1
|
Fang F, Ni K. Expression pattern of RNA demethylase ALKBH5 in fetal and adult human testis. Tissue Cell 2025; 95:102901. [PMID: 40222158 DOI: 10.1016/j.tice.2025.102901] [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: 10/07/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025]
Abstract
N6-methyladenosine (m6A) is a common post-transcriptional modification of RNAs in eukaryotic cells, which is involved in various biological processes. ALKBH5 is one of the m6A demethylases and has been reported to play important roles in mouse testis. But the function of ALKBH5 in human testis remained undiscovered. Here we aimed to analyze the expression and location of ALKBH5 in fetal and adult human testis. We found that fetal human testis is characterized by the formation of testis cords filled with pre-spermatogonia and pre-Sertoli cells, which is significantly distinct from the convoluted seminiferous epithelium in adult testis. ALKBH5 is not only widely expressed in adult human testis, but also expressed in VASA positive pre-spermatogonia, SOX9 positive pre-Sertoli cells, and CYP11A positive pre-Leydig cells in fetal human testis. Moreover, bioinformatics analysis of published RNA-sequencing data (GSE63392) revealed the expression of ALKBH5 in human fetal germ cells is upregulated with the increase of gestational weeks. Thus, our results indicate the potential role of ALKBH5 in fetal human testis development and function.
Collapse
Affiliation(s)
- Fang Fang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke Ni
- Department of Surgery, Wuhan Red Cross Hospital, Wuhan 430015, China.
| |
Collapse
|
2
|
Xu C, Xu X, Huang Y, Shang S, Ma L. RNA methylation: A new promising biomaker in cancer liquid biopsy. Biochim Biophys Acta Rev Cancer 2025; 1880:189337. [PMID: 40315965 DOI: 10.1016/j.bbcan.2025.189337] [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: 01/24/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
RNA methylation is a vital epigenetic modification that regulates gene expression by influencing RNA processes such as transcription, degradation, translation, and transport. Aberrant methylation, including modifications like m6A, m5C, m1A, m7G, and m3C, is closely linked to tumorigenesis and progression. Liquid biopsy, a non-invasive technique analyzing tumor markers in body fluids, offers significant potential for early diagnosis and dynamic monitoring. In this context, RNA methylation, due to its tumor-specific properties, is emerging as a valuable marker. However, significant challenges remain in its clinical application. This review explores the roles of RNA methylation in cancer, recent advances in detection technologies, and its potential as a liquid biopsy marker in tumor management. It highlights its promising applications in cancer diagnosis, prognosis, and personalized treatment in the era of precision oncology.
Collapse
Affiliation(s)
- Chenxin Xu
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xin Xu
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yiwen Huang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Shuang Shang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lifang Ma
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
3
|
Jin ZY, Ling ZQ. PAQR4: From spatial regulation of cell signaling to physiological homeostasis and diseases. Biochim Biophys Acta Rev Cancer 2025; 1880:189314. [PMID: 40194713 DOI: 10.1016/j.bbcan.2025.189314] [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: 11/10/2024] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/09/2025]
Abstract
Progestin and adipoQ receptor family member 4 (PAQR4) gene is a recently discovered seven-transmembrane protein-coding gene that belongs to the PAQR family. An increasing amount of evidence suggests that PAQR4 is upregulated in multiple tumors and participates in tumor progression and chemotherapy resistance via different signaling pathways; PAQR4 regulates cellular ceramide homeostasis by influencing sphingolipid metabolism and glycerol metabolism, and plays a significant role in adipose tissue remodeling. Meanwhile, it is known that the differential expression of PAQR4 is associated with the occurrence of various diseases and is a potential biomarker and therapeutic target. This article conducts a systematic review of the subcellular localization of PAQR4, its topological structure characteristics, and its functions in cancer occurrence, metabolic diseases, and fertility, and provides clues for the future research and translational application of PAQR4.
Collapse
Affiliation(s)
- Zi-Yan Jin
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China; Postgraduate Training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
| | - Zhi-Qiang Ling
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
4
|
Jia C, Lang QF, Yin ZJ, Sun J, Meng QH, Pei TM. Role, mechanism, and application of N6-methyladenosine in hepatobiliary carcinoma. World J Gastrointest Oncol 2025; 17:105140. [DOI: 10.4251/wjgo.v17.i6.105140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/01/2025] [Accepted: 04/22/2025] [Indexed: 06/13/2025] Open
Abstract
Hepatobiliary carcinoma is a frequently occurring and highly invasive cancer within the digestive tract, known for its rapid progression. Due to its difficult diagnosis and treatment in clinical practice, hepatobiliary carcinoma is a serious threat to human life and health. In recent years, the incidence of hepatobiliary carcinoma has gradually increased. N6-methyladenosine (m6A) modification, as a reversible post-transcriptional modification of the adenosine N6 site, is one of the most important RNA modifications in eukaryotes. Emerging research indicates that m6A affects the biological process of cells through the regulation of gene expression. m6A modification also plays a key role in the occurrence and development of various cancers. This review summarizes the role and mechanism of m6A modification in hepatobiliary carcinoma, and discussed its potential clinical application, so as to provide a theoretical reference for the individualized treatment of hepatobiliary carcinoma.
Collapse
Affiliation(s)
- Chen Jia
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Qing-Fu Lang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Zhi-Jie Yin
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Jia Sun
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Qing-Hui Meng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Tie-Min Pei
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
5
|
Zhang S, Huang S, Shao Y, Li C. N6-methyladenosine-dependent upregulation of lnc-28509 increases apoptosis and suppresses bacterial infection in sea cucumber. FISH & SHELLFISH IMMUNOLOGY 2025; 165:110495. [PMID: 40513785 DOI: 10.1016/j.fsi.2025.110495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Revised: 06/05/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
In recent years, lncRNAs (long non-coding RNAs) as well as m6A (N6-methyladenosine) modification have been found to play important roles during pathogens infection induced innate immune response. However, the relationship between the lncRNAs and m6A modification remains unclear. Here, we show that the lncRNA (lnc-28509) from sea cucumber Apostichopus japonicus was significantly induced in response to Vibrio splendidus infection. Knockdown of lnc-28509 inhibits the apoptosis of coelomocytes in sea cucumber and the elimination of Vibrio splendidus. In addition, m6A is highly enriched on lnc-28509 transcripts modified by methyltransferase like 3 (METTL3) upon Vibrio splendidus infection and the METTL3 mediated m6A modification increased lnc-28509 expression through suppressing the decay of lnc-28509. Meanwhile, dual-luciferase reporter assay results revealed that lnc-28509 functions as a miRNA sponge of miR-2012, and miR-2012 inhibits the coelomocytes apoptosis via targeting apoptosis-inducing factor 1 (AIF-1) directly. The results suggested that lnc-28509 regulates apoptosis through sponging miR-2012 and promoting the accumulation of AIF-1. The unique m6A-dependent lncRNA-miRNA interaction can maintain the anti-bacterial role of lnc-28509. Collectively, these data highlight the critical role of m6A modification in the invertebrate lncRNA and reveal a previously unknown mechanism through which lncRNA-dependent cell apoptotic controlled bacterial infection.
