1
|
Zhang L, Yuan J, Yao S, Wen G, An J, Jin H, Tuo B. Role of m5C methylation in digestive system tumors (Review). Mol Med Rep 2025; 31:142. [PMID: 40183387 PMCID: PMC11979572 DOI: 10.3892/mmr.2025.13507] [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: 11/26/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Currently, the incidence of digestive system tumors has been increasing annually, thus becoming a prevalent cause of cancer‑related mortalities. Although significant strides have been made in targeting the molecular mechanisms that underpin the development of these tumors, their treatment and prognosis still pose substantial challenges. This is primarily due to the ambiguity of early diagnostic indicators and the fact that most digestive system tumors are detected at an advanced stage. However, epigenetic modifications are capable of altering the expression of oncogenes and regulating biological processes in cancer. In recent years, the study of methylation in relation to tumor pathogenesis has become a focus of prominent research. Among the various types of methylation, 5‑methylcytosine (m5C) methylation plays a crucial role in the development of digestive system tumors and is anticipated to serve as a novel therapeutic target. However, to date, a comprehensive and systematic review concerning the role of m5C methylation in digestive system tumors is lacking. Consequently, the present study reviewed the role of m5C methylation in digestive system tumors such as esophageal cancer, gastric cancer and hepatocellular carcinoma, with the aim of providing a valuable reference for future research endeavors.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jianbo Yuan
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P.R. China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
2
|
Su L, Xu R, Ren Y, Zhao S, Song L, Meng C, Liu W, Zhou X, Du Z. 5-Methylcytosine methylation predicts cervical cancer prognosis, shaping immune cell infiltration. J Int Med Res 2025; 53:3000605251328301. [PMID: 40219803 DOI: 10.1177/03000605251328301] [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] [Indexed: 04/14/2025] Open
Abstract
BackgroundEpigenetics, encompassing DNA and RNA modifications, has emerged as a prominent area of research in the post-genomic era. Numerous studies have elucidated the impact of epigenetics on tumor regulation. However, the methylation patterns of 5-methylcytosine in cervical cancer as well as its role in the tumor microenvironment and immunotherapy remain poorly understood.MethodsUtilizing a comprehensive dataset encompassing samples from 306 patients with cervical cancer from The Cancer Genome Atlas and Gene Expression Omnibus repositories, we conducted an in-depth analysis to evaluate the potential association between the modification patterns of 5-methylcytosine and the infiltration of cells within the tumor microenvironment, taking into account 11 regulators of 5-methylcytosine modification. Subsequently, we employed stepwise regression and Least Absolute Shrinkage and Selection Operator Cox regression to quantify 5-methylcytosine modification patterns in patients with cervical squamous cell carcinoma and endocervical adenocarcinoma, yielding the 5-methylcytosine score. Our study explored the link between the 5-methylcytosine score and clinical characteristics as well as prognostic outcomes in patients with cervical squamous cell carcinoma and endocervical adenocarcinoma.ResultsA comprehensive analysis of 306 patients with cervical cancer revealed two distinct 5-methylcytosine modification patterns, henceforth labeled as 5-methylcytosine clusters A and B. These clusters exhibited distinct immunological profiles and biological attributes, with 5-methylcytosine cluster A exhibiting a higher degree of immune cell infiltration. Utilizing univariate Cox regression analysis, we identified 367 genes regulated by 5-methylcytosine that were significantly correlated with patient prognosis. This analysis further stratified the samples into three distinct genomic subtypes. Survival analyses indicated that patients belonging to gene cluster C exhibited more favorable survival outcomes than those belonging to gene clusters A and B. Intriguingly, most 5-methylcytosine regulatory factors had higher expression levels in gene cluster B than in gene cluster A. Gene set enrichment analysis of a single sample revealed elevated immune cell infiltration within gene cluster B, indicating a stronger immune response in this cluster. The 5-methylcytosine score feature was utilized to determine the 5-methylcytosine modification pattern in cervical cancer, revealing that patients with low 5-methylcytosine scores exhibited better survival rates, whereas those with high scores had increased mutation frequencies and better treatment responses.ConclusionsThis research underscores the key role of 5-methylcytosine modification patterns in cervical cancer. Analysis of these patterns will deepen our understanding of the cervical cancer tumor microenvironment, paving the way for the development of more refined and effective immunotherapy strategies.
Collapse
Affiliation(s)
- Luyang Su
- Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Ren Xu
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Yanan Ren
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Shixia Zhao
- Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Liyun Song
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Cuiqiao Meng
- Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Weilan Liu
- Physical Examination Center, Hebei General Hospital, Shijiazhuang, Hebei-China
| | - Xuan Zhou
- Department of Obstetrics and Gynecology, Affiliated Hospital of Chengde Medical University, Chengde, Hebei-China
| | - Zeqing Du
- Department of Obstetrics and Gynecology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei-China
| |
Collapse
|
3
|
Hu Y, Chen C, Lin K, Tong X, Huang T, Qiu T, Chen X, Xu J, Xie W, Sun X, Feng S, Lu M, Zhao Z, Chen X, Xue X, Shen X. NSUN2 promotes colorectal cancer progression and increases lapatinib sensitivity by enhancing CUL4B/ErbB-STAT3 signalling in a non-m5C manner. Clin Transl Med 2025; 15:e70282. [PMID: 40156167 PMCID: PMC11953055 DOI: 10.1002/ctm2.70282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025] Open
Abstract
NSUN2, a major methyltransferase that catalyzes m5C methylation in eukaryotes, is known to be implicated in the development of multiple cancers. However, its role in colorectal cancer (CRC) and the related molecular mechanisms have yet to be sufficiently determined. Here, we conducted an analysis of public database (722 CRC patients) and two distinct cohorts from our centre (1559 CRC patients), which revealed that NSUN2 is upregulated in CRC and correlates with unfavourable prognosis. Our analyses also showed that NSUN2 promotes the proliferation and metastasis capabilities of CRC cells. Intriguingly, NSUN2 was found to promote CRC via an m5C-independent mechanism, which has not been previously reported. Overexpression of both wild-type and m5C enzymatic-dead mutant NSUN2 upregulated and activated the ErbB-STAT3 signalling pathway. We also found that both wild-type and the m5C enzymatic-dead mutant NSUN2 closely interacted with CUL4B. Silencing of CUL4B effectively inhibited the m5C-independent function of NSUN2. Moreover, overexpression of NSUN2 enhanced the sensitivity of CRC cells to lapatinib. Taken together, our findings revealed a novel m5C-independent mechanism for NSUN2 in the malignancy and lapatinib sensitivity of CRC via activation of the CUL4B/ErbB-STAT3 pathway, which provides a potential therapeutic strategy for patients with CRC. HIGHLIGHTS: NSUN2 is upregulated in CRC and associated with poor prognosis of CRC patients. NSUN2 promotes CRC malignancy independently of its m5C-enzymatic activity, a mechanism that has not been previously reported. The non-m5C carcinogenic roles of NSUN2 may be mediated through interactions with CUL4B, thereby activating the ErbB-STAT3 signalling pathway. NSUN2-mediated upregulation of ErbB-STAT3 pathway enhances the sensitivity of CRC to lapatinib treatment.
Collapse
Affiliation(s)
- Yuanbo Hu
- Department of General SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of General SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
| | - Chenbin Chen
- Department of General SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Kezhi Lin
- Experiemtial Center of Basic Medicine, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
| | - Xinya Tong
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
| | - Tingting Huang
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
- Department of Obstetrics and Gynecology, Maternal and Child Care Service HospitalYueqingChina
| | - Tianle Qiu
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
| | - Xietao Chen
- Department of General SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
| | - Jun Xu
- Department of General SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
| | - Wangkai Xie
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
| | - Xiangwei Sun
- Department of General SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Shiyu Feng
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
| | - Mingdong Lu
- Department of General SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Zhiguang Zhao
- Department of PathologyThe Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiaodong Chen
- Department of General SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiangyang Xue
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical SciencesWenzhou Medical UniversityWenzhouChina
| | - Xian Shen
- Department of General SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Department of General SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
4
|
Ranga S, Yadav R, Chauhan M, Chhabra R, Ahuja P, Balhara N. Modifications of RNA in cancer: a comprehensive review. Mol Biol Rep 2025; 52:321. [PMID: 40095076 DOI: 10.1007/s11033-025-10419-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
RNA modifications play essential roles in post-transcriptional gene regulation and have emerged as significant contributors to cancer biology. Major chemical modifications of RNA include N6-methyladenosine (m6A), 5-methylcytosine (m5C), N1-methyladenosine (m1A), pseudouridine (ψ), and N7-methylguanosine (m7G). Their dynamic regulation highlights their roles in gene expression modulation, RNA stability, and translation. Advanced high-throughput detection methods, ranging from liquid chromatography-mass spectrometry and high-performance liquid chromatography to next-generation sequencing (NGS) and nanopore direct RNA sequencing, have enabled detailed studies of RNA modifications in cancer cells. Aberrant RNA modifications are associated with the dysregulation of tumor suppressor genes and oncogenes, influencing cancer progression, therapy resistance, and immune evasion. Emerging research suggests the therapeutic potential of targeting RNA-modifying enzymes and their inhibitors in cancer treatment. This review compiles and analyzes the latest findings on RNA modifications, presenting an in-depth discussion of the diverse chemical alterations that occur in RNA and their profound implications in cancer biology. It integrates fundamental principles with cutting-edge research, offering a holistic perspective on how RNA modifications influence gene expression, tumor progression, and therapeutic resistance. It emphasizes the need for further studies to elucidate the complex roles of RNA modifications in cancer, as well as the potential for multimodality therapeutic strategies that exploit the dynamic and reversible nature of these epitranscriptomic marks. It also attempts to highlight the challenges, gaps, and limitations of RNA modifications in cancer that should be tackled before their functional implications. Understanding the interplay between RNA modifications, cancer pathways, and their inhibitors will be crucial for developing promising RNA-based therapeutic approaches to cancer and personalized medicine strategies.
Collapse
Affiliation(s)
- Shalu Ranga
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Ritu Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| | - Meenakshi Chauhan
- Department of Obstetrics and Gynaecology, Pandit Bhagwat Dayal Sharma University of Health Sciences, Rohtak, Haryana, 124001, India
| | - Ravindresh Chhabra
- Department of Biochemistry, Central University of Panjab, Bathinda, Panjab, 151401, India
| | - Parul Ahuja
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Nikita Balhara
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| |
Collapse
|
5
|
Hu X, Liu Y, Zhang S, Liu K, Gu X. The multifaceted role of m5C RNA methylation in digestive system tumorigenesis. Front Cell Dev Biol 2025; 13:1533148. [PMID: 40114967 PMCID: PMC11922842 DOI: 10.3389/fcell.2025.1533148] [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: 11/23/2024] [Accepted: 02/05/2025] [Indexed: 03/22/2025] Open
Abstract
5-Methylcytosine (m5C) is a widespread RNA methylation modification, wherein a methyl group is enzymatically transferred to specific RNA sites by methyltransferases, such as the NSUN family and DNMT2. The m5C modification not only impacts RNA structure and stability but also governs post-transcriptional regulation by influencing RNA transport, translation, and protein interactions. Recently, the functional importance of m5C in complex diseases, including cancer, has gained substantial attention. Increasing evidence highlights the critical roles of m5C in digestive system malignancies, where it contributes to tumor progression by modulating oncogene expression and regulating processes such as tumor cell proliferation, migration, invasion, and resistance to chemotherapy. Furthermore, m5C's involvement in non-coding RNAs reveals additional dimensions in elucidating their roles in cancer. This review summarizes recent advances in m5C RNA methylation research within digestive system tumors, focusing on its functional mechanisms, clinical significance, and potential applications. Specifically, it aims to explore m5C's role in tumor diagnosis, prognosis, and treatment, while proposing future directions to address current challenges and broaden its clinical utility.
Collapse
Affiliation(s)
- Xinjun Hu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yafeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Shujun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Kaijie Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| |
Collapse
|
6
|
Yang Y, Cao L, Xu X, Li D, Deng Y, Li L, Zeng B, Jiang H, Shan L, Huang Y, Xu Y, Ma L. NSUN2/ALYREF axis-driven m 5C methylation enhances PD-L1 expression and facilitates immune evasion in non-small-cell lung cancer. Cancer Immunol Immunother 2025; 74:132. [PMID: 40029463 PMCID: PMC11876480 DOI: 10.1007/s00262-025-03986-5] [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: 12/13/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
Non-small-cell lung cancer (NSCLC) represents a highly prevalent form of malignancy. 5-methylcytosine (m5C) methylation functions as a key post-transcriptional regulatory mechanism linked to cancer progression. The persistent expression of PD-L1 in tumor cells plays a pivotal role in facilitating immune evasion and promoting T-cell exhaustion. However, the involvement of m5C in NSCLC immune evasion remains inadequately understood. This study seeks to explore the function of the m5C methyltransferase NSUN2 in modulating PD-L1 expression and facilitating immune evasion in NSCLC. Our findings indicate elevated levels of NSUN2 and ALYREF in NSCLC, and both promote the growth of NSCLC cells and the progression of lung cancer. Moreover, the expression of PD-L1 in NSCLC tissues positively correlates with NSUN2 and ALYREF expression. We then discovered that PD-L1 acts as a downstream target of NSUN2-mediated m5C modification in NSCLC cells. Knocking down NSUN2 significantly reduces m5C modification of PD-L1 mRNA, thereby decreasing its stability via the m5C reader ALYREF-dependent manner. Furthermore, inhibiting NSUN2 enhanced CD8+ T-cell activation and infiltration mediated by PD-L1, thereby boosting antitumor immunity, as confirmed in both in vitro and in vivo experiments. Collectively, these results suggested that NSUN2/ALYREF/PD-L1 axis plays a critical role in promoting NSCLC progression and tumor cell immune suppression, highlighting its potential as a novel therapeutic strategy for NSCLC immunotherapy.
