1
|
Zhou S, Hui X, Wang W, Zhao C, Jin M, Qin Y, Chen M. SARS-CoV-2 and HCoV-OC43 regulate host m6A modification via activation of the mTORC1 signalling pathway to facilitate viral replication. Emerg Microbes Infect 2025; 14:2447620. [PMID: 39745173 PMCID: PMC11852242 DOI: 10.1080/22221751.2024.2447620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/08/2024] [Accepted: 12/22/2024] [Indexed: 02/25/2025]
Abstract
N6-methyladenosine (m6A) is the most prevalent post-transcriptional modification in eukaryotic RNA and is also present in various viral RNAs, where it plays a crucial role in regulating the viral life cycle. However, the molecular mechanisms through which viruses regulate host RNA m6A methylation are not fully understood. In this study, we reveal that SARS-CoV-2 and HCoV-OC43 infection enhance host m6A modification by activating the mTORC1 signalling pathway. Specifically, the viral non-structural protein nsp14 upregulates the expression of S-adenosylmethionine synthase MAT2A in an mTORC1-dependent manner. This mTORC1-MAT2A axis subsequently stimulates the synthesis of S-adenosylmethionine (SAM). The increase of SAM then enhances the m6A methylation of host RNA and facilitates viral replication. Our findings uncover a molecular mechanism by which viruses regulate host m6A methylation and provide insights into how SARS-CoV-2 hijacks host cellular epitranscriptomic modifications to promote its replication.
Collapse
Affiliation(s)
- Shixiong Zhou
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Xianfeng Hui
- National key laboratory of agricultural microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Weiwei Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Chunbei Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Meilin Jin
- National key laboratory of agricultural microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Yali Qin
- School of Life Sciences, Hubei University, Wuhan, People’s Republic of China
| | - Mingzhou Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
- School of Life Sciences, Hubei University, Wuhan, People’s Republic of China
| |
Collapse
|
2
|
Famà V, Coscujuela Tarrero L, Albanese R, Calviello L, Biffo S, Pelizzola M, Furlan M. Coupling mechanisms coordinating mRNA translation with stages of the mRNA lifecycle. RNA Biol 2025; 22:1-12. [PMID: 40116043 PMCID: PMC11934187 DOI: 10.1080/15476286.2025.2483001] [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: 12/29/2024] [Revised: 03/06/2025] [Accepted: 03/13/2025] [Indexed: 03/23/2025] Open
Abstract
Gene expression involves a series of consequential processes, beginning with mRNA synthesis and culminating in translation. Traditionally studied as a linear sequence of events, recent findings challenge this perspective, revealing coupling mechanisms that coordinate key steps of gene expression, even when spatially and temporally distant. In this review, we focus on translation, the final stage of gene expression, and examine its coupling with key stages of mRNA metabolism: synthesis, processing, export, and decay. For each of these processes, we provide an overview of known instances of coupling with translation. Furthermore, we discuss the role of high-throughput technologies in uncovering these intricate interactions on a genome-wide scale. Finally, we highlight key challenges and propose future directions to advance our understanding of how coupling mechanisms orchestrate robust and adaptable gene expression programs.
Collapse
Affiliation(s)
- Valeria Famà
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan, Italy
- Department of Oncology and Emato-Oncology, University of Milan, Milan, Italy
| | | | | | | | - Stefano Biffo
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Mattia Pelizzola
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Mattia Furlan
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan, Italy
| |
Collapse
|
3
|
Xian Z, Tian L, Yao Z, Cao L, Jia Z, Li G. Mechanism of N6-Methyladenosine Modification in the Pathogenesis of Depression. Mol Neurobiol 2025; 62:5484-5500. [PMID: 39551913 DOI: 10.1007/s12035-024-04614-6] [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: 04/14/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
N6-methyladenosine (m6A) is one of the most common post-transcriptional RNA modifications, which plays a critical role in various bioprocesses such as immunological processes, stress response, cell self-renewal, and proliferation. The abnormal expression of m6A-related proteins may occur in the central nervous system, affecting neurogenesis, synapse formation, brain development, learning and memory, etc. Accumulating evidence is emerging that dysregulation of m6A contributes to the initiation and progression of psychiatric disorders including depression. Until now, the specific pathogenesis of depression has not been comprehensively clarified, and further investigations are warranted. Stress, inflammation, neurogenesis, and synaptic plasticity have been implicated as possible pathophysiological mechanisms underlying depression, in which m6A is extensively involved. Considering the extensive connections between depression and neurofunction and the critical role of m6A in regulating neurological function, it has been increasingly proposed that m6A may have an important role in the pathogenesis of depression; however, the results and the specific molecular mechanisms of how m6A methylation is involved in major depressive disorder (MDD) were varied and not fully understood. In this review, we describe the underlying molecular mechanisms between m6A and depression from several aspects including inflammation, stress, neuroplasticity including neurogenesis, and brain structure, which contain the interactions of m6A with cytokines, the HPA axis, BDNF, and other biological molecules or mechanisms in detail. Finally, we summarized the perspectives for the improved understanding of the pathogenesis of depression and the development of more effective treatment approaches for this disorder.
Collapse
Affiliation(s)
- Zhuohang Xian
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Liangjing Tian
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhixuan Yao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Cao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhilin Jia
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Gangqin Li
- Department of Forensic Psychiatry, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
4
|
Yang L, Ma M, Gao Y, Liu J. Decoding N 6-methyladenosine's dynamic role in stem cell fate and early embryo development: insights into RNA-chromatin interactions. Curr Opin Genet Dev 2025; 91:102311. [PMID: 39908649 DOI: 10.1016/j.gde.2025.102311] [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: 11/09/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
N6-methyladenosine (m6A), a reversible and dynamic RNA modification, plays pivotal roles in regulating stem cell pluripotency and early embryogenesis. Disruptions in m6A homeostasis lead to profound developmental defects, impairing processes such as stem cell self-renewal, lineage specification, oocyte maturation, zygotic genome activation, and maternal RNA degradation after fertilization. Beyond its well-recognized roles in mRNA transport, stability, and translation, recent studies have highlighted m6A's critical role in transcriptional regulation through intricate RNA-chromatin interactions, notably involving chromatin-associated regulatory RNAs (carRNAs) and retrotransposon RNAs. This review delves into the dynamic regulatory landscape of m6A, highlighting its critical interplay with chromatin modifications, and explores its broader implications in stem cell biology and early embryonic development.
Collapse
Affiliation(s)
- Lei Yang
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Mingli Ma
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yawei Gao
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Sycamore Research Institute of Life Sciences, Shanghai 201203, China.
| | - Jun Liu
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China.
| |
Collapse
|
5
|
Ma L, Tao X, Fahim AM, Xu Y, Zhang Y, Li S, Yang G, Pu Y, Wang W, Liu L, Fan T, Wu J, Sun W. Novel insights into the unique characterization of N6-methyladenosine RNA modification and regulating cold tolerance in winter Brassica rapa. Int J Biol Macromol 2025; 303:140460. [PMID: 39919396 DOI: 10.1016/j.ijbiomac.2025.140460] [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: 10/03/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
N6-methyladenosine (m6A) is an mRNA modification considered essential in plants, and is a key player in gene regulation at the transcriptional and translational levels. In present study, we mapped Brassica rapa's whole transcriptome m6A profile under low-temperature stress in different cold tolerant varieties to elucidate the m6A methylation pattern. The distribution of m6A modifications changed significantly under low temperature stress. More 5'UTR m6A was deposited in strong cold-resistant varieties and responded positively to cold resistance by regulating mRNA expression abundance. The increase in m6A abundance was correlated with the increase in mRNA abundance after low temperature stress. ZAT12 might positively regulate its mRNA expression through m6A methylation. MYBC1 might be a negative regulator to cope with low-temperature stress. The hypothetical protein was involved in starch and sucrose metabolic pathways, and that the Low-quality protein was involved in the regulation of DNA binding, DNA-binding, transcription factor activity, and proline biosynthetic processes and leaf senescence pathways. In addition, a number of m6A methyltransferases and m6A demethylases play crucial roles in response to cold stress. These results revealed the critical role of m6A -modified genes under cold stress and provide new insights into the study of cold resistance in winter Brassica rapa.
Collapse
Affiliation(s)
- Li Ma
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China.
| | - Xiaolei Tao
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China
| | - Abbas Muhammad Fahim
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China
| | - Yanxia Xu
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuanyuan Zhang
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China
| | - Shiyi Li
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China
| | - Gang Yang
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuanyuan Pu
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China.
| | - Wangtian Wang
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China.
| | - Lijun Liu
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China
| | - Tingting Fan
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China.
| | - Junyan Wu
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China.
| | - Wancang Sun
- State Key Laboratory of Aridland Crop Science/ College of Agronomy, Gansu Agricultural University, Lanzhou 730070, China.
| |
Collapse
|
6
|
You Q, Yu J, Pan R, Feng J, Guo H, Liu B. Decoding the regulatory roles of circular RNAs in cardiac fibrosis. Noncoding RNA Res 2025; 11:115-130. [PMID: 39759175 PMCID: PMC11697406 DOI: 10.1016/j.ncrna.2024.11.007] [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: 07/30/2024] [Revised: 09/04/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the primary cause of death globally. The evolution of nearly all types of CVDs is characterized by a common theme: the emergence of cardiac fibrosis. The precise mechanisms that trigger cardiac fibrosis are still not completely understood. In recent years, a type of non-coding regulatory RNA molecule known as circular RNAs (circRNAs) has been reported. These molecules are produced during back splicing and possess significant biological capabilities, such as regulating microRNA activity, serving as protein scaffolds and recruiters, competing with mRNA, forming circR-loop structures to modulate transcription, and translating polypeptides. Furthermore, circRNAs exhibit a substantial abundance, notable stability, and specificity of tissues, cells, and time, endowing them with the potential as biomarkers, therapeutic targets, and therapeutic agents. CircRNAs have garnered growing interest in the field of CVDs. Recent investigations into the involvement of circRNAs in cardiac fibrosis have yielded encouraging findings. This study aims to provide a concise overview of the existing knowledge about the regulatory roles of circRNAs in cardiac fibrosis.
Collapse
Affiliation(s)
| | | | - Runfang Pan
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jiaming Feng
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haidong Guo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Baonian Liu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
7
|
Orji OC, Stones J, Rajani S, Markus R, Öz MD, Knight HM. Global Co-regulatory Cross Talk Between m 6A and m 5C RNA Methylation Systems Coordinate Cellular Responses and Brain Disease Pathways. Mol Neurobiol 2025; 62:5006-5021. [PMID: 39499421 PMCID: PMC11880056 DOI: 10.1007/s12035-024-04555-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 10/11/2024] [Indexed: 11/07/2024]
Abstract
N6 adenosine and C5 cytosine modification of mRNAs, tRNAs and rRNAs are regulated by the behaviour of distinct sets of writer, reader and eraser effector proteins which are conventionally considered to function independently. Here, we provide evidence of global cross-regulatory and functional interaction between the m6A and m5C RNA methylation systems. We first show that m6A and m5C effector protein transcripts are subject to reciprocal base modification supporting the existence of co-regulatory post-transcriptional feedback loops. Using global mass spectrometry proteomic data generated after biological perturbation to identify proteins which change in abundance with effector proteins, we found novel co-regulatory cellular response relationships between m6A and m5C proteins such as between the m6A eraser, ALKBH5, and the m5C writer, NSUN4. Gene ontology analysis of co-regulated proteins indicated that m6A and m5C RNA cross-system control varies across cellular processes, e.g. proteasome and mitochondrial mechanisms, and post-translational modification processes such as SUMOylation and phosphorylation. We also uncovered novel relationships between effector protein networks including contributing to intellectual disability pathways. Finally, we provided in vitro confirmation of colocalisation between m6A-RNAs and the m5C reader protein, ALYREF, after synaptic NMDA activation. These findings have important implications for understanding control of RNA metabolism, cellular proteomic responses, and brain disease mechanisms.
