1
|
Jiang JM, Cheng ZW, Zhang L, Tan TF, Zhang T, Shi HB, Hou KF, Xia Q. Proteomic insights into the molecular mechanism of anlotinib inhibition in TP53-mutated non-small cell lung cancer. J Proteomics 2025; 316:105433. [PMID: 40113013 DOI: 10.1016/j.jprot.2025.105433] [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: 09/25/2024] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE Tumor protein 53 (TP53) is the commonly mutated gene in non-small cell lung cancer (NSCLC) that is associated with poor prognosis, and anlotinib exerts inhibitory effects on TP53-mutated NSCLC. The aim of this study was to investigate the inhibitory effect of anlotinib on TP53-mutated NSCLC and its possible mechanism. METHODS The growth ability of TP53-mutated NSCLC cells were tested by Cell counting kit-8 assay. Proteins in TP53-mutated NSCLC cells treated with anlotinib were analyzed using label-free liquid chromatography-mass spectrometry. Differentially represented proteins were analyzed by KEGG, GO, and PPIs. TP53 pathway related proteins were verified using western blotting. RESULTS The cell viability was significantly reduced in TP53-mutated NSCLC cell as opposed to TP53 wild cell by anlotinib treatment. 126 differentially represented proteins (37 upregulated and 89 downregulated) were found between the anlotinib and control groups in TP53-mutated NSCLC cell. Bioinformatics analyses revealed that the differentially represented proteins were primarily involved in catalytic activity, cellular processes, and metabolite interconversion. PANTHER Classification System found that anlotinib mainly impacted the p53 signaling pathway, De novo purine biosynthesis and Integrin signaling. KEGG enrichment and PPI networks of the differentially represented proteins revealed cyclin-dependent kinase 1 (CDK1) and mitogen-activated protein kinase kinase 3 (MAP2K3) as the core protein, which are related to the p53 signaling pathway. Western blotting also revealed that anlotinib significantly suppressed the expression of CDK1 and MAP2K3 in TP53-mutated NSCLC cells, that indicated the possible mechanism may involve the MAP2K3/p53/CDK1 pathway. CONCLUSIONS Our findings showed that anlotinib selectively inhibited the growth of TP53-mutated NSCLC cells and downregulated the expression levels of CDK1 and MAP2K3. The MAP2K3/p53/CDK1 pathway may be the molecular mechanism underlying anlotinib's efficacy in TP53-mutated NSCLC. STATEMENT OF SIGNIFICANCE Tumor protein 53 (TP53) is the commonly mutated gene in non-small cell lung cancer (NSCLC) that is associated with poor prognosis, and anlotinib exerts inhibitory effects on TP53-mutated NSCLC. However, the action mechanism of anlotinib in the treatment of TP53-mutated NSCLC remains unclear. In this study, we used label-free quantitative proteomics to reveal the molecular mechanism of anlotinib inhibition in TP53-mutated NSCLC. We found that anlotinib significantly inhibited the growth of TP53-mutated NSCLC cells and downregulated the expression levels of CDK1 and MAP2K3. The MAP2K3/p53/CDK1 pathway may be the molecular mechanism underlying anlotinib's efficacy in TP53-mutated NSCLC. Our study promotes the use of anti-angiogenic drugs in TP53-mutated NSCLC. It provides new ideas for the treatment of TP53-mutated NSCLC.
Collapse
Affiliation(s)
- Jie-Mei Jiang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zi-Wei Cheng
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Ting-Fei Tan
- Department of Pharmacy, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Teng Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Han-Bing Shi
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Kai-Feng Hou
- Hematologic Lymphoma Center, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Xiao J, Xu Z, Deng Z, Xie J, Qiu Y. METTL3 facilitates osteoblast differentiation and bone regeneration via m6A-dependent maturation of pri-miR-324-5p. Cell Immunol 2025; 413:104974. [PMID: 40398354 DOI: 10.1016/j.cellimm.2025.104974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Osteoblast differentiation is essential for fracture healing and bone regeneration. miR-324-5p has been implicated in osteoporosis, but its precise role in osteogenic differentiation remains unclear. We investigated the function and regulatory mechanisms of miR-324-5p in bone marrow mesenchymal stem cells (BMSCs). METHODS RT-qPCR was used to assess miR-324-5p expression during osteogenic differentiation of BMSCs. ALP, Alizarin Red S (ARS), Oil Red O, and TRAP staining were performed to evaluate osteoblast, adipocyte, and osteoclast differentiation. Rat femoral fracture and calvarial bone defect models were established to assess in vivo bone regeneration. Methylated RNA immunoprecipitation (MeRIP) and luciferase reporter assays were used to investigate METTL3-mediated m6A modification of pri-miR-324-5p and its regulation of ELAVL1. RESULTS miR-324-5p expression increased during osteogenic differentiation, and ALP and ARS staining confirmed enhanced osteoblast activity and mineralization following miR-324-5p overexpression. Meanwhile, Oil Red O staining showed reduced adipogenic differentiation, and TRAP staining demonstrated suppressed osteoclast formation. In vivo, miR-324-5p promoted bone healing, bone mass, and bone regeneration. Mechanistically, METTL3-mediated m6A modification facilitated pri-miR-324-5p maturation, positively regulating its expression. Additionally, miR-324-5p directly targeted ELAVL1, and ELAVL1 overexpression reversed the osteogenic effects of miR-324-5p. CONCLUSION The METTL3/miR-324-5p/ELAVL1 axis plays a crucial role in osteogenic differentiation and bone regeneration, providing new insights into m6A modification-driven osteogenesis.