Collapse
Affiliation(s)
- Yiran Yang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Leiqun Cao
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Xin Xu
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Dan Li
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Yiran Deng
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Lan Li
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Bingjie Zeng
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Haixia Jiang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Liang Shan
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Yiwen Huang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Yunhua Xu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China.
| | - Lifang Ma
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241 West Huaihai Road, Shanghai, 200030, China.
| |
Collapse
|
7
|
Zhang L, Li Y, Li L, Yao F, Cai M, Ye D, Qu Y. Detection, molecular function and mechanisms of m5C in cancer. Clin Transl Med 2025; 15:e70239. [PMID: 40008496 PMCID: PMC11862898 DOI: 10.1002/ctm2.70239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Interest in RNA posttranscriptional modifications, particularly 5-methylcytosine (m5C), has surged in recent years. Studies have shown that m5C plays a key role in cellular processes and is closely linked to tumourigenesis. This growing focus emphasises the importance of understanding the diverse impacts of m5C modifications in both normal cellular functions and cancer development. Moreover, strides in methodologies for discerning m5C have facilitated intricate transcriptome cartography of RNA methylation at the solitary nucleotide echelon. This technical progress has fueled a surge in m5C-centric investigations, facilitating further exploration of this RNA modification. This review provides a comprehensive analysis of the oncogenic potential of m5C RNA modification, elucidating the precise molecular mechanisms driving its role in cancer development. It consolidates current knowledge regarding the biological consequences of m5C RNA modification in tumour cells. Understanding the role of methylation-related processes in tumourigenesis shows promise for advancing cancer diagnosis and therapeutic strategies. HIGHLIGHTS: m5C modifications are dynamically regulated by writers, readers, and erasers, influencing cancer progression, metastasis, and immune evasion. Distinct m5C regulatory networks exist across cancers, modulating oncogenic pathways and therapy responses. m5C signatures serve as biomarkers for cancer prognosis and treatment stratification, highlighting their role in precision oncology.
Collapse
Affiliation(s)
- Linhui Zhang
- Department of UrologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Genitourinary Cancer InstituteShanghaiChina
| | - Yuelong Li
- Department of UrologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Liqing Li
- Department of UrologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Genitourinary Cancer InstituteShanghaiChina
| | - Fei Yao
- Department of NursingFudan University Shanghai Cancer CenterShanghaiChina
| | - Maoping Cai
- Department of UrologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Genitourinary Cancer InstituteShanghaiChina
| | - Dingwei Ye
- Department of UrologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Genitourinary Cancer InstituteShanghaiChina
| | - Yuanyuan Qu
- Department of UrologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Genitourinary Cancer InstituteShanghaiChina
| |
Collapse
|
8
|
Peng J, Lv L, Zhou Y, Wang X, Hu C. PHAX enhanced LIN28B-mediated PBX3 mRNA stability to promote esophageal cancer development. Cancer Sci 2025; 116:808-823. [PMID: 39668567 PMCID: PMC11875786 DOI: 10.1111/cas.16420] [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: 06/06/2024] [Revised: 11/05/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024] Open
Abstract
The abnormal expression of PHAX was observed in esophageal cancer, however, its specific function and mechanism remain to be further elucidated. We demonstrated that PHAX, LIN28B, and PBX3 were upregulated in esophageal cancer, while TET2 was downregulated. Elevated PHAX correlated with adverse outcomes among esophageal cancer patients. PHAX or PBX3 knockdown not only inhibited esophageal cancer cell proliferation, and promoted apoptosis and autophagy in vitro, but it also repressed tumor growth and lung metastasis in mice. Mechanically, PHAX stabilized PBX3 mRNA through interacting with LIN28B. PBX3 directly bound to the TET2 promoter region and inhibited its expression. In conclusion, PHAX directly bound to LIN28B and enhanced LIN28B-mediated stabilization of PBX3 mRNA, leading to upregulation of PBX3. PBX3 then transcriptionally repressed TET2 expression to promote esophageal cancer cell proliferation, and suppress apoptosis and autophagy. Targeting this signaling cascade could represent a promising therapeutic strategy for esophageal cancer.
Collapse
Affiliation(s)
- Jie Peng
- Department of HaematologyXiangya Hospital, Central South UniversityChangshaChina
| | - Liang Lv
- Department of Gastroenterology, Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Yuqian Zhou
- Department of Gastroenterology, Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Xuehong Wang
- Department of Gastroenterology, Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Changmei Hu
- Department of Gastroenterology, Second Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
9
|
Qi Q, Zhong R, Huang Y, Tang Y, Zhang XW, Liu C, Gao CF, Zhou L, Yu J, Wu LY. The RNA M5C methyltransferase NSUN2 promotes progression of hepatocellular carcinoma by enhancing PKM2-mediated glycolysis. Cell Death Dis 2025; 16:82. [PMID: 39924557 PMCID: PMC11808121 DOI: 10.1038/s41419-025-07414-5] [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: 12/14/2023] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. The 5-methylcytosine (m5C) RNA methyltransferase NSUN2 is involved in cell proliferation and metastasis and is upregulated in a variety of cancers. However, the biological function and regulatory mechanism of NSUN2-mediated m5C modification have not been well studied in HCC. Our results showed that NSUN2 is upregulated and associated with poor prognosis in HCC patients after hepatectomy. NSUN2 overexpression significantly promoted HCC growth and metastasis, whereas NSUN2 knockdown had the opposite effect. m5C RNA immunoprecipitation sequencing (m5C-RIP-Seq) revealed that m5C hypermethylation correlates with mRNA overexpression and that NSUN2-mediated m5C hypermethylation promotes metabolism in HCC patients. Mechanistically, our data revealed that PKM2, a terminal enzyme in the glycolytic pathway, is a downstream target of NSUN2-mediated m5C modification. Specifically, NSUN2 could stabilize PKM2 mRNA by increasing the m5C level of the m5C site C773 in the 3'-UTR of PKM2 mRNA. In addition, rescue assays revealed that NSUN2 promotes HCC glycolysis and progression by upregulating PKM2. In conclusion, this study revealed that NSUN2-mediated m5C modification promotes glycolysis and the progression of hepatocellular carcinoma by stabilizing PKM2 mRNA, and provides a potential prognostic factor and therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Qin Qi
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui Zhong
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Huang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao-Wen Zhang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang Liu
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chun-Fang Gao
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Lin Zhou
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Jian Yu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
| | - Lu-Yi Wu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
10
|
Tong R, Li Y, Wang J, Liu C, Liu Y, Li R, Wang X. NSUN2 Knockdown Promotes the Ferroptosis of Colorectal Cancer Cells Via m5C Modification of SLC7A11 mRNA. Biochem Genet 2025:10.1007/s10528-025-11035-0. [PMID: 39920526 DOI: 10.1007/s10528-025-11035-0] [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: 10/23/2024] [Accepted: 01/12/2025] [Indexed: 02/09/2025]
Abstract
The high occurrence and death rates of colorectal cancer (CRC) make it a major health concern. Recent studies have identified NOP2/Sun RNA methyltransferase family member 2 (NSUN2), an RNA methyltransferase, as a key regulator in various tumor types. However, how exactly NSUN2-mediated m5C alteration affects CRC is still a mystery. This study seeks to understand how NSUN2 contributes to the growth and death of colorectal cancer cells. New tissue samples were taken in order to investigate NSUN2 expression in CRC. In vitro tests were performed to evaluate NSUN2's function. We used m5C-methylated-RNA immunoprecipitation and RNA stability experiments to find out how NSUN2 works on Solute carrier family 7 member 11 (SLC7A11, also called xCT). Downregulation of NSUN2 limits CRC cell growth and induces ferroptosis, as we show that NSUN2 was substantially expressed in CRC. In terms of the molecular mechanism, NSUN2 controls the translation and stability of SLC7A11 mRNA by regulating its m5C methylation. Functional tests show that SLC7A11 compensates for the NSUN2 knockdown-induced decrease in cell proliferation. Additionally, SLC7A11 overexpression restores ferroptosis to CRC cells after NSUN2 knockdown. These findings emphasize NSUN2's crucial role in modulating colorectal cancer cell growth and survival via SLC7A11, pointing to promising new therapeutic targets.
Collapse
Affiliation(s)
- Ruibing Tong
- Gastrointestinal Surgery, The First Affiliated Hospital of the Baotou Medical College of Inner Mongolia University of Science and Technology, No. 41 Liyin Road, Kundulun District, Baotou, China.
| | - Yuefeng Li
- Gastrointestinal Surgery, The First Affiliated Hospital of the Baotou Medical College of Inner Mongolia University of Science and Technology, No. 41 Liyin Road, Kundulun District, Baotou, China
| | - Junli Wang
- Gastrointestinal Surgery, The First Affiliated Hospital of the Baotou Medical College of Inner Mongolia University of Science and Technology, No. 41 Liyin Road, Kundulun District, Baotou, China
| | - Chengyu Liu
- Gastrointestinal Surgery, The First Affiliated Hospital of the Baotou Medical College of Inner Mongolia University of Science and Technology, No. 41 Liyin Road, Kundulun District, Baotou, China
| | - Yan Liu
- Gastrointestinal Surgery, The First Affiliated Hospital of the Baotou Medical College of Inner Mongolia University of Science and Technology, No. 41 Liyin Road, Kundulun District, Baotou, China
| | - Rongshuang Li
- Gastrointestinal Surgery, The First Affiliated Hospital of the Baotou Medical College of Inner Mongolia University of Science and Technology, No. 41 Liyin Road, Kundulun District, Baotou, China
| | - Xinghong Wang
- Gastrointestinal Surgery, The First Affiliated Hospital of the Baotou Medical College of Inner Mongolia University of Science and Technology, No. 41 Liyin Road, Kundulun District, Baotou, China
| |
Collapse
|
11
|
Yang S, Zheng Y, Zhou C, Yao J, Yan G, Shen C, Kong S, Xiong Y, Sun Q, Sun Y, Shen H, Bian L, Qian K, Liu X. Multidimensional Proteomic Landscape Reveals Distinct Activated Pathways Between Human Brain Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410142. [PMID: 39716938 PMCID: PMC11831486 DOI: 10.1002/advs.202410142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/11/2024] [Indexed: 12/25/2024]
Abstract
Brain metastases (BrMs) and gliomas are two typical human brain tumors with high incidence of mortalities and distinct clinical challenges, yet the understanding of these two types of tumors remains incomplete. Here, a multidimensional proteomic landscape of BrMs and gliomas to infer tumor-specific molecular pathophysiology at both tissue and plasma levels is presented. Tissue sample analysis reveals both shared and distinct characteristics of brain tumors, highlighting significant disparities between BrMs and gliomas with differentially activated upstream pathways of the PI3K-Akt signaling pathway that have been scarcely discussed previously. Novel proteins and phosphosites such as NSUN2, TM9SF3, and PRKCG_S330 are also detected, exhibiting a high correlation with reported clinical traits, which may serve as potential immunohistochemistry (IHC) biomarkers. Moreover, tumor-specific altered phosphosites and glycosites on FN1 are highlighted as potential therapeutic targets. Further validation of 110 potential noninvasive biomarkers yields three biomarker panels comprising a total of 19 biomarkers (including DES, VWF, and COL1A1) for accurate discrimination of two types of brain tumors and normal controls. In summary, this is a full-scale dataset of two typical human brain tumors, which serves as a valuable resource for advancing precision medicine in cancer patients through targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Shuang Yang
- Institute of Translational MedicineShanghai Jiao Tong UniversityShanghai200241P. R. China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032P. R. China
| | - Yongtao Zheng
- Institute of Translational MedicineShanghai Jiao Tong UniversityShanghai200241P. R. China
- Department of NeurosurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200025P. R. China
| | - Chengbin Zhou
- Department of NeurosurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200025P. R. China
| | - Jun Yao
- Institutes of Biomedical SciencesFudan UniversityShanghai200032P. R. China
| | - Guoquan Yan
- Institutes of Biomedical SciencesFudan UniversityShanghai200032P. R. China
| | - Chengpin Shen
- Shanghai Omicsolution Co., Ltd.Shanghai200000P. R. China
| | - Siyuan Kong
- Institutes of Biomedical SciencesFudan UniversityShanghai200032P. R. China
| | - Yueting Xiong
- Institutes of Biomedical SciencesFudan UniversityShanghai200032P. R. China
| | - Qingfang Sun
- Department of NeurosurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200025P. R. China
| | - Yuhao Sun
- Institute of Translational MedicineShanghai Jiao Tong UniversityShanghai200241P. R. China
- Department of NeurosurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200025P. R. China
| | - Huali Shen
- Institutes of Biomedical SciencesFudan UniversityShanghai200032P. R. China
| | - Liuguan Bian
- Institute of Translational MedicineShanghai Jiao Tong UniversityShanghai200241P. R. China
- Department of NeurosurgeryRuijin HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200025P. R. China
| | - Kun Qian
- Institute of Translational MedicineShanghai Jiao Tong UniversityShanghai200241P. R. China
| | - Xiaohui Liu
- Institute of Translational MedicineShanghai Jiao Tong UniversityShanghai200241P. R. China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032P. R. China
| |
Collapse
|
12
|
Lou N, Gu X, Fu L, Li J, Xue C. Significant roles of RNA 5-methylcytosine methylation in cancer. Cell Signal 2025; 126:111529. [PMID: 39615772 DOI: 10.1016/j.cellsig.2024.111529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 12/06/2024]
Abstract
Cancer stands as a leading cause of mortality and poses an escalating threat to global health. Epigenetic dysregulation is pivotal in the onset and advancement of cancer. Recent research on RNA 5-methylcytosine (m5C) methylation has underscored its significant role in cancer. RNA m5C methylation is a key component in gene expression regulation and is intricately linked to cancer development, offering valuable insights for cancer diagnosis, treatment, and prognosis. This review provides an in-depth examination of the three types of regulators associated with RNA m5C methylation and their biological functions. It further investigates the expression and impact of RNA m5C methylation and its regulators in cancer, focusing on their mechanisms in cancer progression and clinical relevance. The current research on inhibitors targeting RNA m5C methylation-related regulators remains underdeveloped, necessitating further exploration and discovery.