Collapse
Affiliation(s)
- Siyuan Zhang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Sheng Huang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Yina Shao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Chenghua Li
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong, 266237, PR China.
| |
Collapse
|
6
|
Zhao Y, Chen X, Zhang X, Liu H. RNA epigenetic modifications as dynamic biomarkers in cancer: from mechanisms to clinical translation. Biomark Res 2025; 13:81. [PMID: 40483535 PMCID: PMC12145623 DOI: 10.1186/s40364-025-00794-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
RNA modifications are crucial for post-transcriptional gene regulation. Research on RNA modifications has become a novel frontier of epitranscriptomics. Up to now, over 170 kinds of modifications have been identified on mRNA and diverse non-coding RNA. Three classes of proteins (writers, erasers, and readers) regulate the addition, removal, and identification of epigenetic marks, thus affecting RNA biological functions. Increasing evidence identifies the dysregulation of RNA modifications in different cancer types and the therapeutic potential of targeting RNA-modifying enzymes. The ability of RNA modifications to improve mRNA stability and translation efficacy and decrease immunogenicity has been exploited for the clinical use of mRNA cancer vaccines. This review aims to shed light on several vital cap, tail, and internal modifications of RNA with a focus on the connection between RNA epigenetic pathways and cancer pathogenesis. We further explore the clinical potential of RNA modifications as dynamic biomarkers for cancer diagnosis, prognosis, and therapeutic response prediction, addressing both technological challenges and translational opportunities. Finally, we analyze the limitations of current studies and discuss the research focus in the future.
Collapse
Affiliation(s)
- Yingchao Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
| | - Xingli Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China.
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China.
| |
Collapse
|
7
|
Zhao X, Sun Y, Zou J, Wu Y, Huang M, Kong H, Liu G, Gerhardt H, Gu W, Zhang Y, Shang M, Wang X. Protein kinase A regulates ferroptosis by controlling GPX4 m 6A modification through phosphorylation of ALKBH5. Cell Death Differ 2025; 32:1058-1070. [PMID: 39901038 PMCID: PMC12163066 DOI: 10.1038/s41418-025-01453-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/02/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
GPX4-dependent ferroptosis has emerged as a therapeutic strategy for cancer treatment. Here, we demonstrated that protein kinase A (PKA) participates in the regulation of ferroptosis by controlling the m6A modification of GPX4 in an ALKBH5-dependent manner. Notably, we identified ALKBH5, an m6A demethylase, as a novel target of PKA, which drives phosphorylation-dependent degradation of ALKBH5 protein. Moreover, the deletion of ALKBH5 represses ferroptotic cell death by maintaining GPX4 m6A modification and stability. Thus, by regulating ALKBH5-dependent GPX4 stability, PKA acts as a key regulator of ferroptosis. Our study unveils the involvement of PKA in m6A modification, which could control GPX4-dependent ferroptosis and tumor progression.
Collapse
Affiliation(s)
- Xiaocheng Zhao
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| | - Yanxi Sun
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Laboratory of Cell Fate and Metabolic Regulation, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Juan Zou
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Laboratory of Cell Fate and Metabolic Regulation, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yanxia Wu
- Laboratory of Cell Fate and Metabolic Regulation, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Minyi Huang
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Laboratory of Cell Fate and Metabolic Regulation, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Huimin Kong
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Guangda Liu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium. Center for Human Genetics, School of Medicine, University of Leuven, Leuven, Belgium
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 St. Nicholas Avenue, New York, NY, 10032, USA
| | - Yunjiao Zhang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Min Shang
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Xingwu Wang
- Laboratory of Cell Fate and Metabolic Regulation, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| |
Collapse
|
8
|
Lu Z, Lyu Z, Dong P, Liu Y, Huang L. N6-methyladenosine RNA modification in stomach carcinoma: Novel insights into mechanisms and implications for diagnosis and treatment. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167793. [PMID: 40088577 DOI: 10.1016/j.bbadis.2025.167793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
N6-methyladenosine (m6A) RNA methylation is crucially involved in the genesis and advancement of gastric cancer (GC) by controlling various pathobiological aspects including gene expression, signal transduction, metabolism, cell death, epithelial-mesenchymal transition, angiogenesis, and exosome function. Despite its importance, the exact mechanisms by which m6A modification influences GC biology remain inadequately explored. This review consolidates the latest advances in uncovering the mechanisms and diverse roles of m6A in GC and proposes new research and translational directions. Key regulators (writers, readers, and erasers) of m6A, such as METTL3/14/16 and WTAP, significantly affect cancer progression, anticancer immune response, and treatment outcomes. m6A modification also impacts immune cell infiltration and the tumor microenvironment, highlighting its potential as a diagnostic and prognostic marker. Interactions between m6A methylation and non-coding RNAs offer further novel insights into GC development and therapeutic targets. Targeting m6A regulators could enhance immunotherapy response, overcome treatment resistance, and improve oncological and clinical outcomes. Models based on m6A can precisely predict treatment response and prognosis in GC. Additional investigation is needed to fully understand the mechanisms of m6A methylation and its potential clinical applications and relevance (e.g., as precise markers for early detection, prediction of outcome, and response to therapy and as therapeutic targets) in GC. Future research should focus on in vivo studies, potential clinical trials, and the examination of m6A modification in other types of cancers.
Collapse
Affiliation(s)
- Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Zhaojie Lyu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Yunmei Liu
- School of Cultural Heritage and Information Management, Shanghai University, Shanghai, China.
| | - Lei Huang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Shanghai Institute of Pancreatic Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunity and Inflammation, Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
9
|
Xue Y, Cao X, Chen X, Deng X, Deng XW, Ding Y, Dong A, Duan CG, Fang X, Gong L, Gong Z, Gu X, He C, He H, He S, He XJ, He Y, He Y, Jia G, Jiang D, Jiang J, Lai J, Lang Z, Li C, Li Q, Li X, Liu B, Liu B, Luo X, Qi Y, Qian W, Ren G, Song Q, Song X, Tian Z, Wang JW, Wang Y, Wu L, Wu Z, Xia R, Xiao J, Xu L, Xu ZY, Yan W, Yang H, Zhai J, Zhang Y, Zhao Y, Zhong X, Zhou DX, Zhou M, Zhou Y, Zhu B, Zhu JK, Liu Q. Epigenetics in the modern era of crop improvements. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1570-1609. [PMID: 39808224 DOI: 10.1007/s11427-024-2784-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/15/2024] [Indexed: 01/16/2025]
Abstract
Epigenetic mechanisms are integral to plant growth, development, and adaptation to environmental stimuli. Over the past two decades, our comprehension of these complex regulatory processes has expanded remarkably, producing a substantial body of knowledge on both locus-specific mechanisms and genome-wide regulatory patterns. Studies initially grounded in the model plant Arabidopsis have been broadened to encompass a diverse array of crop species, revealing the multifaceted roles of epigenetics in physiological and agronomic traits. With recent technological advancements, epigenetic regulations at the single-cell level and at the large-scale population level are emerging as new focuses. This review offers an in-depth synthesis of the diverse epigenetic regulations, detailing the catalytic machinery and regulatory functions. It delves into the intricate interplay among various epigenetic elements and their collective influence on the modulation of crop traits. Furthermore, it examines recent breakthroughs in technologies for epigenetic modifications and their integration into strategies for crop improvement. The review underscores the transformative potential of epigenetic strategies in bolstering crop performance, advocating for the development of efficient tools to fully exploit the agricultural benefits of epigenetic insights.