Collapse
Affiliation(s)
- Oliver Chukwuma Orji
- Division of Cells, Organisms and Molecular Genetics, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
- Department of Medical Laboratory Sciences, College of Medicine, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Joseph Stones
- Division of Cells, Organisms and Molecular Genetics, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Seema Rajani
- School of Life Sciences Imaging Facility, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Robert Markus
- School of Life Sciences Imaging Facility, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Merve Demirbugen Öz
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Helen Miranda Knight
- Division of Cells, Organisms and Molecular Genetics, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
8
|
Wang H, Wang H, Jia Y, Jin X, Wu H, Yang S, Zhao L, Zhang H, Gu L. The RNA m 6A Methyltransferase PheMTA1 and PheMTA2 of Moso Bamboo Regulate Root Development and Resistance to Salt Stress in Plant. PLANT, CELL & ENVIRONMENT 2025. [PMID: 40165397 DOI: 10.1111/pce.15494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/01/2025] [Accepted: 03/15/2025] [Indexed: 04/02/2025]
Abstract
As the most prevalent RNA modification in eukaryotes, N6-methyladenosine (m6A) plays a crucial role in regulating various biological processes in plants, including embryonic development and flowering. However, the function of m6A RNA methyltransferase in moso bamboo remains poorly understood. In this study, we identified two m6A methyltransferases in moso bamboo, PheMTA1 and PheMTA2. Overexpression of PheMTA1 and PheMTA2 significantly promoted root development and enhanced salt tolerance in rice. Using the HyperTRIBE method, we fused PheMTA1 and PheMTA2 with ADARcdE488Q and introduced them into rice. RNA sequencing (RNA-seq) of the overexpressing rice identified the target RNAs bound by PheMTA1 and PheMTA2. PheMTA1 and PheMTA2 bind to OsATM3 and OsSF3B1, which were involved in the development of root and salt resistance. Finally, we revealed the effects of transcription or alternative splicing on resistance-related genes like OsRS33, OsPRR73, OsAPX2 and OsHAP2E, which are associated with the observed phenotype. In conclusion, our study demonstrates that the m6A methyltransferases PheMTA1 and PheMTA2 from moso bamboo are involved in root development and enhance plant resistance to salt stress.
Collapse
Affiliation(s)
- Huihui Wang
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Huiyuan Wang
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yue Jia
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Basic Forestry and Proteomics Research Center, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaoxia Jin
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hongwei Wu
- College of Forestry, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Siyu Yang
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Basic Forestry and Proteomics Research Center, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Liangzhen Zhao
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Basic Forestry and Proteomics Research Center, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hangxiao Zhang
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Basic Forestry and Proteomics Research Center, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Lianfeng Gu
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Basic Forestry and Proteomics Research Center, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
9
|
Sun N, Wang S, Liu J, Zhang P, Chang Y, Li H, Zhao K, Liu Y, Huang M, Hu Y, Lin Z, Lu Y, Jiang G, Chen W, Huang C, Jin H. XIAP promotes metastasis of bladder cancer cells by ubiquitylating YTHDC1. Cell Death Dis 2025; 16:205. [PMID: 40133252 PMCID: PMC11937301 DOI: 10.1038/s41419-025-07545-9] [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: 06/06/2024] [Revised: 10/24/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
X-linked inhibitor of apoptosis protein (XIAP), a member of the IAP family, is overexpressed in a variety of tumors and plays an important role in tumor progression. Increasing evidence suggests that XIAP promotes metastasis of bladder cancer but the underlying mechanism is not very clear. The RNA N6-methyladenosine (m6A) reader YTHDC1 regulates RNA splicing, nuclear transport, and mRNA stability and is a potential tumor target; however, its ubiquitin E3 ligase has not been described. In this study, screening of proteins that specifically interact with XIAP identified YTHDC1 as its degradation substrate. Ectopic overexpression of XIAP promoted degradation of YTHDC1, and knockout of XIAP upregulated YTHDC1, which inhibited metastasis of bladder cancer. Furthermore, YTHDC1 reduced the expression of matrix metalloproteinase-2 (MMP-2) by destabilizing its mRNA. These experiments revealed that XIAP promotes ubiquitination of YTHDC1, positively regulating expression of the MMP-2 and promoting metastasis of bladder cancer. Collectively, these findings demonstrate that XIAP is a critical regulator of YTHDC1 and pinpoint the XIAP/YTHDC1/MMP-2 axis as a promising target for the treatment of bladder cancer.
Collapse
Affiliation(s)
- Ning Sun
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sijia Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianting Liu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peipei Zhang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yixin Chang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongyan Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kun Zhao
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yijie Liu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mingzhi Huang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Hu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenni Lin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongyong Lu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guosong Jiang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Chen
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Chuanshu Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
10
|
Tsao N, Lombardi PM, Park A, Olabode J, Rodell R, Sun H, Padmanaban S, Brickner JR, Tsai MS, Pollina EA, Chen CK, Mosammaparast N. YTHDC1 cooperates with the THO complex to prevent RNA-damage-induced DNA breaks. Mol Cell 2025; 85:1085-1100.e9. [PMID: 40037355 DOI: 10.1016/j.molcel.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 12/05/2024] [Accepted: 02/05/2025] [Indexed: 03/06/2025]
Abstract
Certain environmental toxins and chemotherapeutics are nucleic acid-damaging agents, causing adducts in DNA and RNA. While most of these adducts occur in RNA, the consequences of RNA damage are largely unexplored. Here, we demonstrate that nuclear RNA damage can result in loss of genome integrity in human cells. Specifically, we show that YTHDC1 regulates alkylation damage responses with the THO complex (THOC). In addition to its established binding to N6-methyladenosine (m6A), YTHDC1 binds to chemically induced N1-methyladenosine (m1A). Without YTHDC1, cells have greater alkylation damage sensitivity and increased DNA breaks, which are rescued by an RNA-specific dealkylase. These RNA-damage-induced DNA breaks (RDIBs) depend on R-loop formation, which is converted to DNA breaks by the XPG nuclease. Strikingly, in the absence of YTHDC1 or THOC, a nuclear RNA m1A methyltransferase is sufficient to induce DNA breaks. Our results provide mechanistic insight into how damaged RNAs can impact genomic integrity.
Collapse
Affiliation(s)
- Ning Tsao
- Department of Pathology & Immunology, Center for Genome Integrity, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Patrick M Lombardi
- Department of Science, Mount St. Mary's University, Emmitsburg, MD 21727, USA
| | - Ajin Park
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jennifer Olabode
- Department of Pathology & Immunology, Center for Genome Integrity, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rebecca Rodell
- Department of Pathology & Immunology, Center for Genome Integrity, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hua Sun
- Department of Pathology & Immunology, Center for Genome Integrity, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shilpa Padmanaban
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua R Brickner
- Department of Pathology & Immunology, Center for Genome Integrity, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Miaw-Sheue Tsai
- Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Elizabeth A Pollina
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chun-Kan Chen
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nima Mosammaparast
- Department of Pathology & Immunology, Center for Genome Integrity, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
11
|
King EM, Midkiff A, McClain K, Kim S, Panfil AR. YTHDF1 and YTHDC1 m 6A reader proteins regulate HTLV-1 tax and hbz activity. J Virol 2025; 99:e0206324. [PMID: 39902961 PMCID: PMC11915865 DOI: 10.1128/jvi.02063-24] [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: 12/04/2024] [Accepted: 01/12/2025] [Indexed: 02/06/2025] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is a retrovirus responsible for adult T-cell leukemia/lymphoma (ATLL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), a progressive neurodegenerative disease. Regulation of viral gene expression plays a key role in viral persistence and pathogenesis. However, the molecular mechanisms underlying this fine-tuned regulation remain poorly understood. Little is known regarding RNA chemical modifications of HTLV-1 RNA and how these affect viral biology and disease development. Post-transcriptional chemical modification of RNA is common in eukaryotes, with N6-methyladenosine (m6A) being the most prevalent. In this study, we investigated the role of m6A RNA modifications on HTLV-1 gene expression. Using MeRIP-Seq, we mapped the sites of m6A modification to the 3' end of the viral genome. We found HTLV-1 RNA, as well as viral oncogene transcripts tax and hbz, contained m6A modifications. m6A-depletion in HTLV-1-transformed cells decreased sense-derived viral genes (Tax, Gag, and Env) and increased antisense-derived Hbz expression. Tax and hbz transcripts were bound by reader proteins YTHDF1 and YTHDC1 in a panel of HTLV-1 T-cell lines. Using expression vectors and shRNA-mediated knockdown, we found that YTHDF1 had opposing effects on viral gene expression, decreasing sense-derived viral genes and increasing antisense-derived Hbz. Upon further study, the YTHDF1 effects on tax abundance were dependent on tax m6A deposition. The nuclear m6A reader protein YTHDC1 affected the abundance of both sense- and antisense-derived viral transcripts and specifically enhanced the nuclear export of tax transcript. Collectively, our results demonstrate global m6A levels and m6A reader proteins YTHDF1 and YTHDC1 regulate HTLV-1 gene expression.IMPORTANCEHuman T-cell leukemia virus type 1 (HTLV-1) persistence and pathogenesis are controlled through tight regulation of viral gene expression. The fate of RNA can be controlled by epigenetic modifications that impact gene expression without altering the DNA sequence. Our study details the impact of N6-methyladenosine (m6A) RNA chemical modifications on HTLV-1 gene expression. We found that reductions in global m6A levels affected viral gene expression, decreasing Tax and other sense-derived viral genes, whereas increasing the antisense-derived Hbz. Our results suggest the oncogenic viral transcripts, tax and hbz, are m6A-modified in cells. We found that these viral RNA modifications are interpreted by reader proteins YTHDF1 and YTHDC1, which dictate the fate of the viral RNA. Understanding HTLV-1 RNA chemical modifications offers potential insights into novel therapeutic strategies for HTLV-1-associated diseases.
Collapse
Affiliation(s)
- Emily M King
- Center for Retrovirus Research and Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Amanda Midkiff
- Center for Retrovirus Research and Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Karsyn McClain
- Center for Retrovirus Research and Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Sanggu Kim
- Center for Retrovirus Research and Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Amanda R Panfil
- Center for Retrovirus Research and Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
- Comprehensive Cancer Center and Solove Research Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
12
|
Cosseddu C, Succu S, Frau A, Mossa F, Versace SV, Brevini TAL, Ledda S, Bebbere D. m6A RNA methylation dynamics during in vitro maturation of cumulus-oocyte complexes derived from adult or prepubertal sheep. J Assist Reprod Genet 2025:10.1007/s10815-025-03444-2. [PMID: 40097858 DOI: 10.1007/s10815-025-03444-2] [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: 10/28/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
PURPOSE N6-methyladenosine (m6A) is the most prevalent base epigenetic modification within eukaryotic mRNAs. It participates in post-transcriptional regulation, including maternal RNA maintenance and decay in mouse oocytes and during maternal-to-zygotic transition. The landscape in other mammalian species remains largely unexplored. The present work analyzed m6A dynamics in sheep cumulus oocyte complexes (COCs), during in vitro maturation. To explore potential relationships with oocyte developmental competence, a previously established model consisting of oocytes derived from adult and prepubertal sheep was adopted. METHODS m6a dynamics were analyzed in terms of m6A RNA methylation abundance in cumulus cells (CCs) by colorimetric assay and expression of key m6A methylation-related proteins (METTL3, METTL14, METTL16, VIRMA, YTHDC1, YTHDC2, YTHDF2, YTHDF3, ALKBH5, and FTO) in both cumulus cells and oocytes by real-time PCR. RESULTS We report the dynamics of m6A in sheep COCs, and reveal alterations in both oocytes and cumulus cells derived from prepubertal donors. These changes were observed in terms of m6A RNA methylation levels and transcript dynamics of several m6A methylation-related proteins. Notably, our study shows that dysregulations occur after IVM. CONCLUSION Overall, this work describes for the first time the dynamics of m6A in sheep COCs and uncovers the involvement of m6A RNA methylation in oocyte developmental potential.