Collapse
Affiliation(s)
- Na Lou
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Leiya Fu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
13
|
Zhang Q, Cao Z, He Y, Liu Z, Guo W. Exploration of the mechanism of 5-Methylcytosine promoting the progression of hepatocellular carcinoma. Transl Oncol 2025; 52:102257. [PMID: 39733743 PMCID: PMC11743813 DOI: 10.1016/j.tranon.2024.102257] [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: 06/22/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024] Open
Abstract
5-Methylcytosine (m5C) is a ubiquitous RNA modification that is closely related to various cellular functions. However, no studies have comprehensively demonstrated the role of m5C in hepatocellular carcinoma (HCC) progression. In this study, six pairs of HCC and adjacent tissue samples were subjected to methylated RNA immunoprecipitation sequencing to identify precise m5C loci. Non-negative matrix factorization (NMF) was used to identify HCC subtypes in TCGA-LIHC cohort. Immune, metabolic, and tumor-related pathways in HCC subtypes with differences in methylation status were analyzed and a prognostic model based on m5C-related genes was constructed. Finally, using RIP and molecular interaction analysis, we demonstrated that YBX1 binds to TPM3 in an m5C dependent manner and regulates HCC progression. Widespread m5C sites were identified and found to be differentially distributed in HCC compared with adjacent tissues. Metabolic processes were inhibited in hypermethylated HCC, whereas immune checkpoint and multiple classical tumor pathways were significantly upregulated. More importantly, we have identified an m5C dependent regulatory axis. The m5C reader YBX1 binds to TPM3 in an M5C dependent manner and promotes the progression of hepatocellular carcinoma. These results provide new evidence for further understanding the comprehensive role of m5C in HCC and the regulatory mechanism of m5C.
Collapse
Affiliation(s)
- Qiyao Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; General Surgery Department of Peking Union Medical College Peking Union Medical College Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100730
| | - Zhen Cao
- General Surgery Department of Peking Union Medical College Peking Union Medical College Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100730
| | - Yuting He
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou 450052, China; Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou 450052, China.
| | - Ziwen Liu
- General Surgery Department of Peking Union Medical College Peking Union Medical College Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100730.
| | - Wenzhi Guo
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou 450052, China; Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou 450052, China.
| |
Collapse
|
14
|
Wang R, Qu J, Chen M, Han T, Liu Z, Wang H. NSUN2 knockdown inhibits macrophage infiltration in diabetic nephropathy via reducing N5-methylcytosine methylation of SOCS1. Int Urol Nephrol 2025; 57:643-653. [PMID: 39382603 DOI: 10.1007/s11255-024-04214-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024]
Abstract
OBJECTIVE N5-methylcytosine (m5C) methylation is involved in various disease progression; however, its role in diabetic nephropathy (DN) has not been studied. The aim of this study was to investigate the role of NSUN2 in DN and the underlying mechanism. METHODS Streptozotocin-induced experimental mouse model was generated to analyze the role of NSUN2 in vivo, and high glucose (HG)-treated Raw264.7 cells were used to assess the effect of NSUN2 on macrophage infiltration in vitro. The regulation of NSUN2 on SOCS1 m5C methylation was evaluated using m5C methylated RNA immunoprecipitation, luciferase reporter analysis, and RNA stability determination assay. RESULTS The results indicated that NSUN2 was highly expressed in the blood and kidney of DN mice. Knockdown of NSUN2 alleviated kidney damage, reduced blood glucose and urine albumin, and suppressed macrophage infiltration in DN mice. Moreover, NSUN2 interacted with SOCS1, and silenced NSUN2 inhibited m5C levels of SOCS1 to reduce SOCS1 mRNA stability. Additionally, interference with NSUN2 suppressed macrophage migration, invasion, and infiltration by positively regulating SOCS1 expression under HG conditions. CONCLUSION In conclusion, silencing of NSUN2 inhibits macrophage infiltration by reducing m5C modification of SOCS1, and thereby attenuates renal injury. The findings suggest a novel regulatory mechanism between NSUN2-mediated m5C modification and DN.
Collapse
Affiliation(s)
- Ru Wang
- Department of Laboratory Medcine, The 305 Hospital of PLA, No.13 Wenjin Street, Xicheng District, Beijing, 100017, China
| | - Jianchang Qu
- Department of Endocrinology, The 305 Hospital of PLA, No.13 Wenjin Street, Xicheng District, Beijing, 100017, China
| | - Meiqiong Chen
- Department of Pathology, The 305 Hospital of PLA, No.13 Wenjin Street, Xicheng District, Beijing, 100017, China
| | - Tenglong Han
- Department of Laboratory Medcine, The 305 Hospital of PLA, No.13 Wenjin Street, Xicheng District, Beijing, 100017, China
| | - Zhipeng Liu
- Medical Department, The 305 Hospital of PLA, No.13 Wenjin Street, Xicheng District, Beijing, 100017, China
| | - Huizhong Wang
- Department of Laboratory Medcine, The 305 Hospital of PLA, No.13 Wenjin Street, Xicheng District, Beijing, 100017, China.
| |
Collapse
|
15
|
Hou X, Dong Q, Hao J, Liu M, Ning J, Tao M, Wang Z, Guo F, Huang D, Shi X, Gao M, Li D, Zheng X. NSUN2-mediated m 5C modification drives alternative splicing reprogramming and promotes multidrug resistance in anaplastic thyroid cancer through the NSUN2/SRSF6/UAP1 signaling axis. Theranostics 2025; 15:2757-2777. [PMID: 40083919 PMCID: PMC11898302 DOI: 10.7150/thno.104713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/14/2025] [Indexed: 03/16/2025] Open
Abstract
Rationale: Anaplastic thyroid carcinoma (ATC) is an extraordinarily aggressive form of thyroid cancer, frequently presenting with locally advanced infiltration or distant metastases at the time of initial diagnosis, thus missing the optimal window for surgical intervention. Consequently, systemic chemotherapy and targeted therapies are vital for improving the prognosis of ATC. However, ATC exhibits significant resistance to conventional treatments, highlighting the need to elucidate the biological mechanisms underlying this drug resistance and identify novel therapeutic targets to overcome it. Methods: We conducted a comprehensive analysis of both bulk and single-cell RNA sequencing (scRNA-seq) data from ATC samples to screen for m5C modification-related genes associated with multidrug resistance (MDR). We then performed IC50 assays, flow cytometry, and employed a spontaneous tumorigenic ATC mouse model with Nsun2 knockout to demonstrate that NSUN2 promotes MDR in ATC. To investigate the mechanisms of NSUN2-mediated drug resistance, we generated NSUN2-knockout ATC cell lines and performed transcriptomic, proteomic, and MeRIP-seq analyses. Additionally, RNA sequencing and alternative splicing analyses were conducted to determine global changes upon NSUN2 knockout. We further explored the underlying mechanisms of the NSUN2/SRSF6/UAP1 axis through glycoprotein staining, denaturing IP ubiquitination, nuclear-cytoplasmic fractionation, and PCR. Lastly, we evaluated the synergistic effects of a small-molecule NSUN2 inhibitor with anticancer agents both in vitro and in vivo. Results: Our findings reveal that NSUN2 expression correlates significantly with MDR in ATC. NSUN2 operates as a "writer" and ALYREF as a "reader" of m5C on SRSF6 mRNA, inducing alternative splicing reprogramming and redirecting the splice form of the UAP1 gene from AGX1 to AGX2. As a result, AGX2 enhances the N-linked glycosylation of ABC transporters, stabilizing them by preventing ubiquitination-mediated degradation. Furthermore, an NSUN2 inhibitor reduces NSUN2 enzymatic activity and diminishes downstream target expression, presenting a novel, promising therapeutic approach to overcome MDR in ATC. Conclusions: These findings suggest that the NSUN2/SRSF6/UAP1 signaling axis plays a vital role in MDR of ATC and identify NSUN2 as a synergistic target for chemotherapy and targeted therapy in ATC.
Collapse
Affiliation(s)
- Xiukun Hou
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Qiman Dong
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Jie Hao
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
- Department of Thyroid and Breast Surgery, Tianjin Union Medical Center, Tianjin 300121, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300121, China
| | - Min Liu
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Junya Ning
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Mei Tao
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Zhongyu Wang
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Fengli Guo
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Dongmei Huang
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Xianle Shi
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Ming Gao
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
- Department of Thyroid and Breast Surgery, Tianjin Union Medical Center, Tianjin 300121, China
- Tianjin Key Laboratory of General Surgery in Construction, Tianjin Union Medical Center, Tianjin 300121, China
| | - Dapeng Li
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, China
| |
Collapse
|
16
|
Li J, Chang W, Li J, Zhao X, Li X. IL-22-mediated microRNA-124-3p/GRB2 axis regulates hyperproliferation and inflammatory response of keratinocytes in psoriasis. Arch Dermatol Res 2025; 317:227. [PMID: 39792268 DOI: 10.1007/s00403-024-03668-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/08/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
Psoriasis is an inflammatory dermatosis that features overproliferation and inflammatory reaction of keratinocytes. A study reported that IL-22 is involved in the pathogenesis of psoriasis by mediating miR-124 to regulate the expression of fibroblast growth factor receptor 2 in keratinocytes. A microRNA may target multiple target genes. Therefore, we speculate that miR-124-3p may also target other downstream genes to affect IL -22-induced keratinocyte function. A possible target gene of miR-124-3p, growth factor receptor-bound protein 2 (GRB2), was screened by analyzing the target gene databases. GRB2 expression was elevated and miR-124-3p expression was decreased in psoriatic lesions compared to psoriatic adjacent normal skins and healthy controls. We performed the following cell experiments in the IL-22-stimulated HaCaT cell model. In keratinocytes transfected with the miR-124-3p mimics, GRB2 expression was significantly lower. We analyzed the regulation of keratinocyte proliferation by GRB2 and miR-124-3p. High levels of GRB2 promoted keratinocyte proliferation and expression of Ki67, PCNA, and K16, which were inhibited by low expression of GRB2. In addition, we found that the effect of GRB2 inhibitors on the proliferation and inflammatory response of keratinocytes was dose-dependent. Finally, we investigated the influence of GRB2 on inflammatory mediators in keratinocytes with the ELISA. After low expression of GRB2, the mRNA expression and secretion of the pro-inflammatory factor were suppressed. When both GRB2 and miR-124-3p were overexpressed, the cellular overproliferation and inflammation caused by GRB2 overexpression were significantly reversed by miR-124-3p. In summary, IL-22-mediated miR-124-3p regulates keratinocyte hyperproliferation and inflammatory response by suppressing GRB2 expression in psoriasis.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Public Health, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenjuan Chang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Junqin Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiya Zhao
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
- Ninth Clinical College of Medicine, Shanxi Medical University, No.5, Dong San Dao Xiang, Jiefang Road, Taiyuan, China
| | - Xinhua Li
- School of Public Health, Shanxi Medical University, Taiyuan, China.
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China.
- Ninth Clinical College of Medicine, Shanxi Medical University, No.5, Dong San Dao Xiang, Jiefang Road, Taiyuan, China.
| |
Collapse
|
17
|
Wu H, Chen S, Li X, Li Y, Shi H, Qing Y, Shi B, Tang Y, Yan Z, Hao Y, Wang D, Liu W. RNA modifications in cancer. MedComm (Beijing) 2025; 6:e70042. [PMID: 39802639 PMCID: PMC11718328 DOI: 10.1002/mco2.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025] Open
Abstract
RNA modifications are emerging as critical cancer regulators that influence tumorigenesis and progression. Key modifications, such as N6-methyladenosine (m6A) and 5-methylcytosine (m5C), are implicated in various cellular processes. These modifications are regulated by proteins that write, erase, and read RNA and modulate RNA stability, splicing, translation, and degradation. Recent studies have highlighted their roles in metabolic reprogramming, signaling pathways, and cell cycle control, which are essential for tumor proliferation and survival. Despite these scientific advances, the precise mechanisms by which RNA modifications affect cancer remain inadequately understood. This review comprehensively examines the role RNA modifications play in cancer proliferation, metastasis, and programmed cell death, including apoptosis, autophagy, and ferroptosis. It explores their effects on epithelial-mesenchymal transition (EMT) and the immune microenvironment, particularly in cancer metastasis. Furthermore, RNA modifications' potential in cancer therapies, including conventional treatments, immunotherapy, and targeted therapies, is discussed. By addressing these aspects, this review aims to bridge current research gaps and underscore the therapeutic potential of targeting RNA modifications to improve cancer treatment strategies and patient outcomes.
Collapse
Affiliation(s)
- Han Wu
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Shi Chen
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Xiang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Yuyang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - He Shi
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Yiwen Qing
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| | - Bohe Shi
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Yifei Tang
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Zhuoyi Yan
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Yang Hao
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Dongxu Wang
- Laboratory Animal CenterCollege of Animal ScienceJilin University, ChangchunJilin provinceChina
| | - Weiwei Liu
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin University, ChangchunJilin provinceChina
- Jilin Provincial Key Laboratory of Tooth Development and Bone RemodelingHospital of StomatologyJilin University, ChangchunJilin provincleChina
| |
Collapse
|
18
|
Ma J, Dong Y, Liu J, Gao S, Quan J. The role of GRB2 in diabetes, diabetes complications and related disorders. Diabetes Obes Metab 2025; 27:23-34. [PMID: 39478285 DOI: 10.1111/dom.16015] [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: 06/10/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 12/06/2024]
Abstract
Growth factor receptor-bound protein 2 (GRB2) is a key adaptor protein involved in multiple signalling pathways, and its dysregulation is associated with various diseases. Type 2 diabetes is a systemic condition characterized by insulin resistance and impaired β-cell function. The complications of diabetes significantly reduce life expectancy and quality of life, imposing a substantial burden on society. However, the role of GRB2 in diabetes and associated complications is largely unknown. Emerging evidence suggests that GRB2 plays a crucial role in insulin resistance, inflammation, immune activation and the regulation of cellular processes such as cell proliferation, growth, metabolism, angiogenesis, apoptosis and differentiation. Dysregulation of GRB2-mediated pathways contributes to the progression of diabetic neuropathy, cognitive dysfunction, nephropathy, retinopathy and related disorders. This review provides a comprehensive overview of the current understanding of the role of GRB2 in diabetes, diabetes complications and related disorders, alongside recent advances in the development of GRB2-targeted therapies. Elucidating the complex role of GRB2 in these disorders provides valuable insights into potential therapeutic strategies targeting GRB2-mediated pathways.
