and regulation network of ncRNAs on ferroptosis in various tumors, aiming to provide a systematic understanding of recently emerging non-coding RNAs and ferroptosis.
Collapse
Affiliation(s)
- Chenxi Hu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiangbo Zeng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuanchao Zhu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zehai Huang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiacheng Liu
- Department of Infectious Diseases, Peking University Hepatology Institute, Peking University People’s Hospital, Beijing, China
| | - Ding Ji
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zaosong Zheng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- *Correspondence: Zaosong Zheng, ; Qiong Wang, ; Wanlong Tan,
| | - Qiong Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- *Correspondence: Zaosong Zheng, ; Qiong Wang, ; Wanlong Tan,
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- *Correspondence: Zaosong Zheng, ; Qiong Wang, ; Wanlong Tan,
| |
Collapse
|