Collapse
Affiliation(s)
- Jing Xiao
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510000, Guangdong, China
| | - Zhiyuan Xu
- Department of Orthopaedics, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, Guangdong, China
| | - Zhiwei Deng
- Department of Orthopaedics, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, Guangdong, China
| | - Juntong Xie
- Department of Orthopaedics, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, Guangdong, China
| | - Yiyan Qiu
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Orthopedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510000, Guangdong, China; Department of Orthopaedics, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen 518172, Guangdong, China.
| |
Collapse
|
3
|
Jia MY, Wu C, Fu Z, Xu WB, Liu J, Wu CY, Zeng XY, Wu YL, Yan H. Targeting the HuR/E2F7 axis synergizes with bortezomib against multiple myeloma. Acta Pharmacol Sin 2025:10.1038/s41401-025-01529-3. [PMID: 40133626 DOI: 10.1038/s41401-025-01529-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/28/2025] [Indexed: 03/27/2025]
Abstract
Multiple myeloma (MM) is a malignant hematological disease caused by the proliferation of abnormal plasma cells in the bone marrow and is still incurable. Relapse and drug resistance are common in MM. New therapeutic targets are urgently needed for MM treatment. Human antigen R (HuR) has been reported to play an important role in the malignant biological behavior of a variety of tumors, but its role in MM remains unclear. In this study, we found that HuR was highly expressed in MM patients and associated with a poor prognosis by analyzing public datasets. We found that targeting HuR with short hairpin RNA (shRNA) or its inhibitor CMLD-2 had significant anti-MM effects both in vitro and in vivo. The overexpression of HuR promotes MM cell proliferation in vitro and in vivo. Moreover, we demonstrated that bortezomib drug sensitivity increased and decreased with the knockdown and overexpression of HuR, respectively. This result provides a rationale for our subsequent combination of CMLD-2 with bortezomib in the treatment of MM. To further explore the mechanism of HuR in MM, we performed RNA sequencing and identified its downstream molecule, E2F7. HuR upregulated E2F7 expression by increasing the stability of its mRNA in MM cells. Higher levels of E2F7 were associated with a poorer prognosis. E2F7 knockdown had anti-MM effects in vitro and in vivo. E2F7 overexpression partially rescued the cell proliferation inhibition and apoptosis caused by targeting HuR in MM cells. We subsequently demonstrated that CMLD-2 synergized with the anti-MM effect of bortezomib both in vitro and in vivo. In conclusion, targeting the HuR/E2F7 axis synergizes with bortezomib against MM. Therefore, the HuR/E2F7 axis may serve as a promising therapeutic target for MM.