Collapse
Affiliation(s)
- Jing Ma
- Department of Endocrinology and Metabolism, Gansu Provincial Hospital, The First Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Endocrinology and Metabolism, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Endocrine and Metabolic Diseases of Gansu Province, Lanzhou, China
| | - Yuyan Dong
- Clinical College of Ningxia Medical University, Yinchuan, China
| | - Juxiang Liu
- Department of Endocrinology and Metabolism, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Endocrine and Metabolic Diseases of Gansu Province, Lanzhou, China
| | - Shuo Gao
- Department of Endocrinology and Metabolism, Gansu Provincial Hospital, The First Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jinxing Quan
- Department of Endocrinology and Metabolism, Gansu Provincial Hospital, The First Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Endocrinology and Metabolism, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Endocrine and Metabolic Diseases of Gansu Province, Lanzhou, China
| |
Collapse
|
19
|
Uddin MB, Wang Z, Yang C. Epitranscriptomic RNA m 6A Modification in Cancer Therapy Resistance: Challenges and Unrealized Opportunities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 12:e2403936. [PMID: 39661414 PMCID: PMC11775542 DOI: 10.1002/advs.202403936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/24/2024] [Indexed: 12/12/2024]
Abstract
Significant advances in the development of new cancer therapies have given rise to multiple novel therapeutic options in chemotherapy, radiotherapy, immunotherapy, and targeted therapies. Although the development of resistance is often reported along with temporary disease remission, there is often tumor recurrence of an even more aggressive nature. Resistance to currently available anticancer drugs results in poor overall and disease-free survival rates for cancer patients. There are multiple mechanisms through which tumor cells develop resistance to therapeutic agents. To date, efforts to overcome resistance have only achieved limited success. Epitranscriptomics, especially related to m6A RNA modification dysregulation in cancer, is an emerging mechanism for cancer therapy resistance. Here, recent studies regarding the contributions of m6A modification and its regulatory proteins to the development of resistance to different cancer therapies are comprehensively reviewed. The promise and potential limitations of targeting these entities to overcome resistance to various anticancer therapies are also discussed.
Collapse
Affiliation(s)
- Mohammad Burhan Uddin
- Department of Pharmaceutical SciencesNorth South UniversityBashundharaDhaka1229Bangladesh
| | - Zhishan Wang
- Stony Brook Cancer CenterStony Brook UniversityStony BrookNY11794USA
| | - Chengfeng Yang
- Stony Brook Cancer CenterStony Brook UniversityStony BrookNY11794USA
- Department of PathologyRenaissance School of MedicineStony Brook UniversityStony BrookNY11794USA
| |
Collapse
|
20
|
Lu Y, Yang L, Feng Q, Liu Y, Sun X, Liu D, Qiao L, Liu Z. RNA 5-Methylcytosine Modification: Regulatory Molecules, Biological Functions, and Human Diseases. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae063. [PMID: 39340806 PMCID: PMC11634542 DOI: 10.1093/gpbjnl/qzae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/12/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024]
Abstract
RNA methylation modifications influence gene expression, and disruptions of these processes are often associated with various human diseases. The common RNA methylation modification 5-methylcytosine (m5C), which is dynamically regulated by writers, erasers, and readers, widely occurs in transfer RNAs (tRNAs), messenger RNAs (mRNAs), ribosomal RNAs (rRNAs), enhancer RNAs (eRNAs), and other non-coding RNAs (ncRNAs). RNA m5C modification regulates metabolism, stability, nuclear export, and translation of RNA molecules. An increasing number of studies have revealed the critical roles of the m5C RNA modification and its regulators in the development, diagnosis, prognosis, and treatment of various human diseases. In this review, we summarized the recent studies on RNA m5C modification and discussed the advances in its detection methodologies, distribution, and regulators. Furthermore, we addressed the significance of RNAs modified with m5C marks in essential biological processes as well as in the development of various human disorders, from neurological diseases to cancers. This review provides a new perspective on the diagnosis, treatment, and monitoring of human diseases by elucidating the complex regulatory network of the epigenetic m5C modification.
Collapse
Affiliation(s)
- Yanfang Lu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, China
| | - Liu Yang
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, China
| | - Qi Feng
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, China
| | - Yong Liu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, China
| | - Xiaohui Sun
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, China
| | - Dongwei Liu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, China
| | - Long Qiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhangsuo Liu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, Zhengzhou 450052, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, China
| |
Collapse
|
21
|
Li Q, Guo G, Chen Y, Lu L, Li H, Zhou Z, Guo J, Gan X, Hu Y, Li Q, Sun M, Liu X. HCP5 Derived Novel Microprotein Triggers Progression of Gastric Cancer through Regulating Ferroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407012. [PMID: 39447131 PMCID: PMC11633528 DOI: 10.1002/advs.202407012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/31/2024] [Indexed: 10/26/2024]
Abstract
The context of long noncoding RNAs (lncRNAs) contains many unannotated open reading frames (ORFs). These ORFs potentially encode novel proteins or peptides with crucial roles in various human cancers, yet the translational potential of these lncRNAs and the functions of the protein products remain largely unexplored, especially in gastric cancer (GC). In this study, a comprehensive analysis is performed and identified a GC associated lncRNA known as HCP5, which contains a non-canonical ORF. Further analysis showed that HCP5-132aa, a microprotein encoded by HCP5 harboring this ORF, is highly expressed in GC cells and tissues, and can promote the proliferation of GC cells by inhibiting ferroptosis. Mechanistically, HCP5-132aa enhances the interaction between YBX1 and ELAVL1, facilitates recognition of YBX1 at the m5C site in the 3'UTR of SLC7A11 and G6PD mRNA, and preserves their stability via ELAVL1. By employing a Cas9/sgRNA delivery system with AAV in vivo, effectively knocked out the HCP5-132aa and inhibition of tumor growth in a patient-derived xenograft model are achieved. These findings demonstrate that the novel protein HCP5-132aa, derived from lncRNA HCP5, mediates the repression of ferroptosis, thereby driving the progression of GC and identifying a new potential therapeutic target for its treatment.
Collapse
Affiliation(s)
- Qiuhui Li
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Guoqing Guo
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Yuli Chen
- Suzhou Cancer Center Core LaboratoryThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhou215001China
| | - Lu Lu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Hanyang Li
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| | - Zihan Zhou
- The First Clinical Medical CollegeNanjing Medical UniversityNanjing211166China
| | - Jiahao Guo
- Suzhou Cancer Center Core LaboratoryThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhou215001China
| | - Xiongkang Gan
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Yanming Hu
- Suzhou Cancer Center Core LaboratoryThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhou215001China
| | - Qiunuo Li
- The First Clinical Medical CollegeNanjing Medical UniversityNanjing211166China
| | - Ming Sun
- Suzhou Cancer Center Core LaboratoryThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhou215001China
| | - Xianghua Liu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166China
| |
Collapse
|
22
|
Cotino‐Nájera S, García‐Villa E, Cruz‐Rosales S, Gariglio P, Díaz‐Chávez J. The role of Lin28A and Lin28B in cancer beyond Let-7. FEBS Lett 2024; 598:2963-2979. [PMID: 39152528 PMCID: PMC11665955 DOI: 10.1002/1873-3468.15004] [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: 05/11/2024] [Revised: 07/02/2024] [Accepted: 07/21/2024] [Indexed: 08/19/2024]
Abstract
Lin28A and Lin28B are paralogous RNA-binding proteins that play fundamental roles in development and cancer by regulating the microRNA family of tumor suppressor Let-7. Although Lin28A and Lin28B share some functional similarities with Let-7 inhibitors, they also have distinct expression patterns and biological functions. Increasing evidence indicates that Lin28A and Lin28B differentially impact cancer stem cell properties, epithelial-mesenchymal transition, metabolic reprogramming, and other hallmarks of cancer. Therefore, it is important to understand the overexpression of Lin28A and Lin28B paralogs in specific cancer contexts. In this review, we summarize the main similarities and differences between Lin28A and Lin28B, their implications in different cellular processes, and their role in different types of cancer. In addition, we provide evidence of other specific targets of each lin28 paralog, as well as the lncRNAs and miRNAs that promote or inhibit its expression, and how this impacts cancer development and progression.
Collapse
Affiliation(s)
- Sandra Cotino‐Nájera
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios Avanzados (CINVESTAV)Mexico CityMexico
| | - Enrique García‐Villa
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios Avanzados (CINVESTAV)Mexico CityMexico
| | - Samantha Cruz‐Rosales
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios Avanzados (CINVESTAV)Mexico CityMexico
| | - Patricio Gariglio
- Departamento de Genética y Biología MolecularCentro de Investigación y de Estudios Avanzados (CINVESTAV)Mexico CityMexico
| | - José Díaz‐Chávez
- Departamento de Biología Celular, Facultad de CienciasUNAMMexico CityMexico
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones BiomédicasUNAM/Instituto Nacional de CancerologíaMexico CityMexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la SaludMexico
| |
Collapse
|
23
|
Zhang X, Zhang Y, Li R, Li Y, Wang Q, Wang Y, Chen X, Wang W, Pang E, Li Y, Wang J, Zheng J, Zhang J. STUB1-mediated ubiquitination and degradation of NSUN2 promotes hepatocyte ferroptosis by decreasing m 5C methylation of Gpx4 mRNA. Cell Rep 2024; 43:114885. [PMID: 39453812 DOI: 10.1016/j.celrep.2024.114885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 08/01/2024] [Accepted: 10/02/2024] [Indexed: 10/27/2024] Open
Abstract
Ferroptosis is an iron-dependent cell death that occurs due to the peroxidation of phospholipids in the cell membrane. In this study, we find that the protein level of NSUN2 is significantly decreased in hepatocyte ferroptosis. This is attributed to STUB1-mediated ubiquitination of NSUN2 at lysines 457 and 654, promoting NSUN2 degradation in ferroptosis. Selenoprotein glutathione peroxidase 4 (GPX4) is a prominent suppressor of ferroptosis. We find that downregulation of NSUN2 diminishes m5C methylation of Gpx4 mRNA 3' UTR. The reduction of NSUN2-mediated Gpx4 mRNA m5C methylation abrogates the interaction between SBP2 and the selenocysteine insertion sequence (SECIS) and leads to inhibition of GPX4 protein expression. Lower GPX4 expression promotes hepatocyte ferroptosis in vivo and in vitro, which is reversed by restoration of NSUN2. These findings shed light on the mechanism of NSUN2 degradation and also indicate that the STUB1-NSUN2-GPX4 axis plays a regulatory role in hepatocyte ferroptosis.
Collapse
Affiliation(s)
- Xiaotian Zhang
- The Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Yihua Zhang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Rongrong Li
- The Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yibo Li
- The Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Qi Wang
- MOE Key Laboratory for Biodiversity Science and Ecological Engineering and Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xinying Chen
- The Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Weihua Wang
- The Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Erli Pang
- MOE Key Laboratory for Biodiversity Science and Ecological Engineering and Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yanyan Li
- Center for Healthy Aging, Changzhi Medical College, Changzhi 046000, China
| | - Jia Wang
- Department of Immunology, Changzhi Medical College, Changzhi 046000, China
| | - Jinping Zheng
- Center for Healthy Aging, Changzhi Medical College, Changzhi 046000, China
| | - Junjie Zhang
- The Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
24
|
Zhang C, Cui Y. Targeting TSPEAR-AS2 suppresses tumor growth and interferon signaling in esophageal cancer. Sci Rep 2024; 14:28768. [PMID: 39567604 PMCID: PMC11579022 DOI: 10.1038/s41598-024-80439-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/19/2024] [Indexed: 11/22/2024] Open
Abstract
Dysregulation of long non-coding RNAs (lncRNAs) and interferon signaling contributes to tumorigenesis and progression; however, their crosstalk in tumor biology remains poorly understood. Here, we showed that TSPEAR-AS2, an lncRNA mediated by METTL1, activates the interferon signaling pathway in esophageal cancer (ESCA). Clinically, we conducted a pan-cancer investigation and identified TSPEAR-AS2 as a novel ESCA-related lncRNA that predicts a worse patient prognosis. The high expression of TSPEAR-AS2 was validated in multiple ESCA cohorts, demonstrating that this lncRNA had good diagnostic performance. METTL1, an RNA N7-methylguanosine (m7G) methyltransferase, was positively correlated with TSPEAR-AS2 expression in ESCA tissues, and METTL1 was found to induce TSPEAR-AS2 expression in ESCA cells. Functionally, the up-regulation of TSPEAR-AS2 promoted proliferation, cell cycle progression, migration, and stemness in ESCA cells, whereas its knockdown attenuated these malignant phenotypes. Furthermore, both TSPEAR-AS2 deficiency and antisense oligonucleotide (ASO)-based intratumoral intervention could significantly suppress tumor growth in vivo. Mechanistically, quantitative proteomic analysis revealed that TSPEAR-AS2 ablation altered the expression of functional proteins related to interferon signaling pathways, such as HLA-E, IRF3, and IFITs. These findings identify a previously unrecognized role of the METTL1/TSPEAR-AS2/interferon signaling axis in ESCA development and suggest that TSPEAR-AS2 is a potential prognostic indicator and therapeutic vulnerability for this malignancy.
Collapse
Affiliation(s)
- Chunyan Zhang
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Yuanbo Cui
- Department of Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China.
- Department of Trauma and Metabolism, Research Institute of Zhengzhou University, Central Hospital Affiliated to Zhengzhou University, ZhengzhouZhengzhou, 450007, China.
| |
Collapse
|
25
|
Wang R, Ding L, Lin Y, Luo W, Xu Z, Li W, Lu Y, Zhu Z, Lu Z, Li F, Mao X, Xia L, Li G. The Quiet Giant: Identification, Effectors, Molecular Mechanism, Physiological and Pathological Function in mRNA 5-methylcytosine Modification. Int J Biol Sci 2024; 20:6241-6254. [PMID: 39664561 PMCID: PMC11628344 DOI: 10.7150/ijbs.101337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/27/2024] [Indexed: 12/13/2024] Open
Abstract
5-Methylcytosine (m5C) is a prevalent nucleotide alteration observed in transfer RNA (tRNA) and ribosomal RNA (rRNA), and it is also widely distributed in the transcriptome, serving as one of the internal modifications of messenger RNA (mRNA) in higher eukaryotes. Increasing evidence has substantiated the presence of m5C in mRNA. As research on m5C progresses, there is an initial comprehension of its molecular mechanisms and biological significance in mRNA. This work aims to provide a comprehensive summary of the most recent advancements in the identification and screening, distribution, molecular functions, and biological effects of m5C in mRNA. We outline the current status of research and provide prospects for potential future applications.