Collapse
Affiliation(s)
- Chiara Cosseddu
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy
| | - Sara Succu
- Department of Veterinary Medicine, Anatomy, University of Sassari, 07100, Sassari, Italy
| | - Adele Frau
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy
| | - Francesca Mossa
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy
| | - Sylvia Virginie Versace
- Department of Veterinary Medicine, Veterinary Teaching Hospital, University of Sassari, 07100, Sassari, Italy
| | - Tiziana A L Brevini
- Laboratory of Biomedical Embryology, Department of Veterinary Medicine and Animal Science and Center for Stem Cell Research, University of Milano, 26900, Lodi, Italy
| | - Sergio Ledda
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy
| | - Daniela Bebbere
- Department of Veterinary Medicine, Obstetrics and Gynecology Clinics, University of Sassari, 07100, Sassari, Italy.
| |
Collapse
|
13
|
Chen C, Zhou W, Zhang Q, He D, Zhao Y, Liu Z, Xia P, Li Q, Ye Z. OSGEP, A Negative Ferroptotic Regulator, Alleviates Cerebral Ischemia-Reperfusion Injury Through Modulating m 6A Methylation of GPX4 mRNA. Neurochem Res 2025; 50:122. [PMID: 40100474 DOI: 10.1007/s11064-025-04367-1] [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: 10/18/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) is a devastating condition that triggers neuronal death and cerebral infarction. O-sialoglycoprotein endopeptidase (OSGEP), identified as a crucial element of the highly conserved KEOPS complex, regulated cellular proliferation and mitochondrial metabolism. Despite its known role in cellular homeostasis, the potential contribution of OSGEP to the development of CIRI remains elusive. This study was designed to investigate the potential role of ferroptosis in the pathogenesis of CIRI and indicate whether OSGEP could suppress ferroptosis to alleviate CIRI by modulating GPX4 m6A methylation. To this end, MCAO and OGD/R models were employed to closely simulate the CIRI. The potent ferroptosis inhibitors conferred prominent neuroprotection in both in vivo and in vitro models. Moreover, OSGEP expression level was not only downregulated in MCAO-treated mice and in cultured cerebrocortical neurons subjected to OGD/R, but also it was related to the prognosis of acute ischemic stroke (AIS) cases. Additionally, OSGEP overexpression exerted potent anti-ferroptotic effects in both MCAO and OGD/R models, while OSGEP depletion exhibited the opposite effect. Moreover, OSGEP regulated GPX4 expression by modulating m6A methylation of its mRNA. Furthermore, the inhibitory effect of OSGEP on ferroptosis was dependent on the presence of GPX4. Specifically, OSGEP knockout exacerbated ferroptosis-like cell death under MCAO condition. Besides, OSGEP regulated GPX4 mRNA stability through competition with YTHDC1 for binding to GPX4 mRNA and forming a complex with HNRNPUL1 in the neuronal primary cultures subjected to OGD/R. These findings highlighted the critical role of OSGEP, as a new contributing anti-ferroptotic factor, in the pathogenesis of CIRI.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wanqing Zhou
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Qian Zhang
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Dehao He
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Yanfei Zhao
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
| | - Zhuoyi Liu
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, China
| | - Pingping Xia
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, China
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, 410008, China.
| |
Collapse
|
14
|
Liu X, Zhang L, Chen J, Shao W. Decoding intricate interactions between m6A modification with mRNAs and non-coding RNAs in cervical cancer: Molecular mechanisms and clinical implications. Cell Signal 2025; 131:111745. [PMID: 40107480 DOI: 10.1016/j.cellsig.2025.111745] [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: 10/10/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
N6-methyladenosine (m6A) methylation is the most prevalent RNA modification that is regulated by three regulatory factors: "writers", "erasers" and "readers". m6A modification regulates RNA stability and other mechanisms, including translation, cleavage, and degradation. Current research has demonstrated that m6A methylation is involved in the regulation of occurrence and development of cancers by controlling the expression of cancer-related genes. This review summarizes the role of m6A modification on messenger RNAs (mRNAs) and non-coding RNAs (ncRNAs) in cervical cancer (CC). We highlight the dual role of m6A regulatory factors, which act as oncogenes or tumor suppressors depending on the cellular context and downstream targets. Additionally, we examine how ncRNAs reciprocally regulate m6A modification in two ways: by guiding the deposition or removal of m6A modifications on RNA targets, and by modulating the expression of m6A regulatory factors. These interactions further contribute to tumor progression. Furthermore, the therapeutic potential of targeting m6A modification has been emphasized in CC. Moreover, recent advances in small-molecule inhibitors targeting m6A regulators and RNA-based therapies which may offer new treatment strategies have been summarized. Finally, we discuss the current challenges in m6A modification research and provide suggestions for future research directions. This review aims to deepen the understanding of m6A modification in CC and contribute to the development of targeted and personalized treatment strategies.
Collapse
Affiliation(s)
- Xuefei Liu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, China; First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Lizhi Zhang
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Ji Chen
- Department of Obstetrics, The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, China
| | - Wei Shao
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
15
|
Gao M, Wu Y, Zhang L, Zhou Y, Liu H, Zhang W, Wang S, Cui Y. YTHDC1 negatively regulates UBE3A to influence RAD51 ubiquitination and inhibit apoptosis in colorectal cancer cells. Sci Rep 2025; 15:8857. [PMID: 40087295 PMCID: PMC11909180 DOI: 10.1038/s41598-025-92925-6] [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: 12/02/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
YTHDC1, a key protein in the m6A-related regulatory network within cells, is involved in multiple cellular processes such as chromatin-related regulation, RNA splicing, and nuclear export. Understanding its role in colorectal cancer (CRC) development and DNA damage repair is critical for the advancement of treatment strategies. Our study found that YTHDC1 was highly expressed in high-malignancy CRC tissues compared with low-malignancy ones. Upon silencing YTHDC1, we observed a pronounced suppression of the proliferation of CRC cell lines, accompanied by a substantial increase in cell apoptosis. Furthermore, we identified RAD51 as a crucial downstream target of YTHDC1. Knocking down YTHDC1 led to a notable decrease in RAD51 protein levels, and silencing RAD51 also inhibited cancer cell proliferation. Interestingly, RNA-sequencing data indicated that the YTHDC1 deletion did not affect RAD51 transcription. However, Western blot revealed that this deletion increased the ubiquitination of RAD51, likely due to the upregulated E3 ligase UBE3A. Ubiquitination experiments subsequently confirmed that RAD51 is indeed one of the substrates of UBE3A. In summary, our study provides novel insights into how YTHDC1 modulates the expression of RAD51 through post-translational modifications. These findings offer valuable information that may potentially contribute to the development of more effective therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Mingwei Gao
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yueguang Wu
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Li Zhang
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yan Zhou
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huijuan Liu
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Weimin Zhang
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shubin Wang
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China.
| | - Yongping Cui
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China.
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
16
|
Huang X, Zhang J, Cun Y, Ye M, Ren Z, Guo W, Ma X, Liu J, Luo W, Sun X, Shao J, Wu Z, Zhu X, Wang J. Spatial control of m 6A deposition on enhancer and promoter RNAs through co-acetylation of METTL3 and H3K27 on chromatin. Mol Cell 2025:S1097-2765(25)00147-9. [PMID: 40101711 DOI: 10.1016/j.molcel.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/08/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Interaction between the N6-methyladenosine (m6A) methyltransferase METTL3 and METTL14 is critical for METTL3 to deposit m6A on various types of RNAs. It remains to be uncovered whether there is spatial control of m6A deposition on different types of RNAs. Here, through genome-wide CRISPR-Cas9 screening in the A549 cell line, we find that H3K27ac acetylase p300-mediated METTL3 acetylation suppresses the binding of METTL3 on H3K27ac-marked chromatin by inhibiting its interaction with METTL14. Consistently, p300 catalyzing the acetylation of METTL3 specifically occurs on H3K27ac-marked chromatin. Disruptive mutations on METTL3 acetylation sites selectively promote the m6A of chromatin-associated RNAs from p300-bound enhancers and promoters marked by H3K27ac, resulting in transcription inhibition of ferroptosis-inhibition-related genes. In addition, PAK2 promotes METTL3 acetylation by phosphorylating METTL3. Inhibition of PAK2 promotes ferroptosis in a manner that depends on the acetylation of METTL3. Our study reveals a spatial-selective way to specifically regulate the deposition of m6A on enhancer and promoter RNAs.
Collapse
Affiliation(s)
- Xiang Huang
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Medical College of Jiaying University, Meizhou 514031, China
| | - Jie Zhang
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Yixian Cun
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Meijun Ye
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhijun Ren
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Medical College of Jiaying University, Meizhou 514031, China
| | - Wenbing Guo
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Medical College of Jiaying University, Meizhou 514031, China
| | - Xiaojun Ma
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiayin Liu
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Weiwei Luo
- GeneMind Biosciences Company Limited, Shenzhen 518000, China
| | - Xiang Sun
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Jingwen Shao
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Zehong Wu
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaofeng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 518000, China
| | - Jinkai Wang
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
17
|
Qin C, Xu F, Yue B, Zhong J, Chai Z, Wang H. SRSF3 and hnRNP A1-mediated m6A-modified circCDK14 regulates intramuscular fat deposition by acting as miR-4492-z sponge. Cell Mol Biol Lett 2025; 30:26. [PMID: 40038607 DOI: 10.1186/s11658-025-00699-6] [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: 07/12/2024] [Accepted: 02/06/2025] [Indexed: 03/06/2025] Open
Abstract
The intramuscular fat (IMF) content of yak beef is critical for determining its quality. Circular RNAs (circRNAs) are a group of endogenous non-coding RNAs that have emerged as important factors in the regulation of IMF deposition. However, the molecular mechanisms through which circRNAs regulate IMF deposition, particularly in yaks, remain unclear. In the present study, a novel circRNA, circCDK14 (originating from the yak's CDK14 gene), was identified by sequencing and RNase R treatment. In our previous study, we successfully established a ceRNA network map and identified miR-4492-z, which interacts with circCDK14. Furthermore, using methylation prediction software, we predicted two genes, SRSF3 and hnRNP A1, that have a strong binding relationship with circCDK14; existing research has confirmed their close association with m6A methylation modifications. On the basis of these findings, we comprehensively evaluated the effects of circCDK14, miR-4492-z, SRSF3 and hnRNP A1 on the proliferation and differentiation of yak intramuscular pre-adipocytes using EdU, CCK-8, BODIPY, Oil Red O and qRT-PCR analyses. Mechanistically, the interaction between circCDK14 and miR-4492-z was validated using a dual-luciferase reporter gene assay and rescue experiments. RIP assays revealed the binding interaction of circCDK14 with SRSF3 and hnRNP A1. The MeRIP experiments showed modification of circCDK14 methylation, with SRSF3 and hnRNP A1 promoting the methylation and translocation of circCDK14 from the nucleus to the cytoplasm. In summary, our results suggest that m6A-modified circCDK14 plays a crucial role as an miR-4492-z sponge in regulating IMF deposition in yaks and that the nuclear export of circCDK14 correlates with the expression levels of SRSF3 and hnRNP A1. This study provides a theoretical basis for the improvement of yak meat quality and promotes the development of molecular yak breeding.
Collapse
Affiliation(s)
- Chunyu Qin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610225, China
| | - Fang Xu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610225, China
| | - Binglin Yue
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610225, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610225, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610225, China
| | - Hui Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, 610225, China.
| |
Collapse
|
18
|
Lancaster CL, Moberg KH, Corbett AH. Post-Transcriptional Regulation of Gene Expression and the Intricate Life of Eukaryotic mRNAs. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70007. [PMID: 40059537 PMCID: PMC11949413 DOI: 10.1002/wrna.70007] [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: 08/22/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025]
Abstract
In recent years, there has been a growing appreciation for how regulatory events that occur either co- or post-transcriptionally contribute to the control of gene expression. Messenger RNAs (mRNAs) are extensively regulated throughout their metabolism in a precise spatiotemporal manner that requires sophisticated molecular mechanisms for cell-type-specific gene expression, which dictates cell function. Moreover, dysfunction at any of these steps can result in a variety of human diseases, including cancers, muscular atrophies, and neurological diseases. This review summarizes the steps of the central dogma of molecular biology, focusing on the post-transcriptional regulation of gene expression.