Collapse
Affiliation(s)
- Ming-Yuan Jia
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chao Wu
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ze Fu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wen-Bin Xu
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jia Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Cheng-Yu Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin-Yi Zeng
- Department of Hematology, Huadong Hospital Affiliated with Fudan University, Shanghai, 200040, China
| | - Ying-Li Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| | - Hua Yan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of General Practice, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
4
|
Zhou H, Shu R, Wu J, Zhou J, Yu Z, Cheng Q, Peng Z, Zhao M. Review of the role and potential clinical value of m6A methylation modifications in the biological process of osteosarcoma. Front Genet 2025; 16:1522622. [PMID: 40176793 PMCID: PMC11961878 DOI: 10.3389/fgene.2025.1522622] [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: 11/04/2024] [Accepted: 02/17/2025] [Indexed: 04/04/2025] Open
Abstract
Osteosarcoma (OS), an aggressive bone tumor, is a substantial threat to the quality of life and survival of affected individuals. Despite recent improvements in OS therapies, the considerable variability and chemotherapy resistance of this cancer necessitate continuous research to discover new treatment targets and biomarkers. Recent epigenetic advances highlight the crucial role of N6-methyladenosine (m6A) methylation in cancer. In OS, m6A methylation has been demonstrated to be a pivotal component in the pathogenesis. This review introduces new findings regarding the association between m6A methylation regulators and OS, and summarizes the potential clinical applications of OS and m6A methylation regulators, including the role of m6A methylation in OS proliferation, growth, apoptosis, and cell migration, invasion, and metastasis; relationship between m6A methylation and OS chemotherapy resistance; and relationship between m6A methylation and OS prognosis. Our review had certain limitations. The interaction between m6A methylation regulators and other oncogenic factors, such as lncRNAs and ncRNAs, is not fully understood. We hope that these potential methods will be translated into clinical applications and effective treatment.
Collapse
Affiliation(s)
- Huaqiang Zhou
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Rongbing Shu
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Jianming Wu
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Jiangjun Zhou
- Department of Orthopedic, The 908Th Hospital of Joint Logistic Support Force of PLA, Nanchang, China
| | - Zhuanyi Yu
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Qiuxin Cheng
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Zhihao Peng
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Min Zhao
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| |
Collapse
|
5
|
Rahmati R, Zarimeidani F, Ahmadi F, Yousefi-Koma H, Mohammadnia A, Hajimoradi M, Shafaghi S, Nazari E. Identification of novel diagnostic and prognostic microRNAs in sarcoma on TCGA dataset: bioinformatics and machine learning approach. Sci Rep 2025; 15:7521. [PMID: 40032929 PMCID: PMC11876432 DOI: 10.1038/s41598-025-91007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
The discovery of unique microRNA (miR) patterns and their corresponding genes in sarcoma patients indicates their involvement in cancer development and suggests their potential use in medical management. MiRs were identified from The Cancer Genome Atlas (TCGA) dataset, with a Deep Neural Network (DNN) employed for novel miR identification. MiRDB facilitated target predictions. Functional enrichment analysis, identify critical pathways, protein-protein interaction network, and diseases/clinical data correlations were explored. COX regression, Kaplan-Meier analyses, and CombioROC was also utilized. The population consisted of 119 females and 142 males, and 1046 miRs were uncovered. Ten miRs was selected for further analysis using DNN. Upon analyzing for gene ontology, it was found that these genes showed enrichment in various activities. We identified a significant association between the overall survival rate of sarcoma patients and miRs levels. The combination of miR.3688 and miR.3936 achieved the greatest diagnostic standing. MiRs have the capability to screen sarcoma patients to identify undetected tumors, predict prognosis, and pinpoint prospective targets for treatment. Further large clinical trials are required to validate our findings.
Collapse
Affiliation(s)
- Rahem Rahmati
- Students Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Lung Transplantation Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Zarimeidani
- Students Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Lung Transplantation Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farnaz Ahmadi
- Lung Transplantation Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hannaneh Yousefi-Koma
- Lung Transplantation Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolreza Mohammadnia
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Hajimoradi
- Lung Transplantation Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shadi Shafaghi
- Lung Transplantation Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Elham Nazari
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Zhou W, Wang X, Yan B, Sun Y. Embryonic Lethal Abnormal Visual-Like Protein 1 Aggravates Caerulein-Induced AR42J Cell Injury and Macrophage M1 Polarization to Accelerate Acute Pancreatitis by Upregulating TRAF6. J Interferon Cytokine Res 2025; 45:20-28. [PMID: 39535226 PMCID: PMC11839522 DOI: 10.1089/jir.2024.0149] [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: 07/26/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Tumor necrosis factor receptor-associated factor 6 (TRAF6) has been found to promote the progression of acute pancreatitis (AP). However, its underlying molecular mechanisms in AP need to be further revealed. Caerulein-induced AR42J cells were used to construct AP cell models. Cell viability and apoptosis were measured by Cell Counting Kit 8 assay and flow cytometry. Levels of inflammatory factors and oxidative stress-related markers were assessed. The medium of AR42J cells was collected for coculturing RAW264.7 cells. Macrophage marker CD86+ cell rates were checked with flow cytometry. The levels of TRAF6, embryonic lethal abnormal visual-like protein 1 (ELAVL1), and inducible nitric oxide synthase (iNOS) were examined by Western blot or quantitative real-time polymerase chain reaction. RNA immunoprecipitation assay was performed to evaluate the interaction between ELAVL1 and TRAF6. TRAF6 mRNA stability was tested using actinomycin D treatment. Caerulein treatment suppressed viability, induced AR42J cell apoptosis, inflammation, oxidative stress, and accelerated macrophage M1 polarization. TRAF6 downregulation could alleviate caerulein-induced AR42J cell injury and macrophage M1 polarization. ELAVL1 interacted with TRAF6 to stabilize its expression. Meanwhile, ELAVL1 knockdown relieved caerulein-induced AR42J cell injury and macrophage M1 polarization, while these effects were abolished by TRAF6 overexpression. TRAF6, stabilized by ELAVL1, promoted caerulein-induced AR42J cell injury and macrophage M1 polarization, suggesting that it might accelerate AP9 progression.