Collapse
Affiliation(s)
- Ruyue Wang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Lifeng Ding
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Yudong Lin
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Wenqin Luo
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Zhehao Xu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Weilin Li
- Department of Urology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, China
| | - Yi Lu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Ziwei Zhu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Zeyi Lu
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Fan Li
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Xudong Mao
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Liqun Xia
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| |
Collapse
|
26
|
Li D, Liu Y, Yang G, He M, Lu L. Recent insights into RNA m5C methylation modification in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189223. [PMID: 39577751 DOI: 10.1016/j.bbcan.2024.189223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
RNA 5-methylcytosine (m5C) methylation involves the addition of a methyl (-CH3) group to the cytosine (C) base within an RNA molecule, forming the m5C modification. m5C plays a role in numerous essential biological processes, including the regulation of RNA stability, nuclear export, and protein translation. Recent studies have highlighted the importance of m5C in the pathogenesis of various diseases, particularly tumors. Emerging evidence indicates that RNA m5C methylation is intricately implicated in the mechanisms underlying hepatocellular carcinoma (HCC). Dysregulation of m5C-associated regulatory factors is common in HCC and shows significant associations with prognosis, treatment response, and clinicopathological features. This review provides an in-depth analysis of the components and functions of m5C regulators, particularly emphasizing their research advancements in the context of HCC.
Collapse
Affiliation(s)
- Danyang Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong Province 519000, PR China
| | - Yanyan Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong Province 519000, PR China
| | - Guang Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong Province 519000, PR China
| | - Mingyu He
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong Province 519000, PR China.
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai Clinical Medical College of Jinan University (Zhuhai People's Hospital), Zhuhai, Guangdong Province 519000, PR China; Guangzhou First Pepople's Hospital, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong Province 510006, PR China.
| |
Collapse
|
27
|
Li P, Huang D. NSUN2-mediated RNA methylation: Molecular mechanisms and clinical relevance in cancer. Cell Signal 2024; 123:111375. [PMID: 39218271 DOI: 10.1016/j.cellsig.2024.111375] [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: 07/06/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Cancer remains a leading cause of morbidity and mortality worldwide, necessitating the ongoing investigation of molecular targets for improved diagnosis, prognosis, and therapy. Among these targets, RNA modifications, particularly N5-methylcytosine (m5C) in RNA, have emerged as critical regulators of gene expression and cellular functions. NOP2/Sun RNA methyltransferase family member 2 (NSUN2) is a key enzyme in m5C modification, significantly influencing various biological processes and tumorigenesis. NSUN2 methylates multiple RNA species, including transfer RNAs (tRNAs), messenger RNAs (mRNAs), and non-coding RNAs, impacting RNA stability, translation efficiency, and cellular stress responses. These modifications, in turn, affect cell proliferation, differentiation, and survival. In cancer, NSUN2 is frequently upregulated, associated with aggressive tumor phenotypes, poor prognosis, and therapy resistance. Its role in oncogenic signaling pathways further underscores its importance in cancer biology. This review offers a comprehensive overview of NSUN2's role in cancer, focusing on its involvement in RNA methylation and its implications for tumor initiation and progression. Additionally, we explore the potential of NSUN2 as a biomarker for cancer diagnosis and prognosis, and its promise as a therapeutic target.
Collapse
Affiliation(s)
- Penghui Li
- Department of gastrointestinal surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471000, Henan, China.
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| |
Collapse
|
28
|
Zhong L, Wu J, Zhou B, Kang J, Wang X, Ye F, Lin X. ALYREF recruits ELAVL1 to promote colorectal tumorigenesis via facilitating RNA m5C recognition and nuclear export. NPJ Precis Oncol 2024; 8:243. [PMID: 39455812 PMCID: PMC11512073 DOI: 10.1038/s41698-024-00737-0] [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: 04/30/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
ALYREF can recognize 5-methylcytosine (m5C) decoration throughout RNAs to regulate RNA metabolism. However, its implications in cancer and precise regulatory mechanisms remain largely elusive. Here, we demonstrated that ALYREF supported colorectal cancer (CRC) growth and migration. Integrated analysis of ALYREF-RIP-Bis-seq and transcriptome profiles identified ribosomal protein S6 kinase B2 (RPS6KB2) and regulatory-associated protein of mTOR (RPTOR) as ALYREF's possible downstream effectors. Mechanistically, ALYREF formed a complex with ELAV like RNA binding protein 1 (ELAVL1) to cooperatively promote m5C recognition and nuclear export of the two mRNAs. Moreover, ALYREF protein was highly expressed in tumor tissues of CRC patients, which predicted their poor prognosis. E2F transcription factor 6 (E2F6)-mediated transactivation gave a molecular insight into ALYREF overexpression. Collectively, ALYREF recruits ELAVL1 to collaboratively facilitate m5C recognition and nuclear export of RPS6KB2 and RPTOR transcripts for colorectal tumorigenesis, providing RNA m5C methylation as promising therapeutic targets and prognostic biomarkers for CRC.
Collapse
Affiliation(s)
- Longhua Zhong
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China
| | - Jingxun Wu
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China
| | - Bingqian Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China
| | - Jiapeng Kang
- Department of Medical Oncology, Zhangzhou Municipal Hospital, Zhangzhou Municipal Hospital Affiliated of Fujian Medical University, Zhangzhou, P. R. China
| | - Xicheng Wang
- Cancer Medical Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China.
| | - Feng Ye
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China.
- Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, China.
| | - Xiaoting Lin
- Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, P. R. China.
- Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, China.
| |
Collapse
|
29
|
Yu L, Xu H, Xiong H, Yang C, Wu Y, Zhang Q. The role of m5C RNA modification in cancer development and therapy. Heliyon 2024; 10:e38660. [PMID: 39444404 PMCID: PMC11497397 DOI: 10.1016/j.heliyon.2024.e38660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/13/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
RNA modifications have been demonstrated to affect the function, stability, processing, and interactions of RNA, including pseudouridylation, acetylation and methylation. RNA methylation products, such as N6-methyladenosine (m6A), 5-methylcytidine (m5C), N7-methylguanosine (m7G), 2'-O-dimethyladenosine (m6Am), and N1-methyladenosine (m1A), have been reported to participate in tumorigenesis and tumor progression. The role of m6A in carcinogenesis has been well studied and summarized. In this review, we described the biological functions of m5C RNA modifications in tumorigenesis and tumor progression. Moreover, we highlighted the molecular mechanisms of m5C RNA modification in oncogenesis. Furthermore, we discussed whether targeting m5C regulator-associated genes could be a novel strategy for improving therapeutic outcomes in patients with cancer.
Collapse
Affiliation(s)
- Li Yu
- Cancer Center, Department of Nursing, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hongen Xu
- Department of Oncology, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou, China
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hanchu Xiong
- Cancer Center, Department of Radiation Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chunju Yang
- Cancer Center, Department of Nursing, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ying Wu
- Cancer Center, Department of Nursing, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiong Zhang
- Cancer Center, Department of Nursing, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
30
|
Yang W, Zhao Y, Yang Y. Dynamic RNA methylation modifications and their regulatory role in mammalian development and diseases. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2084-2104. [PMID: 38833084 DOI: 10.1007/s11427-023-2526-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/15/2023] [Indexed: 06/06/2024]
Abstract
Among over 170 different types of chemical modifications on RNA nucleobases identified so far, RNA methylation is the major type of epitranscriptomic modifications existing on almost all types of RNAs, and has been demonstrated to participate in the entire process of RNA metabolism, including transcription, pre-mRNA alternative splicing and maturation, mRNA nucleus export, mRNA degradation and stabilization, mRNA translation. Attributing to the development of high-throughput detection technologies and the identification of both dynamic regulators and recognition proteins, mechanisms of RNA methylation modification in regulating the normal development of the organism as well as various disease occurrence and developmental abnormalities upon RNA methylation dysregulation have become increasingly clear. Here, we particularly focus on three types of RNA methylations: N6-methylcytosine (m6A), 5-methylcytosine (m5C), and N7-methyladenosine (m7G). We summarize the elements related to their dynamic installment and removal, specific binding proteins, and the development of high-throughput detection technologies. Then, for a comprehensive understanding of their biological significance, we also overview the latest knowledge on the underlying mechanisms and key roles of these three mRNA methylation modifications in gametogenesis, embryonic development, immune system development, as well as disease and tumor progression.
Collapse
Affiliation(s)
- Wenlan Yang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
| | - Yongliang Zhao
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
| | - Yungui Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- China National Center for Bioinformation, Beijing, 100101, China.
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 101408, China.
| |
Collapse
|
31
|
Wang L, Zhang Q, Wang J, Lu H, Zeng W, Zhang T. Vitamin D3 regulates NSUN2 expression and inhibits melanoma cell proliferation and migration. Mol Divers 2024; 28:2863-2874. [PMID: 37688740 DOI: 10.1007/s11030-023-10720-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023]
Abstract
The activated form of vitamin D3 [1,25-dihydroxyvitamin D3; 1,25(OH)2D3] is important for various physiological processes, such as bone mineralization and calcium metabolism, and plays an anticancer role in numerous cancers as well. Its role in melanoma cells has yet to be proven. NOP2/Sun RNA methyltransferase 2 (NSUN2) is a typical RNA methyltransferase and is highly expressed in a variety of cancer cells. However, the molecular mechanisms underlying the role of 1,25(OH)2D3 and NSUN2 in melanoma cells remain largely unknown. The current study showed that 1,25(OH)2D3 could significantly and specifically inhibit the proliferation and migration of melanoma B16 cells. 1,25(OH)2D3 enhances vitamin D receptor expression while simultaneously reducing NSUN2 expression in melanoma cells. Subsequently, knockdown of NSUN2 suppressed B16 cell proliferation and migration. RNA-Seq results illuminated that DNA replication, cell proliferation and cell cycle pathways were enriched, and genes promoting these pathways were reduced after knocking down Nsun2. Dual-luciferase reporter assays showed that 1,25(OH)2D3 downregulated reporter gene expression was controlled by the Nsun2 promoter. The results suggest that 1,25(OH)2D3 binds to the vitamin D response element located upstream of the Nsun2 promoter to downregulate Nsun2 transcription activity and then affects the gene expression pattern related to cell proliferation and the cell cycle, thereby restraining B16 cell proliferation and migration.
Collapse
Affiliation(s)
- Ling Wang
- School of Biological Science and Engineering, Shaanxi University of Technology, Dongguan Street South Campus, Hanzhong, 723001, Shaanxi Province, China
| | - Qiang Zhang
- School of Biological Science and Engineering, Shaanxi University of Technology, Dongguan Street South Campus, Hanzhong, 723001, Shaanxi Province, China
- College of Life Sciences, Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Jinping Wang
- School of Biological Science and Engineering, Shaanxi University of Technology, Dongguan Street South Campus, Hanzhong, 723001, Shaanxi Province, China
| | - Hongzhao Lu
- School of Biological Science and Engineering, Shaanxi University of Technology, Dongguan Street South Campus, Hanzhong, 723001, Shaanxi Province, China
| | - Wenxian Zeng
- School of Biological Science and Engineering, Shaanxi University of Technology, Dongguan Street South Campus, Hanzhong, 723001, Shaanxi Province, China
- QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C, Shaanxi University of Technology, Hanzhong, 723001, China
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, Shaanxi University of Technology, Hanzhong, 723001, China
| | - Tao Zhang
- School of Biological Science and Engineering, Shaanxi University of Technology, Dongguan Street South Campus, Hanzhong, 723001, Shaanxi Province, China.
- QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C, Shaanxi University of Technology, Hanzhong, 723001, China.
- Shaanxi Province Key Laboratory of Bioresources, Shaanxi University of Technology, Hanzhong, 723001, China.
| |
Collapse
|
32
|
Cai P, Li J, An M, Li M, Guo J, Li J, Li X, Chen S, Zhang A, Li P, Liu Y, Zhang W, Fu B. Comprehensive analysis of RNA-5-methylcytosine modification in breast cancer brain metastasis. Future Oncol 2024; 20:2993-3008. [PMID: 39345093 PMCID: PMC11572191 DOI: 10.1080/14796694.2024.2405459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Aim: To delineate the RNA-5-methylcytosine (m5C) modification of breast cancer brain metastasis (BCBM).Methods: Methylated RNA immunoprecipitation next-generation sequencing (MeRIP-seq) was performed to obtain RNA-m5C patterns of BCBM.Results: 1048 hypermethylation and 1866 hypomethylation m5C peaks were identified in BCBM compared with those in breast cancer. The most significant m5C hypermethylated genes included ENG, SHANK1, IGFN1, EVL and MMP9, whereas the most significant m5C hypomethylated genes included AREG, SAA2, TP53I11, KRT7 and LCN2. MeRIP-qPCR data were concordant with the corresponding MeRIP-seq results in terms of the observed m5C levels. Conjoint analysis identified 190 hyper-up genes characterized by concurrent m5C hypermethylation and up-regulation, alongside 284 hypo-down genes exhibiting both m5C hypomethylation and down-regulation.Conclusion: This study presents the first comprehensive analysis of RNA-m5C modification in BCBM.