Collapse
Affiliation(s)
- Carly L. Lancaster
- Department of Biology, Emory College of Arts and Sciences, Atlanta, Georgia, USA
- Department of Cell Biology Emory University School of Medicine, Atlanta, Georgia, USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University Atlanta, Georgia, USA
| | - Kenneth H. Moberg
- Department of Cell Biology Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anita H. Corbett
- Department of Biology, Emory College of Arts and Sciences, Atlanta, Georgia, USA
| |
Collapse
|
19
|
Ru W, Cheng J, Gao Y, Yang K, Qi A, Zhang X, Qi X, Lan X, Liu W, Huang B, Chen H. METTL3-mediated m 6A modification regulates muscle development by promoting TM4SF1 mRNA degradation in P-body via YTHDF2. Int J Biol Macromol 2025; 295:139576. [PMID: 39778834 DOI: 10.1016/j.ijbiomac.2025.139576] [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: 08/23/2024] [Revised: 12/06/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
N6-methyladenosine (m6A), a well-known post-transcriptional modification, is implicated in diverse cellular and physiological processes. However, much remains unknown regarding the precise role and mechanism of m6A modification on muscle development. In this study, we make observation that the levels of m6A and METTL3 are markedly elevated during the differentiation phase (DM) compared to the growth phase (GM) in both C2C12 and bovine myoblasts. Notably, deletion of METTL3 decreased m6A levels, and promoted myoblast proliferation, inhibited myoblast differentiation in vitro. By performing m6A sequencing in both GM and DM myoblast, we further identified that TM4SF1 is involved in m6A -regulated muscle development. Mechanistically, METTL3 increases m6A-modified TM4SF1 transcripts, and subsequently YTHDF2 promotes TM4SF1 mRNA degradation in P-body through liquid-liquid phase separation (LLPS). Additionally, the rescue experiments in vivo showed that overexpressing METTL3 could rescue the attenuated myogenesis induced by TM4SF1 overexpression during muscle regeneration in mice. Collectively, our findings shed light on a regulatory mechanism by which m6A modulates muscle development and raise a new model for m6A-mediated mRNA degradation within P-bodies.
Collapse
Affiliation(s)
- Wenxiu Ru
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jie Cheng
- Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yuee Gao
- Yunnan Academy of Grassland and Animal Science, Kunming 650212, China
| | - Kai Yang
- Yunnan Academy of Grassland and Animal Science, Kunming 650212, China
| | - Ao Qi
- Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaoyan Zhang
- Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xinglei Qi
- Bureau of Animal Husbandry of Biyang County, Biyang, Henan 463700, China
| | - Xianyong Lan
- Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wujun Liu
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China.
| | - Bizhi Huang
- Yunnan Academy of Grassland and Animal Science, Kunming 650212, China.
| | - Hong Chen
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China.
| |
Collapse
|
20
|
Sun B, Yang J, Wang Z, Wang Z, Feng W, Li X, Liu S, Li J, Zhu Y, Zhang P, Wang W. Exon junction complexes regulate osteoclast-induced bone resorption by influencing the NFATc1 m6A distribution through the "shield effect". Clin Transl Med 2025; 15:e70266. [PMID: 40051055 PMCID: PMC11885169 DOI: 10.1002/ctm2.70266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/19/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND The distribution of the m6A methylation modification on the transcriptome is highly regionally selective and is mainly concentrated in abnormally long exons and stop codons. However, in-depth research on the selective mechanism of m6A methylation is still lacking. METHODS In this research, meRIP sequencing, mRNA sequencing, meRIP, luciferase reporter assays and CRISPR/Cas9 conditional knockout mice were used to elucidate the distribution characteristics of NFATc1 m6A. RESULTS METTL14 controls osteoclast-mediated bone resorption by means of the methylation (4249 A) of the NFATc1 gene during osteoclast differentiation. Exon junction complexes (EJCs) selectively protect the m6A methylation sites of the NFATc1 gene. When the methylation sites are located within short exon fragments (50-200 nt), EJCs prevent their hypermethylation and degradation through the "shield effect"; when the methylation sites are located in the 3' UTR region or long exon fragments (greater than 300 nt), the "shield effect" disappears. Downstream, YTHDF2 induced the degradation of hypermethylation NFATc1 transcripts without site restriction. CONCLUSIONS EJCs act as "shields" to regulate the m6A region selectivity of the NFATc1 gene, thereby determining the characteristics of m6A distribution in the gene. Importantly, EJCs can raise the level of m6A methylation of NFATc1 and degrade its mRNA, thereby inhibiting osteoclast differentiation and preserving bone mass. These results will be helpful for identifying potential molecular targets for osteoporosis treatment. KEY POINTS METTL14 controls osteoclast-mediated bone resorption by means of the methylation (4249 A) of the NFATc1 gene during osteoclast differentiation. Exon junction complexes (EJCs) protect the remaining methylation sites of the NFATc1 gene (located in the inner exon fragment of 50-200 nt) from hypermethylation and degradation. The "shield effect" disappears when the exon fragment is extended to 300 nt. Downstream, YTHDF2 induced the degradation of hypermethylation NFATc1 transcripts without site restriction.
Collapse
Affiliation(s)
- Bao Sun
- Department of Oral PathologyShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Jin‐Gang Yang
- Department of StomatologyTongren Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhe Wang
- Department of GynecologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Zheng Wang
- Concordia Institute for Information Systems Engineering, Concordia UniversityMontrealQCCanada
| | - Wei Feng
- Department of EndodonticsCentral Laboratory of Jinan Stomatological Hospital, Jinan Key Laboratory of Oral Tissue Regeneration, Shandong Provincial Health Commission Key Laboratory of Oral Diseases and Tissue RegenerationJinanShandongPeople's Republic of China
| | - Xing Li
- Department of Oral Maxillofacial‐Head Neck OncologyShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Sheng‐Nan Liu
- Department of Oral PathologyShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Jiang Li
- Department of Oral PathologyShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Ya‐Qin Zhu
- Department of General DentistryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Ping Zhang
- Department of Oral and Maxillofacial SurgeryAffiliated Hospital of Stomatology, Nanjing Medical UniversityNanjingChina
| | - Wei Wang
- Department of General DentistryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| |
Collapse
|
21
|
Chen X, Yuan Y, Zhou F, Li L, Pu J, Jiang X. m6A RNA methylation: a pivotal regulator of tumor immunity and a promising target for cancer immunotherapy. J Transl Med 2025; 23:245. [PMID: 40022120 PMCID: PMC11871626 DOI: 10.1186/s12967-025-06221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
M6A modification is one of the most common regulatory mechanisms of gene expression in eukaryotic cells, influencing processes such as RNA splicing, degradation, stability, and protein translation. Studies have shown that m6A methylation is closely associated with tumorigenesis and progression, and it plays a key regulatory role in tumor immune responses. m6A modification participates in regulating the differentiation and maturation of immune cells, as well as related anti-tumor immune responses. In the tumor microenvironment, m6A modification can also affect immune cell recruitment, activation, and polarization, thereby promoting or inhibiting tumor cell proliferation and metastasis, and reshaping the tumor immune microenvironment. In recent years, immunotherapies for tumors, such as immune checkpoint inhibitors and adoptive cell immunotherapy, have been increasingly applied in clinical settings, achieving favorable outcomes. Targeting m6A modifications to modulate the immune system, such as using small-molecule inhibitors to target dysregulated m6A regulatory factors or inducing immune cell reprogramming, can enhance anti-tumor immune responses and strengthen immune cell recognition and cytotoxicity against tumor cells. m6A modification represents a new direction in tumor immunotherapy with promising clinical potential. This review discusses the regulatory role of m6A methylation on immune cells and tumor immune responses and explores new strategies for immunotherapy.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yixiao Yuan
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Fan Zhou
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Jun Pu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650223, China.
- NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, Yunnan, 650500, China.
| | - Xiulin Jiang
- Department of Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
22
|
Kueck NA, Hüwel S, Hoffmann A, Rentmeister A. Quantification of Propargylated RNA Nucleosides After Metabolic Labeling Via the Methylation Pathway. Chembiochem 2025:e202400986. [PMID: 39993262 DOI: 10.1002/cbic.202400986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/19/2025] [Accepted: 02/24/2025] [Indexed: 02/26/2025]
Abstract
RNA modifications are involved in numerous biological processes and vary in different cell types. Methylation is the most widespread type of RNA modification and occurs via S-adenosyl-L-methionine (SAM). We recently developed a metabolic labeling approach based on intracellular formation of a clickable SAM analog (SeAdoYn) and demonstrated its use in mapping methyltransferase (MTase) target sites in mRNA from HeLa cells. Here we investigate how metabolic labeling via the clickable SAM analog modifies four different nucleosides in RNA of HEK293T in comparison to HeLa cells. We find that HEK293T cells retain higher cell viability upon feeding the clickable metabolic SAM precursor. In poly(A)+ RNA we find high Aprop/A levels (0.04 %) and in total RNA (but not poly(A)+ RNA) we detect prop3C, which had not been detected previously in HeLa cells. We discuss the findings in the context of data from the literature with respect to mRNA half-lives in cancer and non-cancer cell lines and suggest that CMTr2 is most likely responsible for the high Aprop level in poly(A)+ RNA.
Collapse
Affiliation(s)
- Nadine A Kueck
- University of Münster, Institute of Biochemistry, Corrensstr. 36, D-48149, Muenster, Germany
| | - Sabine Hüwel
- University of Münster, Institute of Biochemistry, Corrensstr. 36, D-48149, Muenster, Germany
| | - Arne Hoffmann
- Ludwig-Maximilians-University Munich, Department of Chemistry, Butenandtstr. 5-13, Haus F, D-81377, Munich, Germany
| | - Andrea Rentmeister
- Ludwig-Maximilians-University Munich, Department of Chemistry, Butenandtstr. 5-13, Haus F, D-81377, Munich, Germany
| |
Collapse
|
23
|
Shi T, Zhang H, Chen Y. The m6A revolution: transforming tumor immunity and enhancing immunotherapy outcomes. Cell Biosci 2025; 15:27. [PMID: 39987091 PMCID: PMC11846233 DOI: 10.1186/s13578-025-01368-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotes, plays a critical role in the development and progression of various diseases, including cancer, through its regulation of RNA degradation, stabilization, splicing, and cap-independent translation. Emerging evidence underscores the significant role of m6A modifications in both pro-tumorigenic and anti-tumorigenic immune responses. In this review, we provide a comprehensive overview of m6A modifications and examine the relationship between m6A regulators and cancer immune responses. Additionally, we summarize recent advances in understanding how m6A modifications influence tumor immune responses by directly modulating immune cells (e.g., dendritic cells, tumor-associated macrophages, and T cells) and indirectly affecting cancer cells via mechanisms such as cytokine and chemokine regulation, modulation of cell surface molecules, and metabolic reprogramming. Furthermore, we explore the potential synergistic effects of targeting m6A regulators in combination with immune checkpoint inhibitor (ICI) therapies. Together, this review consolidates current knowledge on the role of m6A-mediated regulation in tumor immunity, offering insights into how a deeper understanding of these modifications may identify patients who are most likely to benefit from immunotherapies.
Collapse
Affiliation(s)
- Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| | - Huan Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
| | - Yueqiu Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
| |
Collapse
|
24
|
Chen H, Xuan A, Shi X, Fan T, Xue S, Ruan J, Wang X, Tang S, Qi W, Sun H, Liu C, He S, Ding C, Zhu Z. RNA N6-methyladenosine modification in arthritis: New insights into pathogenesis. Mod Rheumatol 2025; 35:203-214. [PMID: 39235765 DOI: 10.1093/mr/roae080] [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: 01/22/2024] [Accepted: 08/15/2024] [Indexed: 09/06/2024]
Abstract
The commonest type of eukaryotic RNA modification, N6-methyladenosine (m6A), has drawn increased scrutiny in the context of pathological functioning as well as relevance in determination of RNA stability, splicing, transportation, localization, and translation efficiency. The m6A modification plays an important role in several types of arthritis, especially osteoarthritis and rheumatoid arthritis. Recent studies have reported that m6A modification regulates arthritis pathology in cells, such as chondrocytes and synoviocytes via immune responses and inflammatory responses through functional proteins classified as writers, erasers, and readers. The aim of this review was to highlight recent advances relevant to m6A modification in the context of arthritis pathogenesis and detail underlying molecular mechanisms, regulatory functions, clinical applications, and future perspectives of m6A in arthritis with the aim of providing a foundation for future research directions.