Collapse
Affiliation(s)
- Yan Xue
- State Key Laboratory of Wheat Improvement, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang, 261325, China.
| | - Xiaofeng Cao
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Xiangsong Chen
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Xian Deng
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Xing Wang Deng
- State Key Laboratory of Wheat Improvement, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang, 261325, China.
| | - Yong Ding
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Aiwu Dong
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Biophysics, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Cheng-Guo Duan
- Key Laboratory of Plant Design, National Key Laboratory of Plant Molecular Genetics, Shanghai Center for Plant Stress Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Xiaofeng Fang
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Lei Gong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China.
| | - Zhizhong Gong
- State Key Laboratory of Plant Environmental Resilience, Frontiers Science Center for Molecular Design Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China.
| | - Xiaofeng Gu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Chongsheng He
- College of Biology, Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, Hunan Engineering and Technology Research Center of Hybrid Rapeseed, Hunan University, Changsha, 410082, China.
| | - Hang He
- Institute of Advanced Agricultural Sciences, School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China.
| | - Shengbo He
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China.
| | - Xin-Jian He
- National Institute of Biological Sciences, Beijing, 102206, China.
| | - Yan He
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Yuehui He
- School of Advanced Agricultural Sciences, 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.
| | - Danhua Jiang
- Key Laboratory of Seed Innovation, State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianjun Jiang
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, Zhengzhou, 450046, China.
| | - Jinsheng Lai
- State Key Laboratory of Maize Bio-breeding, National Maize Improvement Center, Department of Plant Genetics and Breeding, China Agricultural University, Beijing, 100193, China.
- Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, 100193, China.
- Center for Crop Functional Genomics and Molecular Breeding, China Agricultural University, Beijing, 100193, China.
- Sanya Institute of China Agricultural University, Sanya, 572025, China.
- Hainan Yazhou Bay Seed Laboratory, Sanya, 572025, China.
| | - Zhaobo Lang
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Chenlong Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Stress Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
| | - Qing Li
- National Key Laboratory of Crop Genetic Improvement, Huebei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Xingwang Li
- National Key Laboratory of Crop Genetic Improvement, Huebei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Bao Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China.
| | - Bing Liu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Biophysics, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Xiao Luo
- Shandong Provincial Key Laboratory of Precision Molecular Crop Design and Breeding, Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang, 261325, China.
| | - Yijun Qi
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Weiqiang Qian
- School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China.
| | - Guodong Ren
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Biophysics, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Qingxin Song
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Jiangsu Collaborative Innovation Center for Modern Crop Production, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Xianwei Song
- Key Laboratory of Seed Innovation, State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zhixi Tian
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jia-Wei Wang
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Yuan Wang
- Key Laboratory of Seed Innovation, State Key Laboratory of Plant Genomics and National Center for Plant Gene Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Liang Wu
- Zhejiang Provincial Key Laboratory of Crop Genetic Resources, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China.
| | - Zhe Wu
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Rui Xia
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Horticulture, South China Agricultural University, Guangzhou, 510640, China.
| | - Jun Xiao
- Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Lin Xu
- National Key Laboratory of Plant Molecular Genetics, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Zheng-Yi Xu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, 130024, China.
| | - Wenhao Yan
- National Key Laboratory of Crop Genetic Improvement, Huebei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Hongchun Yang
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Jixian Zhai
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Yijing Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Biophysics, Institute of Plant Biology, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Yusheng Zhao
- Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Xuehua Zhong
- Department of Biology, Washington University in St. Louis, St. Louis, 63130, USA.
| | - Dao-Xiu Zhou
- National Key Laboratory of Crop Genetic Improvement, Huebei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
- Institute of Plant Sciences Paris-Saclay (IPS2), CNRS, INRAE, University Paris-Saclay, Orsay, 91405, France.
| | - Ming Zhou
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yue Zhou
- State Key Laboratory of Protein and Plant Gene Research, School of Advanced Agricultural Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| | - Bo Zhu
- Department of Biological Science, College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China.
| | - Jian-Kang Zhu
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Qikun Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
10
|
Settas N, Bittel AJ, Chen YW. Increased METTL3 Expression and m6A Methylation in Myoblasts of Facioscapulohumeral Muscular Dystrophy. Int J Mol Sci 2025; 26:5170. [PMID: 40507980 PMCID: PMC12155334 DOI: 10.3390/ijms26115170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/20/2025] [Accepted: 05/21/2025] [Indexed: 06/16/2025] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is caused by the aberrant expression of the double homeobox 4 (DUX4) gene. In this study, an analysis of human FSHD muscle biopsies revealed differential expressions of six m6A regulators, including writers, readers and eraser proteins. In immortalized human FSHD myoblasts, we found higher levels of mRNA and protein expression of a major m6A regulator, methyltransferase-like protein 3 (METTL3), in comparison with myoblasts from unaffected siblings (UASbs). Quantification of the overall RNA m6A levels in the FSHD myoblasts revealed significant elevation compared with their UASb, which was reversed to UASb levels following treatment with an antisense oligonucleotide targeting the DUX4 mRNA. Using Oxford Nanopore direct-RNA sequencing, we mapped m6A across the transcriptome and identified genes harboring differential methylated m6A sites, including several involved in iron homeostasis. Western blot protein quantification showed that FSHD myoblasts had higher levels of ferritin-heavy chain-207 isoform and mitoferrin-1. In addition, our data showed elevation in mitochondrial ferrous iron in FSHD myoblasts. Our findings suggest that m6A RNA modifications play a pivotal role in FSHD pathophysiology and may serve as biomarker for this disease.
Collapse
Affiliation(s)
- Nikolaos Settas
- Center for Genetic Medicine Research, Children’s National Research and Innovation Campus, Washington, DC 20012, USA; (N.S.); (A.J.B.)
| | - Adam J Bittel
- Center for Genetic Medicine Research, Children’s National Research and Innovation Campus, Washington, DC 20012, USA; (N.S.); (A.J.B.)
| | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children’s National Research and Innovation Campus, Washington, DC 20012, USA; (N.S.); (A.J.B.)
- Department of Genomics and Precision Medicine, School of Medicine and Health Science, The George Washington University, Washington, DC 20037, USA
- Department of Pediatrics, School of Medicine and Health Science, The George Washington University, Washington, DC 20037, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Science, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
11
|
Wu Y, Xu B, Peng Y, Lin S, Du W, Liu R, Zhang S, Wu J, Zou K, Zhao X. H3K18 lactylation-mediated Ythdf2 activation restrains mouse female germline stem cell proliferation via promoting Ets1 mRNA degradation. Clin Epigenetics 2025; 17:84. [PMID: 40426273 PMCID: PMC12107931 DOI: 10.1186/s13148-025-01890-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Germline stem cells are critical for sustaining fertility by balancing self-renewal and differentiation, and are regulated by genetic and epigenetic programs. Although extensively investigated, the rare female germline stem cells (FGSCs) in mammalian ovaries hinder their application in regenerative medicine. The N6-methyladenosine (m6A) reader YTHDF2 is required for female germ cell competence. However, the mechanistic underpinnings of how YTHDF2 regulates FGSC proliferation remain elusive. RESULTS Here, we show that knockout of Ythdf2 enhances FGSC proliferation in vitro. YTHDF2 binds m6A-modified Ets1 mRNA and facilitates its degradation in an m6A-dependent manner. ETS1 functions as a key downstream effector of YTHDF2, as suppression of ETS1 expression partially reverses the Ythdf2-KO-induced phenotype. Additionally, we demonstrate that YTHDF2/ETS1 axis participates in regulating FGSC proliferation by modulation of proliferation-related gene expression. Moreover, histone lactylation modification H3K18la activates the expression of YTHDF2 in FGSCs. CONCLUSIONS Overall, our study reveals that YTHDF2 intrinsically restrains mouse FGSC proliferation and provides a potential strategy to increase FGSC abundance for its potential clinical application.