Collapse
Affiliation(s)
- Peiying Cai
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Jichao Li
- Department of Clinical Laboratory, Liaocheng Women & Children Hospital, Liaocheng, P.R. China
| | - Meng An
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Min Li
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital; Shandong Provincial Key Medical & Health Laboratory of Precision Medicine for Aging Intervention & Active Health, Liaocheng, P.R. China
| | - Jianran Guo
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital; Shandong Provincial Key Medical & Health Laboratory of Precision Medicine for Aging Intervention & Active Health, Liaocheng, P.R. China
| | - Jun Li
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital; Shandong Provincial Key Medical & Health Laboratory of Precision Medicine for Aging Intervention & Active Health, Liaocheng, P.R. China
| | - Xuan Li
- Department of Molecular Pharmacology Key Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Shen Chen
- Department of Breast & Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Anqi Zhang
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Peng Li
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Yan Liu
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Wei Zhang
- Department of Breast & Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, P.R. China
| | - Bo Fu
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital; Shandong Provincial Key Medical & Health Laboratory of Precision Medicine for Aging Intervention & Active Health, Liaocheng, P.R. China
| |
Collapse
|
33
|
Cheng L, Wang C, Zhao D, Wu S. Narrative review of research progress of RNA m 5C methylation in head and neck malignancies. Transl Cancer Res 2024; 13:5112-5122. [PMID: 39430837 PMCID: PMC11483327 DOI: 10.21037/tcr-24-103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 08/11/2024] [Indexed: 10/22/2024]
Abstract
Background and Objective Head and neck malignancies encompass a spectrum of malignant tumors occurring in the head and neck region, characterized by rapid progression, high recurrence rates, and dismal prognoses. Despite significant advancements in comprehensive surgery-based therapies, the 5-year survival rate for patients has not shown substantial improvement. There is an urgent need to investigate novel targeted therapies. With the advancements in epigenetics, RNA 5-methylcytosine (m5C) methylation, a prevalent form of RNA modification, has been identified by numerous studies as playing a pivotal role in the pathological processes of tumorigenesis and development. However, a comprehensive review within the realm of head and neck malignancies is currently lacking. This study aims to comprehensively review the biological implications of RNA m5C methylation regulators in the pathogenesis and progression of various systemic malignant tumors, with a specific focus on exploring the potential impact of RNA m5C methylation on head and neck malignancies. Methods A literature search on RNA m5C methylation and head and neck malignancies was conducted using PubMed, resulting in the inclusion of 46 relevant articles. The Cancer Genome Atlas (TCGA) database was utilized to analyze the correlation between m5C regulatory factors and clinicopathological features in patients with head and neck squamous cell carcinoma (HNSCC). Key Content and Findings Aberrant expression of RNA m5C methylation regulators is observed in head and neck malignancies, displaying a correlation with the clinicopathological grading of tumors. Conclusions RNA m5C methylation may contribute to the progression of head and neck malignancies and could be associated with an unfavorable prognosis for patients. These findings offer valuable insights for the development of targeted treatments for head and neck malignancies.
Collapse
Affiliation(s)
- Lang Cheng
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Chengtao Wang
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Dan Zhao
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
| | - Shuangjiang Wu
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
| |
Collapse
|
34
|
Tian J, Gao J, Cheng C, Xu Z, Chen X, Wu Y, Fu G, Jin B. NOP2-mediated 5-methylcytosine modification of APOL1 messenger RNA activates PI3K-Akt and facilitates clear cell renal cell carcinoma progression. Int J Biol Sci 2024; 20:4853-4871. [PMID: 39309431 PMCID: PMC11414376 DOI: 10.7150/ijbs.97503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Background: By regulating the functions of multiple RNAs, 5-methylcytosine (m5C) RNA methylation, particularly mediated by NOP2, is involved in tumorigenesis and developments. However, the specific functions and potential mechanisms of m5C, especially involving NOP2, in clear-cell renal cell carcinoma (ccRCC), remain unclear. Methods: NOP2 expression in cell lines and patient tissues was detected using western blotting, quantitative real-time polymerase chain reaction (RT-qPCR), and immunohistochemistry. The biological effects of NOP2 on ccRCC cells were investigated through a series of in vitro and in vivo experiments. To explore the potential regulatory mechanisms by which NOP2 affects ccRCC progression, m5C bisulfite sequencing, RNA-sequencing, RNA immunoprecipitation and methylated RNA immunoprecipitation (RIP/MeRIP) RT-qPCR assay, luciferase reporter assay, RNA stability assay, and bioinformatic analysis were performed. Results: NOP2 expression was significantly upregulated in ccRCC tissues and was associated with poor prognosis. Moreover, loss-of-function and gain-of-function assays demonstrated that NOP2 altered ccRCC cell proliferation, migration, and invasion. Mechanistically, NOP2 stimulated m5C modification of apolipoprotein L1 (APOL1) mRNA, and m5C reader YBX1 stabilized APOL1 mRNA through recognizing and binding to m5C site in the 3'-untranslated regions. Silencing APOL1 expression inhibited ccRCC cell proliferation in vitro and tumor formation in vivo. Furthermore, NOP2/APOL1 affected ccRCC progression via the PI3K-Akt signaling pathway. Conclusion: NOP2 functions as an oncogene in ccRCC by promoting tumor progression through the m5C-dependent stabilization of APOL1, which in turn regulates the PI3K-Akt signaling pathway, suggesting a potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Junjie Tian
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jianguo Gao
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Cheng Cheng
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Zhijie Xu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Xiaoyi Chen
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Yunfei Wu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Guanghou Fu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| | - Baiye Jin
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou 310003, China
| |
Collapse
|
35
|
Tian W, Zhu L, Luo Y, Tang Y, Tan Q, Zou Y, Chen K, Deng X, Tang H, Li H, Cai M, Xie X, Ye F. Autophagy Deficiency Induced by SAT1 Potentiates Tumor Progression in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309903. [PMID: 39073262 PMCID: PMC11423137 DOI: 10.1002/advs.202309903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 06/18/2024] [Indexed: 07/30/2024]
Abstract
Aggressive triple-negative breast cancer (TNBC) still lacks approved targeted therapies, requiring more exploration of its underlying mechanisms. Previous studies have suggested a potential role of SAT1 (Spermidine/Spermine N1-acetyltransferase 1) in cancer, which needs to be further elucidated in breast cancer. In this study, highly expressed SAT1 in TNBC signified worse patient prognoses. And SAT1 knockdown effectively inhibited the proliferation and migration abilities of TNBC cells in vitro and in vivo. In terms of mechanism, the transcription factor JUN enhanced SAT1 transcriptional activity by binding to its promoter region. Then, SAT1 protein in the cytoplasm engaged in directly binding with YBX1 for sustaining YBX1 protein stability via deubiquitylation mediated by the E3 ligase HERC5. Further, SAT1 was found to suppress autophagy remarkably via stabilization of mTOR mRNA with the accumulation of YBX1-mediated methyl-5-cytosine (m5C) modification. These findings proved that SAT1 drives TNBC progression through the SAT1/YBX1/mTOR axis, which may provide a potential candidate for targeted therapy in advanced TNBC.
Collapse
Affiliation(s)
- Wenwen Tian
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, P. R. China
| | - Lewei Zhu
- The First People's Hospital of Foshan, Foshan, 528000, P. R. China
| | - Yongzhou Luo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yuhui Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Qingjian Tan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yutian Zou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kun Chen
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P. R. China
| | - Xinpei Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Hongsheng Li
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, P. R. China
| | - Manbo Cai
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P. R. China
| | - Xiaoming Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Feng Ye
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| |
Collapse
|
36
|
Zhao Z, Zhou Y, Lv P, Zhou T, Liu H, Xie Y, Wu Z, Wang X, Zhao H, Zheng J, Jiang X. NSUN4 mediated RNA 5-methylcytosine promotes the malignant progression of glioma through improving the CDC42 mRNA stabilization. Cancer Lett 2024; 597:217059. [PMID: 38876383 DOI: 10.1016/j.canlet.2024.217059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/16/2024]
Abstract
5-Methylcytosine (m5C) methylation is a significant post-transcriptional modification that play a crucial role in the development and progression of numerous cancers. Whereas the functions and molecular mechanisms underlying m5C methylation in gliomas remain unclear. This study dedicated to explore changes of m5C levels and the clinical significance of the m5C writer NSUN4 in gliomas. We found that high m5C levels were negatively related to prognosis of patients with glioma. Moreover, gain- and loss-of-function experiments revealed the role of NSUN4 in enhancing m5C modification of mRNA to promote the malignant progression of glioma. Mechanistically speaking, NSUN4-mediated m5C alterations regulated ALYREF binding to CDC42 mRNA, thereby impacting the mRNA stability of CDC42. We also demonstrated that CDC42 promoted glioma proliferation, migration, and invasion by activating the PI3K-AKT pathway. Additionally, rescue experiments proved that CDC42 overexpression weaken the inhibitory effect of NSUN4 knockdown on the malignant progression of gliomas in vitro and in vivo. Our findings elucidated that NSUN4-mediated high m5C levels promote ALYREF binding to CDC42 mRNA and regulate its stability, thereby driving the malignant progression of glioma. This provides theoretical support for targeted the treatment of gliomas.
Collapse
Affiliation(s)
- Zhen Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yujie Zhou
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Lv
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ting Zhou
- Department of Gynaecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hanyuan Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Youxi Xie
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhipeng Wu
- Department of Neurosurgery, Weifang People's Hospital, Weifang, China
| | - Xuan Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongyang Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Jianglin Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
37
|
Chen XH, Guo KX, Li J, Xu SH, Zhu H, Yan GR. Regulations of m 6A and other RNA modifications and their roles in cancer. Front Med 2024; 18:622-648. [PMID: 38907157 DOI: 10.1007/s11684-024-1064-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/17/2024] [Indexed: 06/23/2024]
Abstract
RNA modification is an essential component of the epitranscriptome, regulating RNA metabolism and cellular functions. Several types of RNA modifications have been identified to date; they include N6-methyladenosine (m6A), N1-methyladenosine (m1A), 5-methylcytosine (m5C), N7-methylguanosine (m7G), N6,2'-O-dimethyladenosine (m6Am), N4-acetylcytidine (ac4C), etc. RNA modifications, mediated by regulators including writers, erasers, and readers, are associated with carcinogenesis, tumor microenvironment, metabolic reprogramming, immunosuppression, immunotherapy, chemotherapy, etc. A novel perspective indicates that regulatory subunits and post-translational modifications (PTMs) are involved in the regulation of writer, eraser, and reader functions in mediating RNA modifications, tumorigenesis, and anticancer therapy. In this review, we summarize the advances made in the knowledge of different RNA modifications (especially m6A) and focus on RNA modification regulators with functions modulated by a series of factors in cancer, including regulatory subunits (proteins, noncoding RNA or peptides encoded by long noncoding RNA) and PTMs (acetylation, SUMOylation, lactylation, phosphorylation, etc.). We also delineate the relationship between RNA modification regulator functions and carcinogenesis or cancer progression. Additionally, inhibitors that target RNA modification regulators for anticancer therapy and their synergistic effect combined with immunotherapy or chemotherapy are discussed.
Collapse
Affiliation(s)
- Xin-Hui Chen
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, State Key Laboratory of Respiratory Disease, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Kun-Xiong Guo
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, State Key Laboratory of Respiratory Disease, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jing Li
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, State Key Laboratory of Respiratory Disease, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Shu-Hui Xu
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, State Key Laboratory of Respiratory Disease, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Huifang Zhu
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, State Key Laboratory of Respiratory Disease, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Guang-Rong Yan
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, State Key Laboratory of Respiratory Disease, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
38
|
Zheng S, Hu C, Lin Q, Li T, Li G, Tian Q, Zhang X, Huang T, Ye Y, He R, Chen C, Zhou Y, Chen R. Extracellular vesicle-packaged PIAT from cancer-associated fibroblasts drives neural remodeling by mediating m5C modification in pancreatic cancer mouse models. Sci Transl Med 2024; 16:eadi0178. [PMID: 39018369 DOI: 10.1126/scitranslmed.adi0178] [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: 03/29/2023] [Revised: 02/06/2024] [Accepted: 06/25/2024] [Indexed: 07/19/2024]
Abstract
Perineural invasion (PNI) is a biological characteristic commonly observed in pancreatic cancer. Although PNI plays a key role in pancreatic cancer metastasis, recurrence, and poor postoperative survival, its mechanism is largely unclarified. Clinical sample analysis and endoscopic ultrasonographic elasticity scoring indicated that cancer-associated fibroblasts (CAFs) were closely related to the occurrence of PNI. Furthermore, CAF-derived extracellular vesicles (EVs) were involved in PNI in dorsal root ganglion coculture and mouse sciatic nerve models. Next, we demonstrated that CAFs promoted PNI through extracellular vesicle transmission of PNI-associated transcript (PIAT). Mechanistically, PIAT specifically bound to YBX1 and blocked the YBX1-Nedd4l interaction to inhibit YBX1 ubiquitination and degradation. Furthermore, PIAT enhanced the binding of YBX1 and PNI-associated mRNAs in a 5-methylcytosine (m5C)-dependent manner. Mutation of m5C recognition motifs in YBX1 or m5C sites in downstream target genes reversed PIAT-mediated PNI. Consistent with these findings, analyses using a KPC mouse model demonstrated that the PIAT/YBX1 axis enhanced PNI through m5C modification. Clinical data suggested that the PIAT expression in the serum EVs of patients with pancreatic cancer was associated with the degree of neural invasion and prognosis. Our study revealed the important role of the PIAT/YBX1 signaling axis in the tumor microenvironment (TME) in promoting tumor cell PNI and provided a new target for precise interference with CAFs and RNA methylation in the TME to suppress PNI in pancreatic cancer.