Collapse
Affiliation(s)
- Haowei Chen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Anran Xuan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaorui Shi
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tianxiang Fan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Song Xue
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianzhao Ruan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoshuai Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Su'an Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weizhong Qi
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Canzhao Liu
- Department of Cardiovascular Medicine, Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuai He
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology, Royal North Shore Hospital and Sydney Musculoskeletal Health, Kolling Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
25
|
Wang L, Weichselbaum RR, He C. N 6-methyladenosine reader YTHDF2 in cell state transition and antitumor immunity. RNA (NEW YORK, N.Y.) 2025; 31:395-401. [PMID: 39719324 PMCID: PMC11874973 DOI: 10.1261/rna.080259.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Recent studies have revealed that the YTHDF family proteins bind preferentially to the N 6-methyladenosine (m6A)-modified mRNA and regulate the functions of these RNAs in different cell types. YTHDF2, the first identified m6A reader in mammals, has garnered significant attention because of its profound effect to regulate the m6A epitranscriptome in multiple biological processes. Here, we review current knowledge on the mechanisms by which YTHDF2 exerts its functions and discuss recent advances that underscore the multifaceted role of YTHDF2 in development, stem cell expansion, and immune evasion. We also highlight potential therapeutic interventions targeting the m6A/YTHDF2 axis to improve the response to current antitumor therapies.
Collapse
Affiliation(s)
- Liangliang Wang
- The Laboratory of Microbiome and Microecological Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois 60637, USA
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois 60637, USA
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, Illinois 60637, USA
- Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
26
|
Cai Z, Song P, Yu K, Jia G. Advanced reactivity-based sequencing methods for mRNA epitranscriptome profiling. RSC Chem Biol 2025; 6:150-169. [PMID: 39759443 PMCID: PMC11694185 DOI: 10.1039/d4cb00215f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Currently, over 170 chemical modifications identified in RNA introduce an additional regulatory attribute to gene expression, known as the epitranscriptome. The development of detection methods to pinpoint the location and quantify these dynamic and reversible modifications has significantly expanded our understanding of their roles. This review goes deep into the latest progress in enzyme- and chemical-assisted sequencing methods, highlighting the opportunities presented by these reactivity-based techniques for detailed characterization of RNA modifications. Our survey provides a deeper understanding of the function and biological roles of RNA modification.
Collapse
Affiliation(s)
- Zhihe Cai
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
| | - Peizhe Song
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
| | - Kemiao Yu
- Peking-Tsinghua Center for Life Sciences, Peking University Beijing 100871 China
| | - Guifang Jia
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
- Peking-Tsinghua Center for Life Sciences, Peking University Beijing 100871 China
- Beijing Advanced Center of RNA Biology, Peking University Beijing 100871 China
| |
Collapse
|
27
|
Li Z, Lao Y, Yan R, Li F, Guan X, Dong Z. N6-methyladenosine in inflammatory diseases: Important actors and regulatory targets. Gene 2025; 936:149125. [PMID: 39613051 DOI: 10.1016/j.gene.2024.149125] [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: 08/21/2024] [Revised: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
N6-methyladenosine (m6A) is one of the most prevalent epigenetic modifications in eukaryotic cells. It regulates RNA function and stability by modifying RNA methylation through writers, erasers, and readers. As a result, m6A plays a critical role in a wide range of biological processes. Inflammation is a common and fundamental pathological process. Numerous studies have investigated the role of m6A modifications in inflammatory diseases. This review highlights the mechanisms by which m6A contributes to inflammation, focusing on pathogen-induced infectious diseases, autoimmune disorders, allergic conditions, and metabolic disorder-related inflammatory diseases.
Collapse
Affiliation(s)
- Zewen Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Yongfeng Lao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Rui Yan
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Fuhan Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Xin Guan
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhilong Dong
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
28
|
Li P, Lin Y, Ma H, Zhang J, Zhang Q, Yan R, Fan Y. Epigenetic regulation in female reproduction: the impact of m6A on maternal-fetal health. Cell Death Discov 2025; 11:43. [PMID: 39904996 PMCID: PMC11794895 DOI: 10.1038/s41420-025-02324-z] [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: 09/02/2024] [Revised: 01/09/2025] [Accepted: 01/24/2025] [Indexed: 02/06/2025] Open
Abstract
With the development of public health, female diseases have become the focus of current concern. The unique reproductive anatomy of women leads to the development of gynecological diseases gradually become an important part of the socio-economic burden. Epigenetics plays an irreplaceable role in gynecologic diseases. As an important mRNA modification, m6A is involved in the maturation of ovum cells and maternal-fetal microenvironment. At present, researchers have found that m6A is involved in the regulation of gestational diabetes and other reproductive system diseases, but the specific mechanism is not clear. In this manuscript, we summarize the components of m6A, the biological function of m6A, the progression of m6A in the maternal-fetal microenvironment and a variety of gynecological diseases as well as the progression of targeted m6A treatment-related diseases, providing a new perspective for clinical treatment-related diseases.
Collapse
Affiliation(s)
- Peipei Li
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hongyun Ma
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Jiao Zhang
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Qiaorui Zhang
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Ruihua Yan
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Yang Fan
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China.
| |
Collapse
|
29
|
Kim HS, Eun JW, Jang SH, Kim JY, Jeong JY. The diverse landscape of RNA modifications in cancer development and progression. Genes Genomics 2025; 47:135-155. [PMID: 39643826 DOI: 10.1007/s13258-024-01601-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: 10/25/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND RNA modifications, a central aspect of epitranscriptomics, add a regulatory layer to gene expression by modifying RNA function without altering nucleotide sequences. These modifications play vital roles across RNA species, influencing RNA stability, translation, and interaction dynamics, and are regulated by specific enzymes that add, remove, and interpret these chemical marks. OBJECTIVE This review examines the role of aberrant RNA modifications in cancer progression, exploring their potential as diagnostic and prognostic biomarkers and as therapeutic targets. We focus on how altered RNA modification patterns impact oncogenes, tumor suppressor genes, and overall tumor behavior. METHODS We performed an in-depth analysis of recent studies and advances in RNA modification research, highlighting key types and functions of RNA modifications and their roles in cancer biology. Studies involving preclinical models targeting RNA-modifying enzymes were reviewed to assess therapeutic efficacy and potential clinical applications. RESULTS Aberrant RNA modifications were found to significantly influence cancer initiation, growth, and metastasis. Dysregulation of RNA-modifying enzymes led to altered gene expression profiles in oncogenes and tumor suppressors, correlating with tumor aggressiveness, patient outcomes, and response to immunotherapy. Notably, inhibitors of these enzymes demonstrated potential in preclinical models by reducing tumor growth and enhancing the efficacy of existing cancer treatments. CONCLUSIONS RNA modifications present promising avenues for cancer diagnosis, prognosis, and therapy. Understanding the mechanisms of RNA modification dysregulation is essential for developing targeted treatments that improve patient outcomes. Further research will deepen insights into these pathways and support the clinical translation of RNA modification-targeted therapies.
Collapse
Affiliation(s)
- Hyung Seok Kim
- Department of Biochemistry, Kosin University College of Medicine, Seo-Gu, Busan, 49267, South Korea
| | - Jung Woo Eun
- Department of Gastroenterology, Ajou University School of Medicine, 164 Worldcup-Ro, Yeongtong-Gu, Suwon, 16499, South Korea
| | - Se Ha Jang
- Department of Gastroenterology, Ajou University School of Medicine, 164 Worldcup-Ro, Yeongtong-Gu, Suwon, 16499, South Korea
| | - Ji Yun Kim
- Department of Biochemistry, Kosin University College of Medicine, Seo-Gu, Busan, 49267, South Korea
| | - Jee-Yeong Jeong
- Department of Biochemistry, Kosin University College of Medicine, Seo-Gu, Busan, 49267, South Korea.
| |
Collapse
|
30
|
Xu J, Koch J, Schmidt C, Nientiedt M, Neuberger M, Erben P, Michel MS, Rodríguez-Paredes M, Lyko F. Loss of YTHDC1 m 6A reading function promotes invasiveness in urothelial carcinoma of the bladder. Exp Mol Med 2025; 57:118-130. [PMID: 39741187 PMCID: PMC11799412 DOI: 10.1038/s12276-024-01377-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/18/2024] [Accepted: 10/06/2024] [Indexed: 01/02/2025] Open
Abstract
Bladder cancer poses significant clinical challenges due to its high metastatic potential and poor prognosis, especially when it progresses to muscle-invasive stages. Here, we show that the m6A reader YTHDC1 is downregulated in muscle-invasive bladder cancer and is negatively correlated with the expression of epithelial‒mesenchymal transition genes. The functional inhibition or depletion of YTHDC1 increased the migration and invasion of urothelial cells. Integrative analysis of multimodal sequencing datasets provided detailed insights into the molecular mechanisms mediating YTHDC1-dependent phenotypes and identified SMAD6 as a key transcript involved in the invasiveness of urothelial carcinoma of the bladder. Notably, SMAD6 mRNA colocalized less with YTHDC1 in tumoral tissues than in paratumoral tissues, indicating disrupted binding during cancer progression. Our findings establish YTHDC1-dependent m6A reading as a critical epitranscriptomic mechanism regulating bladder cancer invasiveness and provide a paradigm for the epitranscriptomic deregulation of cancer-associated networks.
Collapse
Affiliation(s)
- Jinyun Xu
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Jonas Koch
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Claudia Schmidt
- Core Facility Unit Light Microscopy, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Malin Nientiedt
- Department of Urology and Urosurgery, Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany
| | - Manuel Neuberger
- Department of Urology and Urosurgery, Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany
| | - Philipp Erben
- Department of Urology and Urosurgery, Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany
| | - Maurice Stephan Michel
- Department of Urology and Urosurgery, Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany
| | - Manuel Rodríguez-Paredes
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Frank Lyko
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120, Heidelberg, Germany.
| |
Collapse
|
31
|
Li X, Zheng M, Ma S, Nie F, Yin Z, Liang Y, Yan X, Wen W, Yu J, Liang Y, Huang S, Han H. YTHDC1 is a therapeutic target for B-cell acute lymphoblastic leukemia by attenuating DNA damage response through the KMT2C-H3K4me1/me3 epigenetic axis. Leukemia 2025; 39:308-322. [PMID: 39501105 DOI: 10.1038/s41375-024-02451-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 02/06/2025]
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is an aggressive malignancy characterized by the aberrant accumulation of immature and dysfunctional B cells in bone marrow (BM). Although chemotherapy and other therapies have been widely applied, some patients such as relapsed or drug-refractory (R/R) B-ALL patients exhibit limited response. YT521-B homologous domain-containing protein 1 (YTHDC1) is a nuclear reader of N6-methyladenosine (m6A) RNA modification, which has been implicated in different malignancies including leukemia. In the current study, we show that YTHDC1 is highly expressed in B-ALL cells. YTHDC1 knockdown attenuated B-ALL cell proliferation and cell cycle progression in vitro, and prolonged survival of mice in the human B-ALL xenograft model in vivo attributable to compromised leukemogenesis. Mechanistically, YTHDC1 knockdown significantly increased the accumulation of endogenous and chemotherapeutic agents-induced DNA damage in B-ALL cells. Furthermore, we identified that YTHDC1 binds to and stabilizes m6A-modified KMT2C mRNA. KMT2C is a key enzyme catalyzing histone H3K4 methylation required for the expression of DNA damage response (DDR)-related genes, implying that YTHDC1 inhibitors might improve chemotherapy by attenuating DDR via reducing KMT2C. Indeed, with molecular docking and biochemical experiments, we identified EPZ-5676 as a YTHDC1 inhibitor, and combination of EPZ-5676 with Cytarabine (Ara-c) significantly improved the efficacy of chemotherapy in B-ALL mouse models using YTHDC1high primary and lined B-ALL cells. Collectively, YTHDC1 is required for DDR in B-ALL cells by upregulating DDR-related gene expression via stabilizing m6A-modified KMT2C mRNA, thereby leading to increased histone H3K4 methylation, and targeted inhibition of YTHDC1 is a potentially new therapeutic strategy against B-ALL, especially YTHDC1high B-ALL.