Collapse
Affiliation(s)
- Yunqiang Wu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Xu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yonglin Peng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sang Lin
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenfei Du
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruiqi Liu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China
| | - Ji Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Kang Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, China.
| | - Xiaodong Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
12
|
Cai S, Zhou J, Luo X, Zhang C, Jin S, Ren J, Cui J. Phase transition of WTAP regulates m 6A modification of interferon-stimulated genes. eLife 2025; 13:RP100601. [PMID: 40424294 DOI: 10.7554/elife.100601] [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: 05/29/2025] Open
Abstract
N6-methyladenosine (m6A) is the most prevalent modification of mRNA which controls diverse physiological processes. Although m6A modification has been reported to regulate type I interferon (IFN) responses by targeting the mRNA of IFN-β and the interferon-stimulated genes (ISGs), the detailed mechanism of how m6A methyltransferase complex (MTC) rapidly responds to conduct the modification on nascent mRNA during IFN-β stimulation remains largely unclear. Here, we demonstrate that WTAP, the adaptor protein of m6A MTC, undergoes dephosphorylation-regulated phase transition from aggregates to liquid-like condensates under IFN-β stimulation, thereby mediating m6A modification of a subset of ISGs to restrict their expression. The phase transition of WTAP promotes the interaction with nucleus-translocated transcription factor STAT1, recruits MTC to the promoter regions of ISGs and directs the co-transcriptional m6A modification on ISG mRNAs. Collectively, our findings reveal a novel regulatory role of WTAP phase transition in manipulating signaling pathways and fine-tuning immune response by orchestrating dynamic m6A modification through the cooperation of transcription factors and MTC. Our findings unveil a novel mechanism by which WTAP phase transition controls immune homeostasis via transcription factor-MTC-driven dynamic m6A modification, thereby proposing a potential therapeutic target for alleviating immune dysregulation.
Collapse
Affiliation(s)
- Sihui Cai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jie Zhou
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, China
| | - Xiaotong Luo
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chenqiu Zhang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jian Ren
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Zhou K, Cai H, Zhou Z, Yi D, Yao Y, Jin Z, Huang P. m6A methylation modification of RNA plays a significant role in the occurrence and development of colorectal cancer. Int J Biol Macromol 2025; 315:144666. [PMID: 40424908 DOI: 10.1016/j.ijbiomac.2025.144666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/23/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
Colorectal cancer is the third most common malignant tumor worldwide and ranks second in terms of mortality. N6-methyladenosine (m6A) modification is the most prevalent internal covalent modification in eukaryotic mRNA and is involved in various stages of RNA processing, including splicing, degradation, and export, playing a crucial role in the onset and progression of many diseases. The m6A modification is co-regulated by methyltransferases, demethylases, and methyl-binding proteins, and it has become a hot topic in cancer research. Based on a systematic review of existing studies on the role of m6A modification in colorectal cancer, this article further expands the research horizon in this field and effectively overcomes the limitations of existing reviews that only focus on discussing a single or a class of methylation regulators.
Collapse
Affiliation(s)
- Ke Zhou
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Huazhong Cai
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Zhengrong Zhou
- School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Dehao Yi
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Yuan Yao
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Zhesi Jin
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China
| | - Pan Huang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, PR China; School of Medicine, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
14
|
Martínez-Campos C, Lanz-Mendoza H, Cime-Castillo JA, Peralta-Zaragoza Ó, Madrid-Marina V. RNA Through Time: From the Origin of Life to Therapeutic Frontiers in Transcriptomics and Epitranscriptional Medicine. Int J Mol Sci 2025; 26:4964. [PMID: 40507776 PMCID: PMC12154163 DOI: 10.3390/ijms26114964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/10/2025] [Accepted: 05/15/2025] [Indexed: 06/16/2025] Open
Abstract
This review examines the evolutionary trajectory and functional versatility of RNA, beginning with its proposed involvement in the origin of life and culminating in its current application in therapeutic strategies. We explored the complexity of the transcriptome, splicing mechanisms, and the regulatory functions of non-coding RNAs, especially microRNAs. The processes underlying microRNA biogenesis and activity are discussed in the context of their potential as therapeutic tools. Advances in RNA-based technologies have been further illustrated by the development of mRNA vaccines, representing a significant breakthrough in biomedical innovation. Additionally, we explored the growing field of epitranscriptomics-chemical modifications to RNA that modulate its stability, translation, and function-by analyzing the roles of modification enzymes known as writers, erasers, and readers. Focus is given to how these alterations influence immune reactions and guide the strategic development of future modified mRNA vaccines. Collectively, these advances underscore RNA's multifaceted roles and its transformative potential in the biological and medical sciences.
Collapse
Affiliation(s)
- Cecilia Martínez-Campos
- National Institute of Genomic Medicine (Instituto Nacional de Medicina Genómica, INMEGEN), Periférico Sur 4809, Ciudad de México 14610, Mexico;
| | - Humberto Lanz-Mendoza
- Center for Research on Infectious Diseases, National Institute of Public Health (Instituto Nacional de Salud Pública, INSP), Universidad 655, Cuernavaca 62100, Mexico; (H.L.-M.); (J.A.C.-C.); (Ó.P.-Z.)
| | - Jorge A. Cime-Castillo
- Center for Research on Infectious Diseases, National Institute of Public Health (Instituto Nacional de Salud Pública, INSP), Universidad 655, Cuernavaca 62100, Mexico; (H.L.-M.); (J.A.C.-C.); (Ó.P.-Z.)
| | - Óscar Peralta-Zaragoza
- Center for Research on Infectious Diseases, National Institute of Public Health (Instituto Nacional de Salud Pública, INSP), Universidad 655, Cuernavaca 62100, Mexico; (H.L.-M.); (J.A.C.-C.); (Ó.P.-Z.)
| | - Vicente Madrid-Marina
- Center for Research on Infectious Diseases, National Institute of Public Health (Instituto Nacional de Salud Pública, INSP), Universidad 655, Cuernavaca 62100, Mexico; (H.L.-M.); (J.A.C.-C.); (Ó.P.-Z.)
| |
Collapse
|
15
|
Szydlo K, Santos L, Christian T, Maharjan S, Dorsey A, Masuda I, Jia J, Wu Y, Tang W, Hou YM, Ignatova Z. m6A modification is incorporated into bacterial mRNA without specific functional benefit. Nucleic Acids Res 2025; 53:gkaf425. [PMID: 40401555 PMCID: PMC12096079 DOI: 10.1093/nar/gkaf425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 05/06/2025] [Indexed: 05/23/2025] Open
Abstract
N 6-Methyladenosine (m6A), the most abundant modification in eukaryotic messenger RNAs (mRNAs), has also been found at a low level in bacterial mRNAs. However, enzyme(s) that introduce m6A modification on mRNAs in bacteria remain elusive. In this work, we combine deep-sequencing approaches that identify m6A sites with in vitro biochemical studies to identify putative m6A methyltransferases that would modify Escherichia coli mRNAs. We tested four uncharacterized candidates predicted to encode proteins with putative methyltransferase domains, whose deletion decreased the m6A level. However, in vitro analysis with the purified putative methyltransferases revealed that none of them installs m6A on mRNA. Exposure to heat and oxidative stress also changed the m6A level; however, we found no clear correlation between the m6A change and the specific stress. Considering two deep-sequencing approaches with different resolution, we found that m6A methylation on bacterial mRNAs is very low and appears randomly introduced. These results suggest that, in contrast to eukaryotes, the m6A modification in bacterial mRNA lacks a direct enzymatic recognition mechanism and has no clear biological function.