Collapse
Affiliation(s)
- Shangyou Zheng
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Chonghui Hu
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Qing Lin
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Tingting Li
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China
| | - Guolin Li
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510655, People's Republic of China
| | - Qing Tian
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, People's Republic of China
| | - Xiang Zhang
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Tianhao Huang
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
- Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Yuancheng Ye
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
- Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, People's Republic of China
| | - Rihua He
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
- Shantou University Medical College, Shantou, Guangdong 515041, People's Republic of China
| | - Changhao Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, People's Republic of China
| | - Yu Zhou
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
| | - Rufu Chen
- Department of Pancreas Center, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, People's Republic of China
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510655, People's Republic of China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, People's Republic of China
| |
Collapse
|
39
|
Yu M, Ni M, Xu F, Liu C, Chen L, Li J, Xia S, Diao Y, Chen J, Zhu J, Wu X, Tang M, Li J, Ke G. NSUN6-mediated 5-methylcytosine modification of NDRG1 mRNA promotes radioresistance in cervical cancer. Mol Cancer 2024; 23:139. [PMID: 38970106 PMCID: PMC11225205 DOI: 10.1186/s12943-024-02055-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 06/29/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Radioresistance is the leading cause of death in advanced cervical cancer (CC). Dysregulation of RNA modification has recently emerged as a regulatory mechanism in radiation and drug resistance. We aimed to explore the biological function and clinical significance of 5-methylcytosine (m5C) in cervical cancer radiosensitivity. METHODS The abundance of RNA modification in radiotherapy-resistant and sensitive CC specimens was quantified by liquid chromatography-tandem mass spectrometry. The essential RNA modification-related genes involved in CC radiosensitivity were screened via RNA sequencing. The effect of NSUN6 on radiosensitivity was verified in CC cell lines, cell-derived xenograft (CDX), and 3D bioprinted patient-derived organoid (PDO). The mechanisms of NSUN6 in regulating CC radiosensitivity were investigated by integrative m5C sequencing, mRNA sequencing, and RNA immunoprecipitation. RESULTS We found a higher abundance of m5C modification in resistant CC samples, and NSUN6 was the essential m5C-regulating gene concerning radiosensitivity. NSUN6 overexpression was clinically correlated with radioresistance and poor prognosis in cervical cancer. Functionally, higher NSUN6 expression was associated with radioresistance in the 3D PDO model of cervical cancer. Moreover, silencing NSUN6 increased CC radiosensitivity in vivo and in vitro. Mechanistically, NDRG1 was one of the downstream target genes of NSUN6 identified by integrated m5C-seq, mRNA-seq, and functional validation. NSUN6 promoted the m5C modification of NDRG1 mRNA, and the m5C reader ALYREF bound explicitly to the m5C-labeled NDRG1 mRNA and enhanced NDRG1 mRNA stability. NDRG1 overexpression promoted homologous recombination-mediated DNA repair, which in turn led to radioresistance in cervical cancer. CONCLUSIONS Aberrant m5C hypermethylation and NSUN6 overexpression drive resistance to radiotherapy in cervical cancer. Elevated NSUN6 expression promotes radioresistance in cervical cancer by activating the NSUN6/ALYREF-m5C-NDRG1 pathway. The low expression of NSUN6 in cervical cancer indicates sensitivity to radiotherapy and a better prognosis.
Collapse
Affiliation(s)
- Min Yu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Mengdong Ni
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Fei Xu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Chaohua Liu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Lihua Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jiana Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Siyu Xia
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yixin Diao
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jiaxin Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Zhu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Min Tang
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jiajia Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Guihao Ke
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| |
Collapse
|
40
|
Chen B, Deng Y, Hong Y, Fan L, Zhai X, Hu H, Yin S, Chen Q, Xie X, Ren X, Zhao J, Jiang C. Metabolic Recoding of NSUN2-Mediated m 5C Modification Promotes the Progression of Colorectal Cancer via the NSUN2/YBX1/m 5C-ENO1 Positive Feedback Loop. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309840. [PMID: 38769664 PMCID: PMC11267267 DOI: 10.1002/advs.202309840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/11/2024] [Indexed: 05/22/2024]
Abstract
The RNA modification, 5-methylcytosine (m5C), has recently gained prominence as a pivotal post-transcriptional regulator of gene expression, intricately intertwined with various tumorigenic processes. However, the exact mechanisms governing m5C modifications during the onset and progression of colorectal cancer (CRC) remain unclear. Here, it is determined that the m5C methyltransferase NSUN2 exhibits significantly elevated expression and exerts an oncogenic function in CRC. Mechanistically, NSUN2 and YBX1 are identified as the "writer" and "reader" of ENO1, culminating in the reprogramming of the glucose metabolism and increased production of lactic acid in an m5C-dependent manner. The accumulation of lactic acid derived from CRC cells, in turn, activates the transcription of NSUN2 through histone H3K18 lactylation (H3K18la), and induces the lactylation of NSUN2 at the Lys356 residue (K356), which is crucial for capturing target RNAs. Together, these findings reveal an intriguing positive feedback loop involving the NSUN2/YBX1/m5C-ENO1 signaling axis, thereby bridging the connection between metabolic reprogramming and epigenetic remodeling, which may shed light on the therapeutic potential of combining an NSUN2 inhibitor with immunotherapy for CRC.
Collapse
Affiliation(s)
- Baoxiang Chen
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Yanrong Deng
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Yuntian Hong
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Lifang Fan
- Department of PathologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Xiang Zhai
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Heng Hu
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Siyuan Yin
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Quanjiao Chen
- CAS Key Laboratory of Special Pathogens and BiosafetyCAS Center for Influenza Research and Early WarningWuhan Institute of VirologyChinese Academy of SciencesWuhan430064China
| | - Xiaoyu Xie
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Xianghai Ren
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Jianhong Zhao
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Congqing Jiang
- Department of Colorectal and Anal SurgeryZhongnan Hospital of Wuhan UniversityWuhan430071China
- Hubei Key Laboratory of Intestinal and Colorectal DiseasesZhongnan Hospital of Wuhan UniversityWuhan430071China
- Clinical Center of Intestinal and Colorectal Diseases of Hubei ProvinceZhongnan Hospital of Wuhan UniversityWuhan430071China
| |
Collapse
|
41
|
Li H, Liu H, Zhu D, Dou C, Gang B, Zhang M, Wan Z. Biological function molecular pathways and druggability of DNMT2/TRDMT1. Pharmacol Res 2024; 205:107222. [PMID: 38782147 DOI: 10.1016/j.phrs.2024.107222] [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: 04/09/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
5-methylcytosine (m5C) is among the most common epigenetic modification in DNA and RNA molecules, and plays an important role in the animal development and disease pathogenesis. Interestingly, unlike other m5C DNA methyltransferases (DNMTs), DNMT2/TRDMT1 has the double-substrate specificity and adopts a DNMT-similar catalytic mechanism to methylate RNA. Moreover, it is widely involved in a variety of physiological regulatory processes, such as the gene expression, precise protein synthesis, immune response, and disease occurrence. Thus, comprehending the epigenetic mechanism and function of DNMT2/TRDMT1 will probably provide new strategies to treat some refractory diseases. Here, we discuss recent studies on the spatiotemporal expression pattern and post-translational modifications of DNMT2/TRDMT1, and summarize the research advances in substrate characteristics, catalytic recognition mechanism, DNMT2/TRDMT1-related genes or proteins, pharmacological application, and inhibitor development. This review will shed light on the pharmacological design by targeting DNMT2/TRDMT1 to treat parasitic, viral and oncologic diseases.
Collapse
Affiliation(s)
- Huari Li
- Department of Biochemistry and Molecular Biology, College of Laboratory Medicine, Anhui Province Key Laboratory of Cancer Translational Medicine, and The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, No.2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China; College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan, Hubei Province 430070, PR China.
| | - Huiru Liu
- Department of Biochemistry and Molecular Biology, College of Laboratory Medicine, Anhui Province Key Laboratory of Cancer Translational Medicine, and The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, No.2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Daiyun Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan, Hubei Province 430070, PR China; State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, PR China
| | - Chengli Dou
- Department of Biochemistry and Molecular Biology, College of Laboratory Medicine, Anhui Province Key Laboratory of Cancer Translational Medicine, and The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, No.2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Baocai Gang
- Department of Biochemistry and Molecular Biology, College of Laboratory Medicine, Anhui Province Key Laboratory of Cancer Translational Medicine, and The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, No.2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Mengjie Zhang
- Department of Biochemistry and Molecular Biology, College of Laboratory Medicine, Anhui Province Key Laboratory of Cancer Translational Medicine, and The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, No.2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| | - Ziyu Wan
- Department of Biochemistry and Molecular Biology, College of Laboratory Medicine, Anhui Province Key Laboratory of Cancer Translational Medicine, and The First Affiliated Hospital of Bengbu Medical University, Bengbu Medical University, No.2600 Donghai Avenue, Bengbu, Anhui Province 233030, PR China
| |
Collapse
|
42
|
Li Y, Jin H, Li Q, Shi L, Mao Y, Zhao L. The role of RNA methylation in tumor immunity and its potential in immunotherapy. Mol Cancer 2024; 23:130. [PMID: 38902779 PMCID: PMC11188252 DOI: 10.1186/s12943-024-02041-8] [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: 12/24/2023] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
RNA methylation, a prevalent post-transcriptional modification, has garnered considerable attention in research circles. It exerts regulatory control over diverse biological functions by modulating RNA splicing, translation, transport, and stability. Notably, studies have illuminated the substantial impact of RNA methylation on tumor immunity. The primary types of RNA methylation encompass N6-methyladenosine (m6A), 5-methylcytosine (m5C), N1-methyladenosine (m1A), and N7-methylguanosine (m7G), and 3-methylcytidine (m3C). Compelling evidence underscores the involvement of RNA methylation in regulating the tumor microenvironment (TME). By affecting RNA translation and stability through the "writers", "erasers" and "readers", RNA methylation exerts influence over the dysregulation of immune cells and immune factors. Consequently, RNA methylation plays a pivotal role in modulating tumor immunity and mediating various biological behaviors, encompassing proliferation, invasion, metastasis, etc. In this review, we discussed the mechanisms and functions of several RNA methylations, providing a comprehensive overview of their biological roles and underlying mechanisms within the tumor microenvironment and among immunocytes. By exploring how these RNA modifications mediate tumor immune evasion, we also examine their potential applications in immunotherapy. This review aims to provide novel insights and strategies for identifying novel targets in RNA methylation and advancing cancer immunotherapy efficacy.
Collapse
Affiliation(s)
- Yan Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Haoer Jin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qingling Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Liangrong Shi
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yitao Mao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Luqing Zhao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Pathology, School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
43
|
Zan Y, Ding Y. Pancancer Analysis of NSUN2 with a Focus on Prognostic and Immunological Roles in Endometrial Cancer. Reprod Sci 2024:10.1007/s43032-024-01625-5. [PMID: 38900401 DOI: 10.1007/s43032-024-01625-5] [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: 03/18/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024]
Abstract
The significance of NSUN2 in carcinogenesis is gradually being recognized, yet a comprehensive analysis across pan-cancer remains a pivotal void in existing research. In our investigation, we capitalized on the UCSC Xena platform to evaluate NSUN2 expression levels and their prognostic implications across a range of cancer types. Furthermore, we employed the cBioPortal database to delve into the genomic variations of NSUN2 within human cancers. Our study encompassed the use of molecular docking, genomic tumor profiling, and an assessment of the gene's responsiveness to pharmacological treatments. Additionally, we utilized algorithmic techniques to measure the relationship between NSUN2 expression and key clinical biomarkers, such as microsatellite instability (MSI), tumor mutational burden (TMB), and immune cell infiltration. Our results have established a notable association between NSUN2 and endometrial cancer (UCEC), thereby confirming its clinical significance through an analysis of tumoral expression patterns, mutational spectra, methylation profiles, and drug sensitivity. Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) were crucial tools in elucidating the biological roles of NSUN2 in endometrial cancer. Consistently, elevated NSUN2 expression was associated with unfavorable clinical outcomes and was primarily observed in the context of genetic amplifications. Across 22 distinct tumor types, our analysis revealed a notable correlation between NSUN2 expression and various metrics related to immune cell infiltration, tumor stroma, and immune scores. Notably, higher levels of NSUN2 expression have been linked to a reduced response to certain chemotherapeutic agents, including PHA-793887. In UCEC, a positive correlation between NSUN2 methylation and gene expression hints at a potential epigenetic regulatory mechanism underlying cancer progression. Our study highlights the potential of NSUN2 as a key oncogene and its promising role as a therapeutic target as well as a prognostic biomarker for endometrial cancer. This underscores its potential importance in predicting responses to immunotherapy.
Collapse
Affiliation(s)
- Yuxin Zan
- Institute of Biological Medicine, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Yan Ding
- Institute of Biological Medicine, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| |
Collapse
|
44
|
Hsieh MH, Wei Y, Li L, Nguyen LH, Lin YH, Yong JM, Sun X, Wang X, Luo X, Knutson SK, Bracken C, Daley GQ, Powers JT, Zhu H. Liver cancer initiation requires translational activation by an oncofetal regulon involving LIN28 proteins. J Clin Invest 2024; 134:e165734. [PMID: 38875287 PMCID: PMC11290964 DOI: 10.1172/jci165734] [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: 09/28/2022] [Accepted: 06/11/2024] [Indexed: 06/16/2024] Open
Abstract
It is unknown which posttranscriptional regulatory mechanisms are required for oncogenic competence. Here, we show that the LIN28 family of RNA-binding proteins (RBPs), which facilitate posttranscriptional RNA metabolism within ribonucleoprotein networks, is essential for the initiation of diverse oncotypes of hepatocellular carcinoma (HCC). In HCC models driven by NRASG12V/Tp53, CTNNB1/YAP/Tp53, or AKT/Tp53, mice without Lin28a and Lin28b were markedly impaired in cancer initiation. We biochemically defined an oncofetal regulon of 15 factors connected to LIN28 through direct mRNA and protein interactions. Interestingly, all were RBPs and only 1 of 15 was a Let-7 target. Polysome profiling and reporter assays showed that LIN28B directly increased the translation of 8 of these 15 RBPs. As expected, overexpression of LIN28B and IGFBP1-3 was able to genetically rescue cancer initiation. Using this platform to probe components downstream of LIN28, we found that 8 target RBPs were able to restore NRASG12V/Tp53 cancer formation in Lin28a/Lin28b-deficient mice. Furthermore, these LIN28B targets promote cancer initiation through an increase in protein synthesis. LIN28B, central to an RNP regulon that increases translation of RBPs, is important for tumor initiation in the liver.