Collapse
Affiliation(s)
- Wenyong Zhou
- Department of General Surgery, Cangzhou Central Hospital Cangzhou, Cangzhou, China
| | - Xin Wang
- Department of General Surgery, Cangzhou Hospital of Integrated TCM-WM·Hebei, Cangzhou, China
| | - Bin Yan
- Department of Ultrasound, Cangzhou Central Hospital Cangzhou, Cangzhou, China
| | - Yue Sun
- Department of General Surgery, Cangzhou Hospital of Integrated TCM-WM·Hebei, Cangzhou, China
| |
Collapse
|
7
|
Ren J, Zhao S, Lai J. Role and mechanism of COL3A1 in regulating the growth, metastasis, and drug sensitivity in cisplatin-resistant non-small cell lung cancer cells. Cancer Biol Ther 2024; 25:2328382. [PMID: 38530094 DOI: 10.1080/15384047.2024.2328382] [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: 12/05/2023] [Accepted: 03/05/2024] [Indexed: 03/27/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is among the most difficult malignancies to treat. Type III collagen (COL3A1) can affect the progression and chemoresistance development of NSCLC. We herein explored the mechanism that drives COL3A1 dysregulation in NSCLC. Potential RNA-binding proteins (RBPs) and transcription factors (TFs) that could bind to COL3A1 were searched by bioinformatics. mRNA expression was detected by quantitative PCR. Protein expression was evaluated using immunoblotting and immunohistochemistry. The effects of the variables were assessed by gauging cell growth, invasiveness, migratory capacity, apoptosis, and cisplatin (DDP) sensitivity. The direct YY1/COL3A1 relationship was confirmed by ChIP and luciferase reporter experiments. Xenograft experiments were done to examine COL3A1's function in DDP efficacy. COL3A1 showed enhanced expression in DDP-resistant NSCLC. In H460/DDP and A549/DDP cells, downregulation of COL3A1 exerted inhibitory functions in cell growth, invasiveness, and migration, as well as promoting effects on cell DDP sensitivity and apoptosis. Mechanistically, ELAV-like RNA binding protein 1 (ELAVL1) enhanced the mRNA stability and expression of COL3A1, and Yin Yang 1 (YY1) promoted the transcription and expression of COL3A1. Furthermore, upregulation of COL3A1 reversed ELAVL1 inhibition- or YY1 deficiency-mediated functions in DDP-resistant NSCLC cells. Additionally, COL3A1 downregulation enhanced the anti-tumor efficacy of DDP in vivo. Our investigation demonstrates that COL3A1 upregulation, induced by both RBP ELAVL1 and TF YY1, exerts important functions in phenotypes of NSCLC cells with DDP resistance, offering an innovative opportunity in the treatment of drug-resistant NSCLC.