Collapse
Affiliation(s)
- Klara Szydlo
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Hamburg 20146, Germany
| | - Leonardo Santos
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Hamburg 20146, Germany
| | - Thomas W Christian
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Sunita Maharjan
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Amir Dorsey
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Isao Masuda
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Jingxuan Jia
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, United States
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, United States
| | - Yuan Wu
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, United States
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, United States
| | - Weixin Tang
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, United States
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, United States
| | - Ya-Ming Hou
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Zoya Ignatova
- Institute of Biochemistry and Molecular Biology, University of Hamburg, Hamburg 20146, Germany
| |
Collapse
|
16
|
Xie S, Hagen D, Becker GM, Davenport KM, Shira KA, Stegemiller MR, Thorne JW, Khilji S, Konetchy D, Villamediana P, Murdoch BM, McKay SD. Analyzing the relationship of RNA and DNA methylation with gene expression. Genome Biol 2025; 26:140. [PMID: 40405312 PMCID: PMC12101012 DOI: 10.1186/s13059-025-03617-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 05/14/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND DNA 5-methylcytosine (5mC) and RNA N6-methyladenosine (m6A) methylation are prevalent modifications in eukaryotes, both playing crucial roles in gene regulation. Recent studies have explored their crosstalk and impact on transcription. However, the intricate relationships among 5mC, m6A, and gene expression remain incompletely elucidated. RESULTS We collect data on 5mC, m6A, and gene expression from samples from three tissues from each of four pregnant cattle and sheep. We construct a comprehensive genome-wide self-interaction (same gene) and across-interaction (across genes) network of 5mC and m6A within gene-bodies or promoters and gene expression in both species. Qualitative analysis identifies uniquely expressed genes with specific m6A methylation in each tissue from both species. A quantitative comparison of gene expression ratio between methylated and unmethylated genes for m6A within gene body and promoter, and 5mC within gene body and promoter confirms the positive effect of RNA methylation on gene expression. Importantly, the influence of RNA methylation on gene expression is stronger than that of DNA methylation. The predominant self- and across-interactions are between RNA methylation within gene bodies and gene expression, as well as between RNA methylation within promoters and gene expression in both species. CONCLUSIONS RNA methylation has a stronger effect on gene expression than does DNA methylation within gene bodies and promoters. DNA and RNA methylation in gene-bodies has a greater impact on gene expression than those in promoters. These findings deepen comprehension of the dynamics and complex relationships among the epigenome, epitranscriptome, and transcriptome, offering fresh insights for advancing epigenetics research.
Collapse
Affiliation(s)
- Shangqian Xie
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Darren Hagen
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Gabrielle M Becker
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Kimberly M Davenport
- Department of Animal Sciences, Washington State University, Pullman, WA, 99164, USA
| | - Katie A Shira
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Morgan R Stegemiller
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Jacob W Thorne
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Sarem Khilji
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Denise Konetchy
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Patricia Villamediana
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Brenda M Murdoch
- Department of Animal, Veterinary and Food Sciences, University of Idaho, Moscow, ID, 83844, USA.
| | - Stephanie D McKay
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
17
|
Ding Z, Wang X, Guo S, Kang Y, Cao M, Hu L, Zhang B, Xiong L, Pei J, Yang T, Guo X. Characteristic analysis of N 6-methyladenine in different parts of yak epididymis. BMC Genomics 2025; 26:500. [PMID: 40389816 PMCID: PMC12087211 DOI: 10.1186/s12864-025-11684-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND The epididymis is essential for sperm maturation. During sperm maturation, markable alterations of the payload of small noncoding RNAs are observed in the epididymis, which indicated the role of epigenetic alterations in sperm maturation. However, the N6-Methyladenosine (m6A) modification profile of the epididymis remains unelucidated. Therefore, in this study, we assessed the m6A modification levels in the caput, corpus, and cauda of the yak epididymis using a combination of methylated RNA immunoprecipitation and RNA sequencing. RESULTS The m6A levels were significantly increased in the corpus of the epididymis. Functional enrichment analysis of differentially methylated RNA (DMR) between the corpus and caput group revealed the significant enrichment of DMRs in the gap junction, ErbB signaling pathway, and mTOR signaling pathway, which participate in cell communication and sperm maturation. In addition, the DMRs of cauda-vs-corpus group were enriched in apoptosis, the FoxO signaling pathway, the PI3K-Akt signaling pathway, and the tumor necrosis factor signaling pathway that were associated with sperm autophagy, oxidative stress, and sperm maturation. Furthermore, we identified the key genes exhibiting significant changes in m6A levels but with no differences in RNA levels, including YY1-associated factor 2, forkhead box J2, and forkhead box O1. This finding indicated that m6A modifications affect these genes during translation, thereby participating in sperm maturation. CONCLUSIONS In summary, we generated the m6A profile of the yak epididymis, which will aid in further elucidating the maturation process of sperm and reveal more information related to male infertility.
Collapse
Affiliation(s)
- Ziqiang Ding
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Xingdong Wang
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Shaoke Guo
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Yandong Kang
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Mengli Cao
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Liyan Hu
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Ben Zhang
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Lin Xiong
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Jie Pei
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China
| | - Tao Yang
- Haibei Animal Husbandry Science and Technology Demonstration Park Management Committee, Haibei, 810299, China.
| | - Xian Guo
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China.
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou, 730050, China.
| |
Collapse
|
18
|
Elsabbagh RA, Abdelhady G, Urlaub D, Sandusky M, Khorshid O, Gad MZ, Abou-Aisha K, Watzl C, Rady M. N 6-methyladenosine RNA base modification regulates NKG2D-dependent and cytotoxic genes expression in natural killer cells. BMC Med Genomics 2025; 18:91. [PMID: 40389988 PMCID: PMC12090489 DOI: 10.1186/s12920-025-02147-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/17/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is the most commonly diagnosed cancer in women. N6-methyladenosine (m6A) is the most prevalent internal modification in mammalian mRNAs and plays a crucial role in various biological processes. However, its function in Natural killer (NK) cells in BC remains unclear. NK cells are essential for cancer immunosurveillance. This study aims to assess m6A levels in transcripts involved in the NKG2D cytotoxicity signaling pathway in NK cells of BC patients compared to controls and find out its impact on mRNA levels. Additionally, it evaluates how deliberately altering m6A levels in NK cells affects mRNA and protein expression of NKG2D pathway genes and NK cell functionality. METHODS m6A methylation in transcripts of NKG2D-pathway-related genes in BC patients and controls was determined using methylated RNA immunoprecipitation-reverse transcription-PCR (MERIP-RT-PCR). To deliberately alter m6A levels in primary cultured human NK cells, the m6A demethylases, FTO and ALKBH5, were knocked out using the CRISPR-CAS9 system, and FTO was inhibited using Meclofenamic acid (MA). The impact of m6A alteration on corresponding mRNA and protein levels was assessed using RT-qPCR and Western blot analysis or flow cytometry, respectively. Additionally, NK cell functionality was evaluated through degranulation and 51Cr release cytotoxicity assays. RESULTS Transcripts of NKG2D, an activating receptor that detects stressed non-self tumour cells, had significantly higher m6A levels in the 3' untranslated region (3'UTR) accompanied by a marked reduction in their corresponding mRNA levels in BC patients compared to controls. Conversely, transcripts of ERK2 and PRF1 exhibited significantly lower m6A levels escorted with higher mRNA expression in BC patients relative to controls. The mRNA levels of PI3K, PAK1 and GZMH were also significantly elevated in BC patients. Furthermore, artificially increasing transcripts' m6A levels via MA in cultured primary NK cells reduced mRNA levels of NKG2D pathway genes and death receptor ligands but did not affect protein expression or NK cell functionality. CONCLUSION Transcripts with higher m6A levels in the 3'UTR region were less abundant, and vice versa. However, changes in mRNA levels of the target genes didn't impact their corresponding protein levels or NK cell functionality.