Collapse
Affiliation(s)
- Meng-Hsiung Hsieh
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yonglong Wei
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Lin Li
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Liem H. Nguyen
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yu-Hsuan Lin
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jung M. Yong
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xuxu Sun
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xun Wang
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xin Luo
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | - George Q. Daley
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - John T. Powers
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Hao Zhu
- Children’s Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
45
|
Zhang X, An K, Ge X, Sun Y, Wei J, Ren W, Wang H, Wang Y, Du Y, He L, Li O, Zhou S, Shi Y, Ren T, Yang YG, Kan Q, Tian X. NSUN2/YBX1 promotes the progression of breast cancer by enhancing HGH1 mRNA stability through m 5C methylation. Breast Cancer Res 2024; 26:94. [PMID: 38844963 PMCID: PMC11155144 DOI: 10.1186/s13058-024-01847-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/21/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND RNA m5C methylation has been extensively implicated in the occurrence and development of tumors. As the main methyltransferase, NSUN2 plays a crucial regulatory role across diverse tumor types. However, the precise impact of NSUN2-mediated m5C modification on breast cancer (BC) remains unclear. Our study aims to elucidate the molecular mechanism underlying how NSUN2 regulates the target gene HGH1 (also known as FAM203) through m5C modification, thereby promoting BC progression. Additionally, this study targets at preliminarily clarifying the biological roles of NSUN2 and HGH1 in BC. METHODS Tumor and adjacent tissues from 5 BC patients were collected, and the m5C modification target HGH1 in BC was screened through RNA sequencing (RNA-seq) and single-base resolution m5C methylation sequencing (RNA-BisSeq). Methylation RNA immunoprecipitation-qPCR (MeRIP-qPCR) and RNA-binding protein immunoprecipitation-qPCR (RIP-qPCR) confirmed that the methylation molecules NSUN2 and YBX1 specifically recognized and bound to HGH1 through m5C modification. In addition, proteomics, co-immunoprecipitation (co-IP), and Ribosome sequencing (Ribo-Seq) were used to explore the biological role of HGH1 in BC. RESULTS As the main m5C methylation molecule, NSUN2 is abnormally overexpressed in BC and increases the overall level of RNA m5C. Knocking down NSUN2 can inhibit BC progression in vitro or in vivo. Combined RNA-seq and RNA-BisSeq analysis identified HGH1 as a potential target of abnormal m5C modifications. We clarified the mechanism by which NSUN2 regulates HGH1 expression through m5C modification, a process that involves interactions with the YBX1 protein, which collectively impacts mRNA stability and protein synthesis. Furthermore, this study is the first to reveal the binding interaction between HGH1 and the translation elongation factor EEF2, providing a comprehensive understanding of its ability to regulate transcript translation efficiency and protein synthesis in BC cells. CONCLUSIONS This study preliminarily clarifies the regulatory role of the NSUN2-YBX1-m5C-HGH1 axis from post-transcriptional modification to protein translation, revealing the key role of abnormal RNA m5C modification in BC and suggesting that HGH1 may be a new epigenetic biomarker and potential therapeutic target for BC.
Collapse
Affiliation(s)
- Xuran Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ke An
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xin Ge
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuanyuan Sun
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jingyao Wei
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Weihong Ren
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450000, China
| | - Han Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yueqin Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Lulu He
- Biobank of the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ouwen Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shaoxuan Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yong Shi
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Tong Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yun-Gui Yang
- China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Quancheng Kan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Xin Tian
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshedong Rd, Zhengzhou, Henan, 450052, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
46
|
Hu J, Li P, Dan Y, Chen Z, Lu Y, Chen X, Yan S. COL8A1 Regulates Esophageal Squamous Carcinoma Proliferation and Invasion Through PI3K/AKT Pathway. Ann Surg Oncol 2024; 31:3502-3512. [PMID: 38429534 DOI: 10.1245/s10434-023-14370-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/15/2023] [Indexed: 03/03/2024]
Abstract
PURPOSE Esophageal squamous carcinoma (ESCC) is a gastrointestinal malignancy with a high mortality, but the tumorigenesis is still unclear, restricting the target therapy development of ESCC. We explored the role of COL8A1 in ESCC development. METHODS Tissue microarrays were used to investigate the expression level of COL8A1 in ESCC tissues. The association between COL8A1 and the overall survival of ESCC patients was assessed. The effect of differential COL8A1 expression on tumor growth was investigated by the xenograft model. The regulation of COL8A1 on tumor growth, migration, and invasion was studied by using ESCC cell lines. The signal transduction pathways involved in COL8A1 were bioinformatically profiled and validated. RESULTS The COL8A1 was significantly expressed in cancerous tissues and was associated with poor prognosis in patients with ESCC. In vivo, the tumor growth obviously declined after inhibition of the COL8A1 expression. The abilities of cell proliferation and invasion were both decreased when the expression of COL8A1 was knockdown in ESCC cell line. Furthermore, we found the inactivation of the PI3K/AKT pathway that was mediated by knockdown of COL8A1 in ESCC cells, which was reversed with COL8A1 overexpression, whereas the cell proliferation and invasion ability were restored. CONCLUSIONS This is the first report that COL8A1 promote ESCC progression, which hopefully will provide a theoretical basis for clinical targeting of ESCC.
Collapse
Affiliation(s)
- Jing Hu
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Pengbo Li
- The Affiliated Lihuili Hospital, Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Yanggang Dan
- Department of Cardiothoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Zhe Chen
- Department of Cardiothoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Yeting Lu
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Xue Chen
- Department of Radiation Oncology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Senxiang Yan
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
47
|
Liu L, Chen Y, Zhang T, Cui G, Wang W, Zhang G, Li J, Zhang Y, Wang Y, Zou Y, Ren Z, Xue W, Sun R. YBX1 Promotes Esophageal Squamous Cell Carcinoma Progression via m5C‐Dependent SMOX mRNA Stabilization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302379. [PMID: 38566431 PMCID: PMC11132058 DOI: 10.1002/advs.202302379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 02/22/2024] [Indexed: 04/04/2024]
Abstract
The modification and recognition of 5-methylcytosine (m5C) are involved in the initiation and progression of various tumor types. However, the precise role and potential mechanism of Y-box-binding protein 1 (YBX1) in esophageal squamous cell carcinoma (ESCC) remains unclear. Here, it is found that YBX1 is frequently upregulated in ESCC compared with matched nontumor tissues. Gain- and loss-of-function assays show that YBX1 promoted the proliferation and metastasis of ESCC cells both in vitro and in vivo. Functional studies revealed that NOP2/Sun RNA methyltransferase family member 2 (NSUN2) is a critical RNA methyltransferase that facilitates YBX1-mediated ESCC progression. Mechanistically, integrated analysis based on RNA immunoprecipitation sequencing (RIP-seq) and m5C methylated RNA immunoprecipitation and sequencing (MeRIP-seq) assays identified spermine oxidase (SMOX) as a target gene containing an m5C site in its coding sequence (CDS) region, which coincided well with the binding site of YBX1. Overexpression of SMOX-WT but not SMOX-Mut partially restored the proliferation and invasion ability of ESCC cells curbed by YBX1 knockdown. Moreover, YBX1 activated the mTORC1 signaling pathway by stabilizing SMOX mRNA. The study reveals that YBX1 promotes ESCC development by stabilizing SMOX mRNA in an m5C-dependent manner, thus providing a valuable therapeutic target for ESCC.
Collapse
Affiliation(s)
- Liwen Liu
- Precision Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Academy of Medical SciencesZhengzhou UniversityZhengzhouHenan450052China
| | - Yu Chen
- Academy of Medical SciencesZhengzhou UniversityZhengzhouHenan450052China
| | - Tao Zhang
- Department of OncologyThe First Hospital of Lanzhou UniversityLanzhouGansu730000China
| | - Guangying Cui
- Precision Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Weiwei Wang
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Guizhen Zhang
- Precision Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Academy of Medical SciencesZhengzhou UniversityZhengzhouHenan450052China
| | - Jianhao Li
- Precision Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Yize Zhang
- Precision Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Yun Wang
- Precision Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Yawen Zou
- Precision Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Zhigang Ren
- Precision Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Wenhua Xue
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Ranran Sun
- Precision Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| |
Collapse
|
48
|
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
|
49
|
Wang J, Ren H, Xu C, Yu B, Cai Y, Wang J, Ni X. Identification of m6A/m5C-related lncRNA signature for prediction of prognosis and immunotherapy efficacy in esophageal squamous cell carcinoma. Sci Rep 2024; 14:8238. [PMID: 38589454 PMCID: PMC11001862 DOI: 10.1038/s41598-024-58743-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/02/2024] [Indexed: 04/10/2024] Open
Abstract
N6-methyladenosine (m6A) and 5-methylcytosine (m5C) RNA modifications have garnered significant attention in the field of epigenetic research due to their close association with human cancers. This study we focus on elucidating the expression patterns of m6A/m5C-related long non-coding RNAs (lncRNAs) in esophageal squamous cell carcinoma (ESCC) and assessing their prognostic significance and therapeutic potential. Transcriptomic profiles of ESCC were derived from public resources. m6A/m5C-related lncRNAs were obtained from TCGA using Spearman's correlations analysis. The m6A/m5C-lncRNAs prognostic signature was selected to construct a RiskScore model for survival prediction, and their correlation with the immune microenvironment and immunotherapy response was analyzed. A total of 606 m6A/m5C-lncRNAs were screened, and ESCC cases in the TCGA cohort were stratified into three clusters, which showed significantly distinct in various clinical features and immune landscapes. A RiskScore model comprising ten m6A/m5C-lncRNAs prognostic signature were constructed and displayed good independent prediction ability in validation datasets. Patients in the low-RiskScore group had a better prognosis, a higher abundance of immune cells (CD4 + T cell, CD4 + naive T cell, class-switched memory B cell, and Treg), and enhanced expression of most immune checkpoint genes. Importantly, patients with low-RiskScore were more cline benefit from immune checkpoint inhibitor treatment (P < 0.05). Our findings underscore the potential of RiskScore system comprising ten m6A/m5C-related lncRNAs as effective biomarkers for predicting survival outcomes, characterizing the immune landscape, and assessing response to immunotherapy in ESCC.
Collapse
Affiliation(s)
- Jianlin Wang
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213003, Jiangsu, China
- Center for Medical Physics, Nanjing Medical University, Changzhou, 213003, Jiangsu, China
| | - Huiwen Ren
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213003, Jiangsu, China
| | - Chao Xu
- Department of Radiotherapy, Jiangyin People's Hospital, Jiangyin, 214400, Jiangsu, China
| | - Bo Yu
- Department of Radiotherapy, Jiangyin People's Hospital, Jiangyin, 214400, Jiangsu, China
| | - Yiling Cai
- Department of Radiotherapy, Jiangyin People's Hospital, Jiangyin, 214400, Jiangsu, China
| | - Jian Wang
- Department of Radiotherapy, Jiangyin People's Hospital, Jiangyin, 214400, Jiangsu, China.
| | - Xinye Ni
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213003, Jiangsu, China.
- Center for Medical Physics, Nanjing Medical University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
50
|
Wu R, Sun C, Chen X, Yang R, Luan Y, Zhao X, Yu P, Luo R, Hou Y, Tian R, Bian S, Li Y, Dong Y, Liu Q, Dai W, Fan Z, Yan R, Pan B, Feng S, Wu J, Chen F, Yang C, Wang H, Dai H, Shu M. NSUN5/TET2-directed chromatin-associated RNA modification of 5-methylcytosine to 5-hydroxymethylcytosine governs glioma immune evasion. Proc Natl Acad Sci U S A 2024; 121:e2321611121. [PMID: 38547058 PMCID: PMC10998593 DOI: 10.1073/pnas.2321611121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/28/2024] [Indexed: 04/02/2024] Open
Abstract
Malignant glioma exhibits immune evasion characterized by highly expressing the immune checkpoint CD47. RNA 5-methylcytosine(m5C) modification plays a pivotal role in tumor pathogenesis. However, the mechanism underlying m5C-modified RNA metabolism remains unclear, as does the contribution of m5C-modified RNA to the glioma immune microenvironment. In this study, we demonstrate that the canonical 28SrRNA methyltransferase NSUN5 down-regulates β-catenin by promoting the degradation of its mRNA, leading to enhanced phagocytosis of tumor-associated macrophages (TAMs). Specifically, the NSUN5-induced suppression of β-catenin relies on its methyltransferase activity mediated by cysteine 359 (C359) and is not influenced by its localization in the nucleolus. Intriguingly, NSUN5 directly interacts with and deposits m5C on CTNNB1 caRNA (chromatin-associated RNA). NSUN5-induced recruitment of TET2 to chromatin is independent of its methyltransferase activity. The m5C modification on caRNA is subsequently oxidized into 5-hydroxymethylcytosine (5hmC) by TET2, which is dependent on its binding affinity for Fe2+ and α-KG. Furthermore, NSUN5 enhances the chromatin recruitment of RBFOX2 which acts as a 5hmC-specific reader to recognize and facilitate the degradation of 5hmC caRNA. Notably, hmeRIP-seq analysis reveals numerous mRNA substrates of NSUN5 that potentially undergo this mode of metabolism. In addition, NSUN5 is epigenetically suppressed by DNA methylation and is negatively correlated with IDH1-R132H mutation in glioma patients. Importantly, pharmacological blockage of DNA methylation or IDH1-R132H mutant and CD47/SIRPα signaling synergistically enhances TAM-based phagocytosis and glioma elimination in vivo. Our findings unveil a general mechanism by which NSUN5/TET2/RBFOX2 signaling regulates RNA metabolism and highlight NSUN5 targeting as a potential strategy for glioma immune therapy.
Collapse
Affiliation(s)
- Ruixin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai200040, China
| | - Chunming Sun
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Neurology, Huashan hospital, Fudan University, Shanghai200040, China
| | - Xi Chen
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Runyue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Yuxuan Luan
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Microbiology, Key Laboratory of Medical Molecular Virology (Ministry of Education/ National Health Commission/ Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Xiang Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Panpan Yu
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan hospital, Fudan University, Shanghai200032, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan hospital, Fudan University, Shanghai200032, China
| | - Ruotong Tian
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Shasha Bian
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Microbiology, Key Laboratory of Medical Molecular Virology (Ministry of Education/ National Health Commission/ Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Yuli Li
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Yinghua Dong
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Logistics, Dalian No.3 People’s hospital Affiliated to Dalian Medical University, Dalian116033, China
| | - Qian Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Weiwei Dai
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Microbiology, Key Laboratory of Medical Molecular Virology (Ministry of Education/ National Health Commission/ Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Zhuoyang Fan
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Rucheng Yan
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Binyang Pan
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai200040, China
| | - Siheng Feng
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Jing Wu
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai200040, China
| | - Fangzhen Chen
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai200040, China
| | - Changle Yang
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai200040, China
| | - Hanlin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Haochen Dai
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Minfeng Shu
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
- Department of Microbiology, Key Laboratory of Medical Molecular Virology (Ministry of Education/ National Health Commission/ Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, China
| |
Collapse
|