Collapse
Affiliation(s)
- Jiankun Ren
- Nursing School, Hebi Polytechnic, Hebi City, China
| | - Songwei Zhao
- Nursing School, Hebi Polytechnic, Hebi City, China
| | - Junyu Lai
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Chen X, Han Q, Song J, Pu Y. Identification and validation of a novel defined stress granule-related gene signature for predicting the prognosis of ovarian cancer via bioinformatics analysis. Medicine (Baltimore) 2024; 103:e40608. [PMID: 39809219 PMCID: PMC11596697 DOI: 10.1097/md.0000000000040608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/01/2024] [Indexed: 01/16/2025] Open
Abstract
Ovarian cancer (OC) is a malignant gynecological cancer with an extremely poor prognosis. Stress granules (SGs) are non-membrane organelles that respond to stressors; however, the correlation between SG-related genes and the prognosis of OC remains unclear. This systematic analysis aimed to determine the expression levels of SG-related genes between high- and low-risk groups of patients with OC and to explore the prognostic value of these genes. RNA-sequencing data and clinical information from GSE18520 and GSE14407 in the Gene Expression Omnibus (GEO) and ovarian plasmacytoma adenocarcinoma in The Cancer Genome Atlas (TCGA) were downloaded. SG-related genes were obtained from GeneCards, the Molecular Signatures Database, and the literature. First, 13 SG-related genes were identified in the prognostic model using least absolute shrinkage and selection operator (LASSO) Cox regression. The prognostic value of each SG-related gene for survival and its relationship with clinical characteristics were evaluated. Next, we performed a functional enrichment analysis of SG-related genes. The protein-protein interactions (PPI) of SG-related genes were visualized using Cytoscape with STRING. According to the median risk score from the LASSO Cox regression, a 13-gene signature was created. All patients with OC in TCGA cohort and GEO datasets were classified into high- and low-risk groups. Five SG-related genes were differentially expressed between the high- and low-risk OC groups in the GEO datasets. The 13 SG-related genes were related to several important oncogenic pathways (TNF-α signaling, PI3K-AKT-mTOR signaling, and WNT-β-catenin signaling) and several cellular components (cytoplasmic stress granule, cytoplasmic ribonucleoprotein granule, and ribonucleoprotein granule). The PPI network identified 11 hub genes with the strongest interactions with ELAVL1. These findings indicate that SG-related genes (DNAJA1, ELAVL1, FBL, GRB7, MOV10, PABPC3, PCBP2, PFN1, RFC4, SYNCRIP, USP10, ZFP36, and ZFP36L1) can be used to predict OC prognosis.
Collapse
Affiliation(s)
- Xiaoqi Chen
- Department of Gynecology, Affiliated Hospital of Qinghai University, Xining, China
| | - Qianqian Han
- Department of Colorectal and Anal Surgery, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Jing Song
- Department of Gynecologic Oncology, Affiliated Hospital of Qinghai University, Xining, China
| | - Yongqiang Pu
- Department of Gastrointestinal Oncology, Affiliated Hospital of Qinghai University, Xining, China
| |
Collapse
|
9
|
Patke R, Harris AE, Woodcock CL, Thompson R, Santos R, Kumari A, Allegrucci C, Archer N, Gudas LJ, Robinson BD, Persson JL, Fray R, Jeyapalan J, Rutland CS, Rakha E, Madhusudan S, Emes RD, Muyangwa-Semenova M, Alsaleem M, de Brot S, Green W, Ratan H, Mongan NP, Lothion-Roy J. Epitranscriptomic mechanisms of androgen signalling and prostate cancer. Neoplasia 2024; 56:101032. [PMID: 39033689 PMCID: PMC11295630 DOI: 10.1016/j.neo.2024.101032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Prostate cancer (PCa) is the second most common cancer diagnosed in men. While radical prostatectomy and radiotherapy are often successful in treating localised disease, post-treatment recurrence is common. As the androgen receptor (AR) and androgen hormones play an essential role in prostate carcinogenesis and progression, androgen deprivation therapy (ADT) is often used to deprive PCa cells of the pro-proliferative effect of androgens. ADTs act by either blocking androgen biosynthesis (e.g. abiraterone) or blocking AR function (e.g. bicalutamide, enzalutamide, apalutamide, darolutamide). ADT is often effective in initially suppressing PCa growth and progression, yet emergence of castrate-resistant PCa and progression to neuroendocrine-like PCa following ADT are major clinical challenges. For this reason, there is an urgent need to identify novel approaches to modulate androgen signalling to impede PCa progression whilst also preventing or delaying therapy resistance. The mechanistic convergence of androgen and epitranscriptomic signalling offers a potential novel approach to treat PCa. The epitranscriptome involves covalent modifications of mRNA, notably, in the context of this review, the N(6)-methyladenosine (m6A) modification. m6A is involved in the regulation of mRNA splicing, stability, and translation, and has recently been shown to play a role in PCa and androgen signalling. The m6A modification is dynamically regulated by the METTL3-containing methyltransferase complex, and the FTO and ALKBH5 RNA demethylases. Given the need for novel approaches to treat PCa, there is significant interest in new therapies that target m6A that modulate AR expression and androgen signalling. This review critically summarises the potential benefit of such epitranscriptomic therapies for PCa patients.