Collapse
Affiliation(s)
- Xinxin Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, PR China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, Guangdong, PR China
| | - Minhua Zheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, PR China
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Fengze Nie
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, PR China
| | - Zhiqiang Yin
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Yanan Liang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, PR China
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Xianchun Yan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Weihong Wen
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, PR China
| | - Jianhua Yu
- Division of Hematology & Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA, USA
| | - Yingmin Liang
- Department of Hematology, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, PR China
| | - Siyong Huang
- Department of Hematology, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, PR China.
| | - Hua Han
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
32
|
Lee ES, Smith HW, Wang YE, Ihn SS, Scalize de Oliveira L, Kejiou NS, Liang YL, Nabeel-Shah S, Jomphe RY, Pu S, Greenblatt JF, Palazzo AF. N-6-methyladenosine (m6A) promotes the nuclear retention of mRNAs with intact 5' splice site motifs. Life Sci Alliance 2025; 8:e202403142. [PMID: 39626965 PMCID: PMC11629677 DOI: 10.26508/lsa.202403142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
In humans, misprocessed mRNAs containing intact 5' Splice Site (5'SS) motifs are nuclear retained and targeted for decay by ZFC3H1, a component of the Poly(A) Exosome Targeting complex, and U1-70K, a component of the U1 snRNP. In S. pombe, the ZFC3H1 homolog, Red1, binds to the YTH domain-containing protein Mmi1 and targets certain RNA transcripts to nuclear foci for nuclear retention and decay. Here we show that YTHDC1 and YTHDC2, two YTH domain-containing proteins that bind to N-6-methyladenosine (m6A) modified RNAs, interact with ZFC3H1 and U1-70K, and are required for the nuclear retention of mRNAs with intact 5'SS motifs. Disruption of m6A deposition inhibits both the nuclear retention of these transcripts and their accumulation in YTHDC1-enriched foci that are adjacent to nuclear speckles. Endogenous RNAs with intact 5'SS motifs, such as intronic poly-adenylated transcripts, tend to be m6A-modified at low levels. Thus, the m6A modification acts on a conserved quality control mechanism that targets misprocessed mRNAs for nuclear retention and decay.
Collapse
Affiliation(s)
- Eliza S Lee
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Harrison W Smith
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Yifan E Wang
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Sean Sj Ihn
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | | | - Nevraj S Kejiou
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Yijing L Liang
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Canada
| | - Syed Nabeel-Shah
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Robert Y Jomphe
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Shuye Pu
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Jack F Greenblatt
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | | |
Collapse
|
33
|
Li Y, Yin M, Wang J, Zhao X, Xu J, Wang W, Fu B. Epitranscriptome profiles reveal participation of the RNA methyltransferase gene OsMTA1 in rice seed germination and salt stress response. BMC PLANT BIOLOGY 2025; 25:115. [PMID: 39865266 PMCID: PMC11771074 DOI: 10.1186/s12870-025-06134-4] [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: 11/07/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND RNA m6A methylation installed by RNA methyltransferases plays a crucial role in regulating plant growth and development and environmental stress responses. However, the underlying molecular mechanisms of m6A methylation involved in seed germination and stress responses are largely unknown. In the present study, we surveyed global m6A methylation in rice seed germination under salt stress and the control (no stress) using an osmta1 mutant and its wild type. RESULTS The knockout of OsMTA1 resulted in a decreased level of m6A methylation and delayed seed germination, together with increased oxidative damage in the osmta1-1 mutant, especially under salt stress, indicating that OsMTA1 performs a crucial function in rice seed germination and salt stress response. Comparative analysis of m6A profiling using methylated RNA immunoprecipitation sequencing revealed that a unique set of genes that functioned in seed germination, cell growth, and development, including OsbZIP78 and OsA8, were hypomethylated in osmta1-1 embryos and germinating seeds. Numerous genes involved in plant growth and stress response were hypomethylated in the osmta1-1 mutant during seed germination under salt stress. Further combined analysis of the m6A methylome and transcriptome revealed that the loss of function of OsMTA1 had a more complex impact on gene expression in osmta1-1. Several hypomethylated genes with a negative role in growth and development, such as OsHsfA7 and OsHDAC3, were highly up-regulated in the osmta1-1 mutant under the control condition. In contrast, several hypomethylated genes positively associated with stress response were down-regulated, whereas a different set of hypomethylated genes that functioned as negative regulators of growth and stress response were up-regulated in the osmta1-1 mutant under salt stress. These results further demonstrated that OsMTA1-mediated m6A methylation modulated rice seed germination and salt stress response by regulating transcription of a unique set of genes with diverse functions. CONCLUSION Our results reveal a crucial role for the m6A methyltransferase gene OsMTA1 in regulating rice seed germination and salt stress response, and provide candidate genes to assist in breeding new stress-tolerant rice varieties.
Collapse
Affiliation(s)
- Yingbo Li
- State Key Laboratory of Crop Gene Resources and Breeding/Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, South Zhong-Guan-Cun Street 12#, Beijing, 100081, China
| | - Ming Yin
- State Key Laboratory of Crop Gene Resources and Breeding/Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, South Zhong-Guan-Cun Street 12#, Beijing, 100081, China
| | - Juan Wang
- State Key Laboratory of Crop Gene Resources and Breeding/Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, South Zhong-Guan-Cun Street 12#, Beijing, 100081, China
| | - Xiuqin Zhao
- State Key Laboratory of Crop Gene Resources and Breeding/Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, South Zhong-Guan-Cun Street 12#, Beijing, 100081, China
| | - Jianlong Xu
- State Key Laboratory of Crop Gene Resources and Breeding/Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, South Zhong-Guan-Cun Street 12#, Beijing, 100081, China
| | - Wensheng Wang
- State Key Laboratory of Crop Gene Resources and Breeding/Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, South Zhong-Guan-Cun Street 12#, Beijing, 100081, China.
- National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya, 572024, China.
| | - Binying Fu
- State Key Laboratory of Crop Gene Resources and Breeding/Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, South Zhong-Guan-Cun Street 12#, Beijing, 100081, China.
| |
Collapse
|
34
|
Zhang H, Lu W, Tang H, Chen A, Gao X, Zhu C, Zhang J. Novel Insight of N6-Methyladenosine in Cardiovascular System. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:222. [PMID: 40005339 PMCID: PMC11857502 DOI: 10.3390/medicina61020222] [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: 11/28/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
N6-methyladenosine (m6A) is the most common and abundant internal co-transcriptional modification in eukaryotic RNAs. This modification is catalyzed by m6A methyltransferases, known as "writers", including METTL3/14 and WTAP, and removed by demethylases, or "erasers", such as FTO and ALKBH5. It is recognized by m6A-binding proteins, or "readers", such as YTHDF1/2/3, YTHDC1/2, IGF2BP1/2/3, and HNRNPA2B1. Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Recent studies indicate that m6A RNA modification plays a critical role in both the physiological and pathological processes involved in the initiation and progression of CVDs. In this review, we will explore how m6A RNA methylation impacts both the normal and disease states of the cardiovascular system. Our focus will be on recent advancements in understanding the biological functions, molecular mechanisms, and regulatory factors of m6A RNA methylation, along with its downstream target genes in various CVDs, such as atherosclerosis, ischemic diseases, metabolic disorders, and heart failure. We propose that the m6A RNA methylation pathway holds promise as a potential therapeutic target in cardiovascular disease.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Wei Lu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Haoyue Tang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Aiqun Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Xiaofei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| | - Congfei Zhu
- Department of Cardiology, Lianshui County People’s Hospital, Affiliated Hospital of Kangda College, Nanjing Medical University, Huaian 223400, China
| | - Junjie Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; (H.Z.); (W.L.); (H.T.); (A.C.); (X.G.)
| |
Collapse
|
35
|
Yang Y, Ni WJ, Yang Y, Liao J, Yang Y, Li J, Zhu X, Guo C, Xie F, Leng XM. Research progress on N6-methyladenosine RNA modification in osteosarcoma: functions, mechanisms, and potential clinical applications. Med Oncol 2025; 42:55. [PMID: 39853585 DOI: 10.1007/s12032-024-02597-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/30/2024] [Indexed: 01/26/2025]
Abstract
Osteosarcoma (OS) is the most commonly diagnosed primary malignant bone tumor in children and adolescents. Despite significant advancements in therapeutic strategies against OS over the past few decades, the prognosis for this disease remains poor, largely due to its high invasiveness and challenges associated with its treatment. N6-methyladenosine (m6A) modification is one of the most abundant epigenetic modifications of RNAs, and many studies have highlighted its crucial role in OS. This article provides a comprehensive summary and introduction to m6A regulators, including methyltransferases, demethylases, and binding proteins. The article emphasizes how regulated m6A modifications can either promote or inhibit OS. It also delves into the mechanisms by which m6A-modified messenger RNAs (mRNAs) and noncoding RNAs (ncRNAs) participate in signaling pathways such as the Wnt/β-catenin, PI3K/AKT, and STAT3 pathways, and discusses these mechanisms in detail. Given the abnormal expression of m6A regulators in OS, the article also explores their potential applications as biomarkers or therapeutic targets in clinical settings. It is anticipated that this review will provide new insights into the diagnosis and treatment of OS.
Collapse
Affiliation(s)
- Ying Yang
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Wen-Juan Ni
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular of Ministry of Education, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Yadong Yang
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Junnan Liao
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Yuqian Yang
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Jianwei Li
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Xiuzhi Zhu
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Chun Guo
- Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, 98 Chengxiang Road, Youjiang District, Baise, 533000, Guangxi, China
- Department of Human Anatomy, School of Basic Medical Sciences, Youjiang Medical University for Nationalities, 98 Chengxiang Road, Youjiang District, Baise, 533000, Guangxi, People's Republic of China
| | - Fuhua Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Xiao-Min Leng
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China.
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular of Ministry of Education, Gannan Medical University, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
36
|
Zhang S, Wang R, Luo K, Gu S, Liu X, Wang J, Zhang L, Sun L. Dynamics and regulatory roles of RNA m 6A methylation in unbalanced genomes. eLife 2025; 13:RP100144. [PMID: 39853090 PMCID: PMC11759410 DOI: 10.7554/elife.100144] [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] [Indexed: 01/26/2025] Open
Abstract
N6-methyladenosine (m6A) in eukaryotic RNA is an epigenetic modification that is critical for RNA metabolism, gene expression regulation, and the development of organisms. Aberrant expression of m6A components appears in a variety of human diseases. RNA m6A modification in Drosophila has proven to be involved in sex determination regulated by Sxl and may affect X chromosome expression through the MSL complex. The dosage-related effects under the condition of genomic imbalance (i.e. aneuploidy) are related to various epigenetic regulatory mechanisms. Here, we investigated the roles of RNA m6A modification in unbalanced genomes using aneuploid Drosophila. The results showed that the expression of m6A components changed significantly under genomic imbalance, and affected the abundance and genome-wide distribution of m6A, which may be related to the developmental abnormalities of aneuploids. The relationships between methylation status and classical dosage effect, dosage compensation, and inverse dosage effect were also studied. In addition, we demonstrated that RNA m6A methylation may affect dosage-dependent gene regulation through dosage-sensitive modifiers, alternative splicing, the MSL complex, and other processes. More interestingly, there seems to be a close relationship between MSL complex and RNA m6A modification. It is found that ectopically overexpressed MSL complex, especially the levels of H4K16Ac through MOF, could influence the expression levels of m6A modification and genomic imbalance may be involved in this interaction. We found that m6A could affect the levels of H4K16Ac through MOF, a component of the MSL complex, and that genomic imbalance may be involved in this interaction. Altogether, our work reveals the dynamic and regulatory role of RNA m6A modification in unbalanced genomes, and may shed new light on the mechanisms of aneuploidy-related developmental abnormalities and diseases.