Collapse
Affiliation(s)
- Raghda A Elsabbagh
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt
| | - Ghada Abdelhady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt
| | - Doris Urlaub
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), TU Dortmund, Dortmund, Germany
| | - Mina Sandusky
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), TU Dortmund, Dortmund, Germany
| | - Ola Khorshid
- Medical Oncology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Z Gad
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt
| | - Khaled Abou-Aisha
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt
| | - Carsten Watzl
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), TU Dortmund, Dortmund, Germany.
| | - Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, the German University in Cairo, Cairo, Egypt.
- Faculty of Biotechnology, German International University, New Administrative Capital, Egypt.
| |
Collapse
|
19
|
Tepe ML, Chen Y, Carso A, Zhou H. MapID-based quantitative mapping of chemical modifications and expression of human transfer RNA. Cell Chem Biol 2025; 32:752-766.e7. [PMID: 40318625 DOI: 10.1016/j.chembiol.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/12/2025] [Accepted: 04/11/2025] [Indexed: 05/07/2025]
Abstract
Detection and quantification of tRNA chemical modifications are critical for understanding their regulatory functions in biology and diseases. However, tRNA-seq-based methods for modification mapping encountered challenges both experimentally (poor processivity of heavily modified tRNAs during reverse transcription or RT) and bioinformatically (frequent reads misalignment to highly similar tRNA genes). Here, we report "MapID-tRNA-seq" where we deployed an evolved reverse transcriptase (RT-1306) into tRNA-seq and developed "MapIDs" that reduce redundancy of the human tRNA genome and explicitly annotate genetic variances. RT-1306 generated robust mutations against m1A and m3C, and RT stops against multiple bulky roadblock modifications. MapID-assisted data processing enabled systematic exclusion of false-positive discoveries of modifications which arise from reads misalignment onto similar genes. We applied MapID-tRNA-seq into mapping m1A, m3C and expression levels of tRNAs in three mammary cell lines, which revealed cell-type dependent modification sites and potential translational regulation of the reduced mitochondrial activities in breast cancer.
Collapse
Affiliation(s)
- Mitchel L Tepe
- Chemistry Department, Boston College, Chestnut Hill, MA 02467, USA
| | - Yitan Chen
- Chemistry Department, Boston College, Chestnut Hill, MA 02467, USA
| | - Allison Carso
- Chemistry Department, Boston College, Chestnut Hill, MA 02467, USA
| | - Huiqing Zhou
- Chemistry Department, Boston College, Chestnut Hill, MA 02467, USA.
| |
Collapse
|
20
|
Li C, Chen K, Li X, Xiong X. Epitranscriptome-epigenome interactions in development and disease mechanisms. Trends Genet 2025:S0168-9525(25)00097-6. [PMID: 40374434 DOI: 10.1016/j.tig.2025.04.009] [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: 01/17/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 05/17/2025]
Abstract
Crosstalk between epitranscriptomic modifications to RNA and epigenomic modifications to DNA and histones plays fundamental roles in development and disease. Here, we summarize two major regulatory modes of the crosstalk between the epigenome and epitranscriptome. In the 'cis mode', the crosstalk occurs co-transcriptionally, with direct interactions observed between epigenetic modifications mediated by their regulators. In the 'trans mode', the modification of an epigenetic layer regulates the expression of another epigenetic layer's writers/erasers and subsequently induces downstream epigenetic alteration. Additionally, we focus on the functional roles of the crosstalk mechanism in physiological and pathological contexts, including development, differentiation, cancer, and complex genetic diseases. Lastly, we discuss the potential future directions for a systematic understanding of epigenetic crosstalk in development and disease.
Collapse
Affiliation(s)
- Chengyu Li
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Kexuan Chen
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Xiaoyu Li
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China
| | - Xushen Xiong
- The Second Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 311121, China; State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 311121, China.
| |
Collapse
|
21
|
Feng A, Liang Y, Fu P, Dong Y, Black SM, Wang T. Endotoxin-induced m6A RNA methylation landscape in lung endothelial cells: role of METTL3 in regulating inflammation and injury during acute lung injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167907. [PMID: 40379220 DOI: 10.1016/j.bbadis.2025.167907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/31/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Acute Lung Injury (ALI) involves diffuse alveolar damage, neutrophil infiltration, and pulmonary edema, with unacceptable mortality. Bacterial lipopolysaccharide (LPS) activates inflammatory pathways in ALI, which are then regulated by transcriptional and post-transcriptional pathways to affect gene expression. RNA methylation, N6-methyladenosine, is the main m6A mRNA modification that controls the expression of various genes in different environments. There are very few facts about LPS's effect on m6A RNA methylation. This study will explore the m6A RNA methylation landscape in lung endothelial cells (ECs) to understand its role in lung inflammation. In this study, lung endothelial cells were treated with LPS, and the dynamics of mRNA m6A methylation were examined through m6A-methylated RNA sequencing. RNA abundance was measured with RNA-seq, and global protein expression and m6A-binding proteins were identified using mass spectrometry (MS). Following LPS treatment, global m6A methylation levels increased along with the upregulation and nuclear translocation of METTL3 protein, while demethylase activity remained unchanged. METTL3 drove LPS-induced m6A methylation and endothelial injury, as shown by selective METTL3 siRNA and the inhibitor STM2457. MeRIP-seq analyses revealed increased m6A sites near the 5' UTR in LPS-treated cells, with m6A methylation correlating positively with gene expression. The metabolic and apoptosis pathways were shown to be more enriched in different types of methylated exons. METTL3-mediated m6A methylation targeted inflammatory genes, enhancing protein expression in chemokine signaling and MAPK pathways. STM2457 effectively mitigated LPS- or CLP-induced experimental ALI. According to this paper, LPS-mediated m6A RNA methylation is described in terms of genomic structure. Modulation of m6A methylation exerts influence over LPS-mediated endothelial gene expression and the ensuing inflammatory response.
Collapse
Affiliation(s)
- Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Yishu Dong
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA; Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA; Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
22
|
Jiang Y, Liang X, Sun H, Yin P, Zhou J, Yu C. Immunomodulatory role of RNA modifications in sex hormone-dependent cancers. Front Immunol 2025; 16:1513037. [PMID: 40406121 PMCID: PMC12095187 DOI: 10.3389/fimmu.2025.1513037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Recent studies have identified that RNA epigenetic modifications, including m6A, m1A, m5C, etc, play pivotal roles in tumor progression. These modifications influence mRNA stability, RNA processing, translational efficiency, and decoding precision. However, comprehensive reviews detailing the connection between m6A RNA modifications and hormone-dependent cancers in both male and female populations remain scarce(breast cancer, ovarian cancer, and endometrial cancer, prostate cancer). In this article, we explore the cellular and molecular roles of various RNA modifications alongside the key elements of the tumor microenvironment. We examine how these RNA modifications influence the development of hormone-dependent cancers through their impact on immune mechanisms. By enhancing our understanding of the function of RNA modifications within the immune systems of four specific tumors, we offer fresh insights for their potential applications in diagnosis and treatment.