Collapse
Affiliation(s)
- Rodhan Patke
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Anna E Harris
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Corinne L Woodcock
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Rachel Thompson
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Rute Santos
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Amber Kumari
- Biodiscovery Institute, University of Nottingham, UK
| | - Cinzia Allegrucci
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Nathan Archer
- School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Brian D Robinson
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Jenny L Persson
- Department of Molecular Biology, Umea University, Umea, Sweden
| | - Rupert Fray
- School of Biosciences, University of Nottingham, UK
| | - Jennie Jeyapalan
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Catrin S Rutland
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK
| | - Emad Rakha
- School of Medicine, University of Nottingham, UK; Nottingham University NHS Trust, Nottingham, UK
| | - Srinivasan Madhusudan
- School of Medicine, University of Nottingham, UK; Nottingham University NHS Trust, Nottingham, UK
| | - Richard D Emes
- Research and Innovation, Nottingham Trent University, UK
| | | | - Mansour Alsaleem
- Biodiscovery Institute, University of Nottingham, UK; Unit of Scientific Research, Applied College, Qassim University, Qassim, Saudi Arabia
| | - Simone de Brot
- Institute of Animal Pathology, University of Bern, Switzerland
| | - William Green
- Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Hari Ratan
- Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Nigel P Mongan
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
| | - Jennifer Lothion-Roy
- Biodiscovery Institute, University of Nottingham, UK; School of Veterinary Medicine and Science, University of Nottingham, UK.
| |
Collapse
|
10
|
Qu Y, Gao N, Zhang S, Gao L, He B, Wang C, Gong C, Shi Q, Li Z, Yang S, Xiao Y. Role of N6-methyladenosine RNA modification in cancer. MedComm (Beijing) 2024; 5:e715. [PMID: 39252821 PMCID: PMC11381670 DOI: 10.1002/mco2.715] [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: 03/17/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
N6-methyladenosine (m6A) is the most abundant modification of RNA in eukaryotic cells. Previous studies have shown that m6A is pivotal in diverse diseases especially cancer. m6A corelates with the initiation, progression, resistance, invasion, and metastasis of cancer. However, despite these insights, a comprehensive understanding of its specific roles and mechanisms within the complex landscape of cancer is still elusive. This review begins by outlining the key regulatory proteins of m6A modification and their posttranslational modifications (PTMs), as well as the role in chromatin accessibility and transcriptional activity within cancer cells. Additionally, it highlights that m6A modifications impact cancer progression by modulating programmed cell death mechanisms and affecting the tumor microenvironment through various cancer-associated immune cells. Furthermore, the review discusses how microorganisms can induce enduring epigenetic changes and oncogenic effect in microorganism-associated cancers by altering m6A modifications. Last, it delves into the role of m6A modification in cancer immunotherapy, encompassing RNA therapy, immune checkpoint blockade, cytokine therapy, adoptive cell transfer therapy, and direct targeting of m6A regulators. Overall, this review clarifies the multifaceted role of m6A modification in cancer and explores targeted therapies aimed at manipulating m6A modification, aiming to advance cancer research and improve patient outcomes.
Collapse
Affiliation(s)
- Yi Qu
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Nannan Gao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Shengwei Zhang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Limin Gao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Bing He
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Chao Wang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Chunli Gong
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Qiuyue Shi
- Department of Gastroenterology the First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Zhibin Li
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Shiming Yang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Yufeng Xiao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| |
Collapse
|
11
|
Rajan AAN, Hutchins EJ. Post-transcriptional regulation as a conserved driver of neural crest and cancer-cell migration. Curr Opin Cell Biol 2024; 89:102400. [PMID: 39032482 PMCID: PMC11346372 DOI: 10.1016/j.ceb.2024.102400] [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: 05/02/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024]
Abstract
Cells have evolved mechanisms to migrate for diverse biological functions. A process frequently deployed during metazoan cell migration is the epithelial-mesenchymal transition (EMT). During EMT, adherent epithelial cells undergo coordinated cellular transitions to mesenchymalize and reduce their intercellular attachments. This is achieved via tightly regulated changes in gene expression, which modulates cell-cell and cell-matrix adhesion to allow movement. The acquisition of motility and invasive properties following EMT allows some mesenchymal cells to migrate through complex environments to form tissues during embryogenesis; however, these processes may also be leveraged by cancer cells, which often co-opt these endogenous programs to metastasize. Post-transcriptional regulation is now emerging as a major conserved mechanism by which cells modulate EMT and migration, which we discuss here in the context of vertebrate development and cancer.