Collapse
Affiliation(s)
- Shuai Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Ruixue Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Kun Luo
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Shipeng Gu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Xinyu Liu
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Junhan Wang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Ludan Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal UniversityBeijingChina
| | - Lin Sun
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal UniversityBeijingChina
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal UniversityBeijingChina
| |
Collapse
|
37
|
King EM, Panfil AR. Dynamic Roles of RNA and RNA Epigenetics in HTLV-1 Biology. Viruses 2025; 17:124. [PMID: 39861913 PMCID: PMC11769288 DOI: 10.3390/v17010124] [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: 12/16/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Since the discovery of RNA in the early 1900s, scientific understanding of RNA form and function has evolved beyond protein coding. Viruses, particularly retroviruses like human T-cell leukemia virus type 1 (HTLV-1), rely heavily on RNA and RNA post-transcriptional modifications to regulate the viral lifecycle, pathogenesis, and evasion of host immune responses. With the emergence of new sequencing technologies in the last decade, our ability to dissect the intricacies of RNA has flourished. The ability to study RNA epigenetic modifications and splice variants has become more feasible with the recent development of third-generation sequencing technologies, such as Oxford nanopore sequencing. This review will highlight the dynamic roles of known RNA and post-transcriptional RNA epigenetic modifications within HTLV-1 biology, including viral hbz, long noncoding RNAs, microRNAs (miRNAs), transfer RNAs (tRNAs), R-loops, N6-methyladenosine (m6A) modifications, and RNA-based therapeutics and vaccines.
Collapse
Affiliation(s)
- Emily M. King
- Center for Retrovirus Research, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Amanda R. Panfil
- Center for Retrovirus Research, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
- Center for RNA Biology, Comprehensive Cancer Center, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
38
|
Cheng Y, Shang Y, Zhang S, Fan S. The interplay between RNA m6A modification and radiation biology of cancerous and non-cancerous tissues: a narrative review. Cancer Biol Med 2025; 21:j.issn.2095-3941.2024.0415. [PMID: 39831771 PMCID: PMC11745087 DOI: 10.20892/j.issn.2095-3941.2024.0415] [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: 09/24/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
The diverse radiation types in medical treatments and the natural environment elicit complex biological effects on both cancerous and non-cancerous tissues. Radiation therapy (RT) induces oncological responses, from molecular to phenotypic alterations, while simultaneously exerting toxic effects on healthy tissue. N6-methyladenosine (m6A), a prevalent modification on coding and non-coding RNAs, is a key epigenetic mark established by a set of evolutionarily conserved enzymes. The interplay between m6A modification and radiobiology of cancerous and non-cancerous tissues merits in-depth investigation. This review summarizes the roles of m6A in the biological effects induced by ionizing radiation and ultraviolet (UV) radiation. It begins with an overview of m6A modification and its detection methods, followed by a detailed examination of how m6A dynamically regulates the sensitivity of cancerous tissues to RT, the injury response in non-cancerous tissues, and the toxicological effects of UV exposure. Notably, this review underscores the importance of novel regulatory mechanisms of m6A and their potential clinical applications in identifying epigenetically modulated radiation-associated biomarkers for cancer therapy and estimation of radiation dosages. In conclusion, enzyme-mediated m6A-modification triggers alterations in target gene expression by affecting the metabolism of the modified RNAs, thus modulating progression and radiosensitivity in cancerous tissues, as well as radiation effects on normal tissues. Several promising avenues for future research are further discussed. This review highlights the importance of m6A modification in the context of radiation biology. Targeting epi-transcriptomic molecules might potentially provide a novel strategy for enhancing the radiosensitivity of cancerous tissues and mitigating radiation-induced injury to normal tissues.
Collapse
Affiliation(s)
- Yajia Cheng
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yue Shang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Shuqin Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Saijun Fan
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
39
|
Li BX, Wu MY, Wang ZH, Zhou DM, Li JQ, Lu BF, Lin XL, Zhao Y, Sheng XJ. Mechanism of hsa_circ_0069443 promoting early pregnancy loss through ALKBH5/FN1 axis in trophoblast cells. iScience 2025; 28:111608. [PMID: 39868042 PMCID: PMC11758834 DOI: 10.1016/j.isci.2024.111608] [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: 12/26/2023] [Revised: 06/30/2024] [Accepted: 12/12/2024] [Indexed: 01/28/2025] Open
Abstract
Studies have shown that circRNAs play an important regulatory role in trophoblast function and embryonic development. Based on sequencing and functional experiments, we found that hsa_circ_0069443 can regulate the function of trophoblast cells, and its presence is found in the exosomes secreted by trophoblast cells. It is known that exosomes mediate the interaction between the uterus and embryo, which is crucial for successful pregnancy. We found that trophoblast cell-derived exosomes overexpressing hsa_circ_0069443 promoted the migration and invasion of endometrial stromal cells as well as the EMT process of endometrial glandular epithelial cells, and this process promotes embryo implantation and adhesion, thus proving that a decrease in hsa_circ_0069443 may be the key factor leading to early pregnancy loss. This study also found that hsa_circ_0069443 can bind to the RNA-binding protein demethylase ALKBH5, affecting the overall m6A level of trophoblast cells, and hsa_circ_0069443 and ALKBH5 can regulate the expression level of FN1, verifying the role of the 0069443/ALKBH5/FN1 axis in trophoblast cells and endometrial stromal cells. In summary, this study demonstrates that hsa_circ_0069443 may be a key factor leading to early pregnancy loss, and the regulation of the hsa_circ_0069443/ALKBH5/FN1 axis may provide new insights into early diagnostic markers for early pregnancy loss.
Collapse
Affiliation(s)
- Bai-xue Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Mei-yao Wu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Department of Gynecology, Queen Mary Hospital, Hong Kong, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Zhi-hui Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Dong-mei Zhou
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Jian-qi Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Bing-feng Lu
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Xiao-ling Lin
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Yang Zhao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Xiu-jie Sheng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Department of Gynecologic Oncology Research Office, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangzhou Key Laboratory of Targeted Therapy for Gynecologic Oncology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
- Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| |
Collapse
|
40
|
Xie X, Fang Z, Zhang H, Wang Z, Li J, Jia Y, Shang L, Cao F, Li F. The role of N(6)-methyladenosine (m6a) modification in cancer: recent advances and future directions. EXCLI JOURNAL 2025; 24:113-150. [PMID: 39967906 PMCID: PMC11830918 DOI: 10.17179/excli2024-7935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/18/2024] [Indexed: 02/20/2025]
Abstract
N(6)-methyladenosine (m6A) modification is the most abundant and prevalent internal modification in eukaryotic mRNAs. The role of m6A modification in cancer has become a hot research topic in recent years and has been widely explored. m6A modifications have been shown to regulate cancer occurrence and progression by modulating different target molecules. This paper reviews the recent research progress of m6A modifications in cancer and provides an outlook on future research directions, especially the development of molecularly targeted drugs. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Xiaozhou Xie
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhen Fang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haoyu Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuchen Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
41
|
Chen L, Li X, Liu H, He F, Li M, Long R, Wang X, Kang J, Yang Q. Comprehensive analysis of epigenetic modifications in alfalfa under cadmium stress. JOURNAL OF HAZARDOUS MATERIALS 2025; 482:136545. [PMID: 39577281 DOI: 10.1016/j.jhazmat.2024.136545] [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: 06/04/2024] [Revised: 09/30/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Epigenetics plays an important role in plant growth and development and in environmental adaptation. Alfalfa, an important forage crop, is rich in nutrients. However, little is known about the molecular regulatory mechanisms underlying the response of alfalfa to cadmium (Cd) stress. Here, we performed DNA methylation (5mC), RNA methylation (m6A) and transcriptomic sequencing analyses of alfalfa roots under Cd stress. Whole-genome methylation sequencing and transcriptomic sequencing revealed that Cd stress reduced DNA methylation levels. Moreover, a reduced 5mC methylation level was associated with decreased expression of several DNA methyltransferase genes. Compared with those under normal (CK) conditions, the m6A modification levels under Cd stress were greater and were positively correlated with gene expression in alfalfa roots. We also found a negative correlation between the 5mC level and the m6A level, especially in CG and CHG contexts. In yeast, the overexpression of MsNARMP5 (natural resistance-associated macrophage protein) and MsPCR2 (plant cadmium resistance 2), which are modified by 5mC or m6A, significantly increased Cd stress tolerance. These results provide candidate genes for future studies on the mechanism of Cd stress tolerance in alfalfa roots and valuable information for studying heavy metal stress in alfalfa breeding.
Collapse
Affiliation(s)
- Lin Chen
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Xianyang Li
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Hao Liu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Fei He
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Mingna Li
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ruicai Long
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xue Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junmei Kang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qingchuan Yang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
42
|
Su N, Yu X, Duan M, Shi N. Recent advances in methylation modifications of microRNA. Genes Dis 2025; 12:101201. [PMID: 39524539 PMCID: PMC11550756 DOI: 10.1016/j.gendis.2023.101201] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 11/16/2024] Open
Abstract
microRNAs (miRNAs) are short single-stranded non-coding RNAs between 21 and 25 nt in length in eukaryotic organisms, which control post-transcriptional gene expression. Through complementary base pairing, miRNAs generally bind to their target messenger RNAs and repress protein production by destabilizing the messenger RNA and translational silencing. They regulate almost all life activities, such as cell proliferation, differentiation, apoptosis, tumorigenesis, and host-pathogen interactions. Methylation modification is the most common RNA modification in eukaryotes. miRNA methylation exists in different types, mainly N6-methyladenosine, 5-methylcytosine, and 7-methylguanine, which can change the expression level and biological mode of action of miRNAs and improve the activity of regulating gene expression in a very fine-tuned way with flexibility. In this review, we will summarize the recent findings concerning methylation modifications of miRNA, focusing on their biogenesis and the potential role of miRNA fate and functions.
Collapse
Affiliation(s)
| | | | | | - Ning Shi
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| |
Collapse
|
43
|
Rupareliya M, Shende P. Influence of RNA Methylation on Cancerous Cells: A Prospective Approach for Alteration of In Vivo Cellular Composition. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1474:79-103. [PMID: 39259424 DOI: 10.1007/5584_2024_820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
RNA methylation is a dynamic and ubiquitous post-transcriptional modification that plays a pivotal role in regulating gene expression in various conditions like cancer, neurological disorders, cardiovascular diseases, viral infections, metabolic disorders, and autoimmune diseases. RNA methylation manifests across diverse RNA species including messenger RNA (mRNA), ribosomal RNA (rRNA), and transfer RNA (tRNA), exerting pivotal roles in gene expression regulation and various biological phenomena. Aberrant activity of writer, eraser, and reader proteins enables dysregulated methylation landscape across diverse malignancy transcriptomes, frequently promoting cancer pathogenesis. Numerous oncogenic drivers, tumour suppressors, invasion/metastasis factors, and signalling cascade components undergo methylation changes that modulate respective mRNA stability, translation, splicing, transport, and protein-RNA interactions accordingly. Functional studies confirm methylation-dependent alterations drive proliferation, survival, motility, angiogenesis, stemness, metabolism, and therapeutic evasion programs systemically. Methyltransferase overexpression typifies certain breast, liver, gastric, and other carcinomas correlating with adverse clinical outcomes like diminished overall survival. Mapping efforts uncover nodal transcripts for targeted drug development against hyperactivated regulators including METTL3. Some erasers and readers also suitable lead candidates based on apparent synthetic lethality. Proteomic screens additionally highlight relevant methylation-sensitive effector pathways amenable to combinatorial blockade, reversing compensatory signalling mechanisms that facilitate solid tumour progression. Quantifying global methylation burdens and responsible enzymes clinically predicts patient prognosis, risk stratification for adjuvant therapy, and overall therapeutic responsiveness.
Collapse
Affiliation(s)
- Manali Rupareliya
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, India.
| |
Collapse
|
44
|
Tian S, Song Y, Guo L, Zhao H, Bai M, Miao M. Epigenetic Mechanisms in Osteoporosis: Exploring the Power of m 6A RNA Modification. J Cell Mol Med 2025; 29:e70344. [PMID: 39779466 PMCID: PMC11710941 DOI: 10.1111/jcmm.70344] [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: 09/05/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Osteoporosis, recognised as a metabolic disorder, has emerged as a significant burden on global health. Although available treatments have made considerable advancements, they remain inadequately addressed. In recent years, the role of epigenetic mechanisms in skeletal disorders has garnered substantial attention, particularly concerning m6A RNA modification. m6A is the most prevalent dynamic and reversible modification in eukaryotes, mediating various metabolic processes of mRNAs, including splicing, structural conversion, translation, translocation and degradation and serves as a crucial component of epigenetic modification. Research has increasingly validated that m6A plays a vital role in the proliferation, differentiation, migration, invasion,and repair of bone marrow mesenchymal stem cells (BMSCs), osteoblasts and osteoclasts, all of which impact the whole process of osteoporosis pathogenesis. Continuous efforts have been made to target m6A regulators and natural products derived from traditional medicine, which exhibit multiple biological activities such as anti-inflammatory and anticancer effects, have emerged as a valuable resources for m6A drug discovery. This paper elaborates on m6A methylation and its regulatory role in osteoporosis, emphasising its implications for diagnosis and treatment, thereby providing theoretical references.