Collapse
Affiliation(s)
- Yujia Jiang
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaolan Liang
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hongyi Sun
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ping Yin
- Department of Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhou
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chaoqin Yu
- Gynecology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
23
|
Gao B, Pan H, Zhou X, Yu L, Gao Y, Zhang T, Gao X, Hou J. RNA demethylase ALKBH5 regulates cell cycle progression in DNA damage response. Sci Rep 2025; 15:16059. [PMID: 40341728 PMCID: PMC12062394 DOI: 10.1038/s41598-025-01207-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 05/05/2025] [Indexed: 05/11/2025] Open
Abstract
RNA N6-methyladenosine (m6A) modification plays a crucial role in the DNA damage response, while the detailed mechanisms remain to be explored. In this study, we report the involvement of the m6A demethylase ALKBH5 in X-ray-induced DNA damage response. Depletion of ALKBH5 reduces X-ray-induced DNA damage, induces G2/M phase arrest and reduces cell apoptosis. RNA sequencing and m6A sequencing analysis reveal that ALKBH5 removes m6A modifications from its target mRNAs and suppresses their expression. A subset of mRNAs encoding cyclin dependent kinase inhibitors, such as CDKN1A and CDKN2B, show increased stability and expression upon ALKBH5 knockdown. Subsequently, the upregulation of CDKN1A and CDKN2B contributes to G2/M phase arrest to facilitate DNA repair. Our findings unveil the epigenetic regulation of cell cycle checkpoint by ALKBH5 in X-ray-induced DNA damage, offering potential targets for DNA damage-based therapy for cancers.
Collapse
Grants
- 82372727, 82073110, 82071729 the National Natural Science Foundation of China
- 82372727, 82073110, 82071729 the National Natural Science Foundation of China
- LZ23H160003 Natural Science Foundation of Zhejiang Province
- LTGY24H040005, LTGY24H040006, LTGY23H040004, LTGY23H040005 the Science Technology Department of Zhejiang Province, China
- WKJ-ZJ-2449, 2023KY368 the Health Commission of Zhejiang Province, China
Collapse
Affiliation(s)
- Bo Gao
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China
| | - Haitao Pan
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China
| | - Xiaoling Zhou
- Environmental Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lei Yu
- Environmental Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yunyi Gao
- Environmental Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Tao Zhang
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.
| | - Xiangwei Gao
- Environmental Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Jingyu Hou
- Environmental Medicine, School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
24
|
Gong T, Zeng Z, Huang Z, Luo Y, Cao X, Li H, Zhao Y, Han D, Li D, Zhong M. Downregulation of METTL3 enhances TRADD-mediated apoptosis in inflammatory bowel disease. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2799-6. [PMID: 40347213 DOI: 10.1007/s11427-024-2799-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/07/2024] [Indexed: 05/12/2025]
Abstract
Dysregulation of RNA N6-methyladenosine (m6A) modification in intestinal epithelial cells (IECs) compromises intestinal homeostasis, which is critical for maintaining gastrointestinal functions, immunity, and barrier integrity in inflammatory bowel disease (IBD). Here we explored the role of m6A modification, particularly through methyltransferase like 3 (METTL3), in IBD pathology and the apoptosis of intestinal stem cells (ISCs). Reduced m6A RNA methylation and METTL3 expression were detected in IBD tissues, which correlated with increased ISC apoptosis and spontaneous enteritis in METTL3-deficient models; mechanistically, Mettl3 depletion increased TRADD expression in a m6A-dependent manner, thereby augmenting the TNF-induced apoptosis pathway, whereas pharmacological inhibition of TRADD ameliorated the apoptotic phenotype in METTL3-deficient models and improved survival rates in the enteritis mouse model, suggesting a novel therapeutic avenue for IBD management. Collectively, METTL3-mediated m6A RNA methylation plays a pivotal role in maintaining intestinal homeostasis and is activated in ISCs to mitigate the hyperactivity of endogenous inflammatory signals; by modulating TRADD transcript metabolism, METTL3 limits excessive ISC apoptosis, providing insights into IBD pathogenesis and treatment strategies.
Collapse
Affiliation(s)
- Tingyue Gong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhiyang Zeng
- Department of Central Laboratory, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai, 201499, China
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zurui Huang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Future Technology, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Luo
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xiya Cao
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Hao Li
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yongheng Zhao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Dali Han
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- College of Future Technology, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- China National Center for Bioinformation, Beijing, 100101, China.
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Joint Research Center for Translational Medicine, ECNU-Fengxian Hospital, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Ming Zhong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
25
|
Li P, Zhang C, Yin W, Tao M, Niu Z, Cui Y, Wu D, Gao F. From bone marrow mesenchymal stem cells to diseases: the crucial role of m 6A methylation in orthopedics. Stem Cell Res Ther 2025; 16:228. [PMID: 40329380 PMCID: PMC12057228 DOI: 10.1186/s13287-025-04364-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Elucidating the molecular mechanisms underlying orthopedic diseases is crucial for guiding therapeutic strategies and developing innovative interventions. N6-methyladenosine (m6A)-an epitranscriptomic modification-has emerged as a key regulator of cellular fate and tissue homeostasis. Specifically, m6A plays a pivotal role in several RNA biological processes such as precursor RNA splicing, 3'-end processing, nuclear export, translation, and stability. Recent advancements indicate that m6A methylation regulates stem cell proliferation and osteogenic differentiation by modulating various signaling pathways. Extensive research has shown that abnormalities in m6A methylation contribute significantly to the onset and progression of various orthopedic diseases such as osteoporosis (OP), osteoarthritis (OA), rheumatoid arthritis (RA), and bone tumors. This review aims to summarize the key proteases involved in m6A methylation and their functions. The detailed mechanisms by which m6A methylation regulates osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) through direct and indirect ways are also discussed, with a focus on specific molecular pathways. Finally, this review analyzes the roles and mechanisms of m6A modification in the development and progression of multiple orthopedic diseases, offering a comprehensive understanding of the pathophysiology of these conditions and proposing new directions and molecular targets for innovative treatment strategies.