Collapse
Affiliation(s)
- Arvind Arul Nambi Rajan
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Erica J Hutchins
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
12
|
Lv K, Xie P, Yang Q, Luo M, Li C. hsa_circ_0101050 regulated by ZC3H13 enhances tumorigenesis in papillary thyroid cancer via m 6A modification. Heliyon 2024; 10:e32913. [PMID: 38988519 PMCID: PMC11234023 DOI: 10.1016/j.heliyon.2024.e32913] [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: 03/05/2024] [Revised: 05/23/2024] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
While the regulatory roles of circular RNAs (circRNAs) and zinc finger CCCH-type containing 13 (ZC3H13) were previously reported in various human cancers, the mechanisms underlying their interaction in papillary thyroid cancer (PTC) remain unclear. We aimed to determine the role of hsa_circ_0101050 and its regulatory relationship with ZC3H13 in PTC. The expression levels of hsa_circ_0101050 and ZC3H13 were determined in tumor samples and adjacent normal tissues from 46 patients with PTC and in two PTC cell lines (IHH-4 and PTC-1) using quantitative reverse transcription-polymerase chain reaction. The roles of hsa_circ_0101050 and ZC3H13 in cell viability, wound healing, and migration were determined using knockdown and overexpression approaches in PTC cell lines, and a xenograft model in nude mice was used to determine their role in vivo. Methylated RNA immunoprecipitation assay was used to analyze N6-methyladenosine (m6A) modification of hsa_circ_0101050 by ZC3H13. We found hsa_circ_0101050 overexpression and ZC3H13 downregulation in PTC samples and PTC cell lines. In PTC cell lines, silencing hsa_circ_0101050 reduced cell viability and migration whereas its overexpression promoted an aggressive PTC phenotype. ZC3H13 increased the m6A modification of hsa_circ_0101050 and repressed its expression. ZC3H13 overexpression inhibited PTC cell viability, migration, and invasion, which were reversed in cells overexpressing hsa_circ_0101050. Taken together, these results suggested that the downregulation of hsa_circ_0101050 mediated by ZC3H13 through m6A modification contributed to its oncogenic effect in PTC development, revealing the ZC3H13-m6A-hsa_circ_0101050 as a potential therapeutic target in PTC.
Collapse
Affiliation(s)
- Kun Lv
- Department of Tradition Chinese Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Ping Xie
- Department of Tradition Chinese Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Qian Yang
- Department of Tradition Chinese Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Meng Luo
- Department of Tradition Chinese Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, Hubei, China
| | - Chan Li
- Department of Tradition Chinese Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan 430060, Hubei, China
| |
Collapse
|
13
|
Mehmood R. Ramifications of m6A Modification on ncRNAs in Cancer. Curr Genomics 2024; 25:158-170. [PMID: 39087001 PMCID: PMC11288162 DOI: 10.2174/0113892029296712240405053201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 08/02/2024] Open
Abstract
N6-methyladenosine (m6A) is an RNA modification wherein the N6-position of adenosine is methylated. It is one of the most prevalent internal modifications of RNA and regulates various aspects of RNA metabolism. M6A is deposited by m6A methyltransferases, removed by m6A demethylases, and recognized by reader proteins, which modulate splicing, export, translation, and stability of the modified mRNA. Recent evidence suggests that various classes of non- coding RNAs (ncRNAs), including microRNAs (miRNAs), circular RNAs (circRNAs), and long con-coding RNAs (lncRNAs), are also targeted by this modification. Depending on the ncRNA species, m6A may affect the processing, stability, or localization of these molecules. The m6A- modified ncRNAs are implicated in a number of diseases, including cancer. In this review, the author summarizes the role of m6A modification in the regulation and functions of ncRNAs in tumor development. Moreover, the potential applications in cancer prognosis and therapeutics are discussed.
Collapse
Affiliation(s)
- Rashid Mehmood
- Department of Life Sciences, College of Science and General Studies, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
14
|
Feng C, Tian Q, Tang X, Yu J, Li H, Geng C, Xu L. microRNA-9a-5p disrupts the ELAVL1/VEGF axis to alleviate traumatic brain injury. Exp Neurol 2024; 375:114721. [PMID: 38342180 DOI: 10.1016/j.expneurol.2024.114721] [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: 05/29/2023] [Revised: 08/07/2023] [Accepted: 02/08/2024] [Indexed: 02/13/2024]
Abstract
Plasma microRNA (miR)-9 has been identified as a promising diagnostic biomarker for traumatic brain injury (TBI). This study aims to investigate the possible role and mechanisms of miR-9a-5p affecting TBI. Microarray-based gene expression profiling of TBI was used for screening differentially expressed miRNAs and genes. TBI rat models were established. miR-9a-5p, ELAVL1 and VEGF expression in the brain tissue of TBI rats was detected. The relationship among miR-9a-5p, ELAVL1 and VEGF was tested. TBI modeled rats were injected with miR-9a-5p-, ELAVL1 or VEGF-related sequences to identify their effects on TBI. miR-9a-5p was poorly expressed in the brain tissue of rats with TBI. ELAVL1 was a downstream target gene of miR-9a-5p, which could negatively regulate its expression. Enforced miR-9a-5p expression prevented brain tissue damage in TBI rats by targeting ELAVL1. Meanwhile, ELAVL1 could increase the expression of VEGF, which was highly expressed in the brain tissue of rats with TBI. In addition, ectopically expressed miR-9a-5p alleviated brain tissue damage in TBI rats by downregulating the ELAVL1/VEGF axis. Overall, miR-9a-5p can potentially reduce brain tissue damage in TBI rats by targeting ELAVL1 and down-regulating VEGF expression.