Collapse
Affiliation(s)
- Shuo Tian
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| | - Yagang Song
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| | - Lin Guo
- School of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Hui Zhao
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| | - Ming Bai
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| | - Mingsan Miao
- Academy of Traditional Chinese MedicineHenan University of Chinese MedicineZhengzhouChina
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu‐YaoZhengzhouChina
| |
Collapse
|
45
|
Li K, Liang Y, Li X, Yang M, Wang M, Li F, Qi X, Zhou J, Fu W, Li L. Rapid and direct detection of m 6A methylation by DNAzyme-based and smartphone-assisted electrochemical biosensor. Biosens Bioelectron 2025; 267:116788. [PMID: 39316869 DOI: 10.1016/j.bios.2024.116788] [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: 07/14/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
m6A methylation detection is crucial for understanding RNA functions, revealing disease mechanisms, guiding drug development and advancing epigenetics research. Nevertheless, high-throughput sequencing and liquid chromatography-based traditional methods still face challenges to rapid and direct detection of m6A methylation. Here we report a DNAzyme-based and smartphone-assisted electrochemical biosensor for rapid detection of m6A. We initially identified m6A methylation-sensitive DNAzyme mutants through site mutation screening. These mutants were then combined with tetrahedral DNA to modify the electrodes, creating a 3D sensing interface. The detection of m6A was accomplished by using DNAzyme to capture and cleave the m6A sequence. The electrochemical biosensor detected the m6A sequence at nanomolar concentrations with a low detection limit of 0.69 nM and a wide detection range from 10 to 104 nM within 60 min. As a proof of concept, the 3'-UTR sequence of rice was selected as the m6A analyte. Combined with a smartphone, our biosensor shows good specificity, sensitivity, and easy-to-perform features, which indicates great prospects in the field of RNA modification detection and epigenetic analysis.
Collapse
Affiliation(s)
- Kai Li
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yu Liang
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xinran Li
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Mengrui Yang
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Min Wang
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Fukai Li
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xin Qi
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jian Zhou
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Wei Fu
- Development Center of Science and Technology, Ministry of Agriculture and Rural Affairs, Beijing 100176, China.
| | - Liang Li
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
46
|
Chen J, Wu H, Zuo T, Wu J, Chen Z. METTL3‑mediated N6‑methyladenosine modification of MMP9 mRNA promotes colorectal cancer proliferation and migration. Oncol Rep 2025; 53:9. [PMID: 39540393 DOI: 10.3892/or.2024.8842] [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: 07/11/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
N6‑methyladenosine (m6A) is the predominant chemical modification of eukaryotic mRNA, dynamically mediated by the RNA methyltransferase, methyltransferase-like 3 (METTL3). m6A modification plays a critical role in cancer progression through post‑transcriptional regulation in various types of cancer. However, the role of METTL3 and its associated m6A modification in colorectal tumorigenesis remains to be fully elucidated. In the present study, it was demonstrated that METTL3 expression and the m6A levels were both upregulated in colorectal cancer (CRC) and positively associated with clinical progression, based on the bioinformatics analysis of cancer databases. Furthermore, knockdown and overexpression of METTL3 notably affected CRC cell viability, apoptosis and migration in vitro. Similarly, xenograft animal models confirmed that METTL3 promoted CRC tumorigenicity in vivo. Mechanistically, it was revealed that the m6A modification of matrix metallopeptidase 9 (MMP9) mRNA mediated by METTL3 promoted its expression in CRC by decreasing its degradation. Collectively, the findings of the present study suggested that the METTL3/MMP9 axis could serve as a novel promising therapeutic candidate for CRC.
Collapse
Affiliation(s)
- Jie Chen
- Department of Central Laboratory, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Henglan Wu
- Department of Nephrology, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Ting Zuo
- Department of Anesthesia Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Jianming Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Zhiheng Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
47
|
Yu J, Sun W, Zhao X, Chen Y. The therapeutic potential of RNA m(6)A in lung cancer. Cell Commun Signal 2024; 22:617. [PMID: 39736743 DOI: 10.1186/s12964-024-01980-5] [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: 07/05/2024] [Accepted: 12/04/2024] [Indexed: 01/01/2025] Open
Abstract
Lung cancer (LC) is a highly malignant and metastatic form of cancer. The global incidence of and mortality from LC is steadily increasing; the mean 5-year overall survival (OS) rate for LC is less than 20%. This frustrating situation may be attributed to the fact that the pathogenesis of LC remains poorly understood and there is still no cure for mid to advanced LC. Methylation at the N6-position of adenosine (N6mA) of RNA (m(6)A) is widely present in human tissues and organs, and has been found to be necessary for cell development and maintenance of homeostasis. However, numerous basic and clinical studies have demonstrated that RNA m(6)A is deregulated in many human malignancies including LC. This can drive LC malignant characteristics such as proliferation, stemness, invasion, epithelial-mesenchymal transition (EMT), metastasis, and therapeutic resistance. Intriguingly, an increasing number of studies have also shown that eliminating RNA m(6)A dysfunction can exert significant anti-cancer effects on LC such as suppression of cell proliferation and viability, induction of cell death, and reversal of treatment insensitivity. The current review comprehensively discusses the therapeutic potential of RNA m(6)A and its underlying molecular mechanisms in LC, providing useful information for the development of novel LC treatment strategies.
Collapse
Affiliation(s)
- Jingran Yu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Shenyang , Liaoning, 110022, China
| | - Wei Sun
- Department of Radiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, China
| | - Xiangxuan Zhao
- Center for Innovative Engineering Technology in Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, No.79 Chongshandong Road, Shenyang, 110847, China.
- Health Sciences Institute, China Medical University, Puhe Road, Shenyang North New Area, Shenyang, 110022, China.
| | - Yingying Chen
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Shenyang , Liaoning, 110022, China.
| |
Collapse
|
48
|
Wang YY, Ye LH, Zhao AQ, Gao WR, Dai N, Yin Y, Zhang X. M6A modification regulates tumor suppressor DIRAS1 expression in cervical cancer cells. Cancer Biol Ther 2024; 25:2306674. [PMID: 38372700 PMCID: PMC10878024 DOI: 10.1080/15384047.2024.2306674] [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: 10/22/2023] [Accepted: 01/14/2024] [Indexed: 02/20/2024] Open
Abstract
DIRAS family GTPase 1 (DIRAS1) has been reported as a potential tumor suppressor in other human cancer. However, its expression pattern and role in cervical cancer remain unknown. Knockdown of DIRAS1 significantly promoted the proliferation, growth, migration, and invasion of C33A and SiHa cells cultured in vitro. Overexpression of DIRAS1 significantly inhibited the viability and motility of C33A and SiHa cells. Compared with normal cervical tissues, DIRAS1 mRNA levels were significantly lower in cervical cancer tissues. DIRAS1 protein expression was also significantly reduced in cervical cancer tissues compared with para-cancerous tissues. In addition, DIRAS1 expression level in tumor tissues was significantly negatively correlated with the pathological grades of cervical cancer patients. DNA methylation inhibitor (5-Azacytidine) and histone deacetylation inhibitor (SAHA) resulted in a significant increase in DIRAS1 mRNA levels in C33A and SiHa cells, but did not affect DIRAS1 protein levels. FTO inhibitor (FB23-2) significantly down-regulated intracellular DIRAS1 mRNA levels, but significantly up-regulated DIRAS1 protein levels. Moreover, the down-regulation of METTL3 and METTL14 expression significantly inhibited DIRAS1 protein expression, whereas the down-regulation of FTO and ALKBH5 expression significantly increased DIRAS1 protein expression. In conclusion, DIRAS1 exerts a significant anti-oncogenic function and its expression is significantly downregulated in cervical cancer cells. The m6A modification may be a key mechanism to regulate DIRAS1 mRNA stability and protein translation efficiency in cervical cancer.
Collapse
Affiliation(s)
- Yu-Yan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Lian-Hua Ye
- Department of Internal Medicine, Zigong Fourth People’s Hospital, Zigong, Sichuan, China
| | - An-Qi Zhao
- Department of Obstetrics and Gynecology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei-Ran Gao
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ning Dai
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yu Yin
- Operating Rooms, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xin Zhang
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
49
|
Zhao Y, Li J, Dian M, Bie Y, Peng Z, Zhou Y, Zhou B, Hao W, Wang X. Role of N6-methyladenosine methylation in nasopharyngeal carcinoma: current insights and future prospective. Cell Death Discov 2024; 10:490. [PMID: 39695216 DOI: 10.1038/s41420-024-02266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a distinct type of head and neck squamous cell carcinoma prevalent in Southern China, Southeast Asia, and North Africa. Despite advances in treatment options, the prognosis for advanced NPC remains poor, underscoring the urgent need to explore its underlying mechanisms and develop novel therapeutic strategies. Epigenetic alterations have been shown to play a key role in NPC progression. Recent studies indicate that dysregulation of RNA modifications in NPC specifically affects tumor-related transcripts, influencing various oncogenic processes. This review provides a comprehensive overview of altered RNA modifications and their regulators in NPC, with a focus on m6A and its regulatory mechanisms. We discuss how m6A RNA modification influences gene expression and affects NPC initiation and progression at the molecular level, analyzing its impact on cancer-related biological functions. Understanding these modifications could reveal new biomarkers and therapeutic targets for NPC, offering promising directions for future research and precision medicine.
Collapse
Affiliation(s)
- YaYan Zhao
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie Li
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - MeiJuan Dian
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - YaNan Bie
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - ZhiTao Peng
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - BingQian Zhou
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - WeiChao Hao
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - XiCheng Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
50
|
Wang W, Jin Y, Xie Z, He M, Li J, Wang Z, Ma S, Zhang W, Tong J. When animal viruses meet N 6-methyladenosine (m 6A) modifications: for better or worse? Vet Res 2024; 55:171. [PMID: 39695760 DOI: 10.1186/s13567-024-01424-5] [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: 06/01/2024] [Accepted: 09/07/2024] [Indexed: 12/20/2024] Open
Abstract
N6-methyladenosine (m6A) is a prevalent and dynamic RNA modification, critical in regulating gene expression. Recent research has shed light on its significance in the life cycle of viruses, especially animal viruses. Depending on the context, these modifications can either enhance or inhibit the replication of viruses. However, research on m6A modifications in animal virus genomes and the impact of viral infection on the host cell m6A landscape has been hindered due to the difficulty of detecting m6A sites at a single-nucleotide level. This article summarises the methods for detecting m6A in RNA. It then discusses the progress of research into m6A modification within animal viruses' infections, such as influenza A virus, porcine epidemic diarrhoea virus, porcine reproductive, and respiratory syndrome virus. Finally, the review explores how m6A modification affects the following three aspects of the replication of animal RNA viruses: the regulation of viral genomic RNA function, the alteration of the m6A landscape in cells after viral infection, and the modulation of antiviral immunity through m6A modification. Research on m6A modifications in viral RNA sheds light on virus-host interactions at a molecular level. Understanding the impact of m6A on viral replication can help identify new targets for antiviral drug development and may uncover novel regulatory pathways that could potentially enhance antiviral immune responses.
Collapse
Affiliation(s)
- Wenjing Wang
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Yufei Jin
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Ziyun Xie
- College of Veterinary Medicine, Hebei Agriculture University, Baoding, 071001, China
| | - Mei He
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Jing Li
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Zihan Wang
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Saiya Ma
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Wuchao Zhang
- College of Veterinary Medicine, Hebei Agriculture University, Baoding, 071001, China.
| | - Jie Tong
- College of Life Sciences, Hebei University, Baoding, 071002, China.
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China.
| |
Collapse
|