Collapse
Affiliation(s)
- Chenxi Feng
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215000, PR China
| | - Qiuyan Tian
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou 215000, PR China
| | - Xiaojuan Tang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou 215000, PR China
| | - Jian Yu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou 215000, PR China
| | - Hong Li
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou 215000, PR China
| | - Changxing Geng
- School of Mechanical and Electrical Engineering, Soochow University, Suzhou 215000, PR China.
| | - Lixiao Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215000, PR China.
| |
Collapse
|
15
|
Wang X, Liu D, Hua K, Fang L. LncRNA HOST2 promotes NSUN2-mediated breast cancer progression via interaction with ELAVL1. Cell Signal 2024; 117:111112. [PMID: 38387687 DOI: 10.1016/j.cellsig.2024.111112] [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: 11/30/2023] [Revised: 02/09/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Breast cancer (BC) is the most prevalent malignant tumor in women worldwide with high morbidity and mortality. NSUN2, a crucial RNA methyltransferase, plays a pivotal role in regulating the proliferation and metastasis of tumor cells. Our study demonstrated that NSUN2 is upregulated in BC tissues and cell lines, and its high expression is associated with a poor prognosis in BC patients. Knockout of NSUN2 exerted inhibitory effects on the proliferation and migration of BC cells in vitro and in vivo. Mechanistic investigations revealed that the RNA-binding protein ELAVL1 can bind to NSUN2 mRNA and increase its stability. Additionally, we identified HOST2, a long non-coding RNA, as a key player in blocking the ubiquitin-dependent proteasomal degradation of ELAVL1, thereby influencing the stability of NSUN2 mRNA. In conclusion, this study revealed for the first time that HOST2 maintains NSUN2 mRNA stability by blocking ubiquitin-dependent degradation of ELAVL1, which in turn affects BC progression. HOST2/ELAVL1/NSUN2 oncogenic cascade has the potential to be a novel therapeutic target for BC.
Collapse
Affiliation(s)
- Xuehui Wang
- Department of Thyroid and Breast Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Institute of Breast Disease, School of Medicine, Tongji University, Shanghai 200072, China
| | - Diya Liu
- Department of Thyroid and Breast Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Institute of Breast Disease, School of Medicine, Tongji University, Shanghai 200072, China
| | - Kaiyao Hua
- Department of Thyroid and Breast Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Institute of Breast Disease, School of Medicine, Tongji University, Shanghai 200072, China
| | - Lin Fang
- Department of Thyroid and Breast Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Institute of Breast Disease, School of Medicine, Tongji University, Shanghai 200072, China.
| |
Collapse
|
16
|
Han Z, Yi X, Li J, Zhang T, Liao D, You J, Ai J. RNA m 6A modification in prostate cancer: A new weapon for its diagnosis and therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:188961. [PMID: 37507057 DOI: 10.1016/j.bbcan.2023.188961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/21/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
Prostate cancer (PCa) is the most common malignant tumor and the second leading cause of cancer-related mortality in men worldwide. Despite significant advances in PCa therapy, the underlying molecular mechanisms have yet to be fully elucidated. Recently, epigenetic modification has emerged as a key player in tumor progression, and RNA-based N6-methyladenosine (m6A) epigenetic modification was found to be crucial. This review summarizes comprehensive state-of-art mechanisms underlying m6A modification, its implication in the pathogenesis, and advancement of PCa in protein-coding and non-coding RNA contexts, its relevance to PCa immunotherapy, and the ongoing clinical trials for PCa treatment. This review presents potential m6A-based targets and paves a new avenue for diagnosing and treating PCa, providing new guidelines for future related research through a systematic review of previous results.
Collapse
Affiliation(s)
- Zeyu Han
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Xianyanling Yi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jin Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Tianyi Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Dazhou Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jia You
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu 610041, China.
| |
Collapse
|