Collapse
Affiliation(s)
- Peng Li
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Chu Zhang
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Wen Yin
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Mijia Tao
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Zhipeng Niu
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Yutao Cui
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China.
| | - Dankai Wu
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China.
| | - Feng Gao
- Traumatic orthopedics, The Second Hospital of Jilin University, Changchun, 130041, P. R. China.
| |
Collapse
|
26
|
Huang F, Wang Y, Zhang X, Gao W, Li J, Yang Y, Mo H, Prince E, Long Y, Hu J, Jiang C, Kang Y, Chen Z, Hu YC, Zeng C, Yang L, Chen CW, Chen J, Huang H, Weng H. m 6A/IGF2BP3-driven serine biosynthesis fuels AML stemness and metabolic vulnerability. Nat Commun 2025; 16:4214. [PMID: 40328743 PMCID: PMC12056023 DOI: 10.1038/s41467-025-58966-1] [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: 07/31/2024] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Metabolic reprogramming of amino acids represents a vulnerability in cancer cells, yet the mechanisms underlying serine metabolism in acute myeloid leukemia (AML) and leukemia stem/initiating cells (LSCs/LICs) remain unclear. Here, we identify RNA N6-methyladenosine (m6A) modification as a key regulator of serine biosynthesis in AML. Using a CRISPR/Cas9 screen, we find that depletion of m6A regulators IGF2BP3 or METTL14 sensitizes AML cells to serine and glycine (SG) deprivation. IGF2BP3 recognizies m6A on mRNAs of key serine synthesis pathway (SSP) genes (e.g., ATF4, PHGDH, PSAT1), stabilizing these transcripts and sustaining serine production to meet the high metabolic demand of AML cells and LSCs/LICs. IGF2BP3 silencing combined with dietary SG restriction potently inhibits AML in vitro and in vivo, while its deletion spares normal hematopoiesis. Our findings reveal the critical role of m6A modification in the serine metabolic vulnerability of AML and highlight the IGF2BP3/m6A/SSP axis as a promising therapeutic target.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Humans
- Serine/biosynthesis
- Serine/metabolism
- RNA-Binding Proteins/metabolism
- RNA-Binding Proteins/genetics
- Animals
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Mice
- Cell Line, Tumor
- Glycine/metabolism
- Methyltransferases/metabolism
- Methyltransferases/genetics
- CRISPR-Cas Systems
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- Gene Expression Regulation, Leukemic
Collapse
Affiliation(s)
- Feng Huang
- The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | | | - Xiuxin Zhang
- Bioland Laboratory, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Weiwei Gao
- Guangzhou National Laboratory, Guangzhou, China
- University of Science and Technology of China, Hefei, China
| | - Jingwen Li
- Guangzhou National Laboratory, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Ying Yang
- Guangzhou National Laboratory, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Hongjie Mo
- Guangzhou National Laboratory, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Emily Prince
- Department of Systems Biology and Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
| | - Yifei Long
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiacheng Hu
- Bioland Laboratory, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Chuang Jiang
- Guangzhou National Laboratory, Guangzhou, China
- Bioland Laboratory, Guangzhou, China
| | - Yalin Kang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhenhua Chen
- Department of Systems Biology and Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
| | - Yueh-Chiang Hu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Chengwu Zeng
- Department of Hematology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - Lu Yang
- Department of Systems Biology and Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
| | - Chun-Wei Chen
- Department of Systems Biology and Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA
| | - Jianjun Chen
- Department of Systems Biology and Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Monrovia, CA, USA.
- The Gehr Family Center for Leukemia Research, Beckman Research Institute of City of Hope, Monrovia, CA, USA.
| | - Huilin Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Hengyou Weng
- The First Affiliated Hospital, The Fifth Affiliated Hospital, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
- Bioland Laboratory, Guangzhou, China.
| |
Collapse
|
27
|
Li D, Hu L, Wei Q, Kang L, Sun Y, Jiang Y. Regulatory Mechanism of DHCR7 Gene Expression by Estrogen in Chicken Granulosa Cells of Pre-Hierarchical Follicles. Biomolecules 2025; 15:668. [PMID: 40427562 PMCID: PMC12108600 DOI: 10.3390/biom15050668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
The difference in chicken egg production is closely related to the efficiency of follicle selection, which is marked by granulosa cell differentiation and progesterone production with cholesterol as the substrate. The conversion of 7-dehydrocholesterol to cholesterol catalyzed by 7-Dehydrocholesterol reductase (DHCR7) is the rate-limiting step in cholesterol synthesis. Our previous study revealed that estrogen enhanced the mRNA expression of three DHCR7 transcript variants (T1, T3, and T4) in a dose-dependent manner in the granulosa cells of chicken pre-hierarchical follicles (Pre-GCs). This study investigates the molecular mechanisms through which estrogen regulates DHCR7 in chicken Pre-GCs. At the transcriptional level, through CUT&RUN-qPCR, we found that under basal conditions, sterol-regulatory element binding protein 2 (SREBP2) bound to the promoters of three DHCR7 transcript variants to promote cholesterol synthesis in Pre-GCs to maintain low cholesterol levels; meanwhile upon estrogen treatment, estrogen receptors α and β bound to the regulatory regions of three chicken DHCR7 transcript variants, leading to a reduction in the interaction between SREBP2 and DHCR7. At the translational level, the upstream open reading frames (uORFs) and N6-methyladenosine (m6A) modification in the 5'UTR of different DHCR7 transcripts differentially regulate the expression of T3 and T4, as detected by dual-luciferase reporter assays, but this regulation is not affected by estrogen. This study systematically explores the molecular mechanisms through which estrogen upregulates DHCR7 expression in chicken Pre-GCs and provides a clue for understanding the molecular mechanisms underlying cholesterol synthesis in chicken ovarian follicles.
Collapse
Affiliation(s)
- Dandan Li
- College of Animal Science and Technology, Shandong Agricultural University, 7 Panhe Street, Taian 271017, China; (D.L.); (L.H.)
| | - Longxiao Hu
- College of Animal Science and Technology, Shandong Agricultural University, 7 Panhe Street, Taian 271017, China; (D.L.); (L.H.)
| | - Qingqing Wei
- College of Animal Science and Technology, Shandong Agricultural University, 7 Panhe Street, Taian 271017, China; (D.L.); (L.H.)
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian 271017, China
| | - Li Kang
- College of Animal Science and Technology, Shandong Agricultural University, 7 Panhe Street, Taian 271017, China; (D.L.); (L.H.)
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian 271017, China
| | - Yi Sun
- College of Animal Science and Technology, Shandong Agricultural University, 7 Panhe Street, Taian 271017, China; (D.L.); (L.H.)
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian 271017, China
| | - Yunliang Jiang
- College of Animal Science and Technology, Shandong Agricultural University, 7 Panhe Street, Taian 271017, China; (D.L.); (L.H.)
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, Shandong Agricultural University, Taian 271017, China
| |
Collapse
|
28
|
Shang M, Qin J, Zhao B, Luo Q, Wang H, Yang C, Liu H, Ran J, Yang W. Recent Advance in Sensitive Detection of Demethylase FTO. Chembiochem 2025; 26:e202400995. [PMID: 39714929 DOI: 10.1002/cbic.202400995] [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: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
Methylation modification is a critical regulatory mechanism in epigenetics and plays a significant role in various biological processes. N6-methyladenosine (m6A) is the most common modification found in RNA. The fat mass and obesity-associated protein (FTO) facilitate the demethylation of m6A in RNA, and its abnormal expression is closely linked to the development of several diseases. As a result, FTO has the potential to serve as an important biomarker for clinical disease diagnosis. Despite its significance, there has been a lack of comprehensive reviews addressing advancements in detection methods for the demethylase FTO. This review provides an overview of the progress in FTO detection methods, ranging from traditional approaches to innovative techniques, with a particular emphasis on recently reported advancements. These novel detection methods can be categorized into strategies based on enzymes, functional nucleic acids (FNA), and conformational changes. We summarize the principles and applications of these detection methods and discuss the current challenges and prospects in this field.
Collapse
Affiliation(s)
- Min Shang
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Jingying Qin
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Bingyue Zhao
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Qian Luo
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Huimin Wang
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Changying Yang
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Hanghang Liu
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Jiabing Ran
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| | - Wei Yang
- College of Biological and Pharmaceutical Sciences, Key Laboratory of Natural Products, China Three Gorges University, 443002, Yichang, Hubei, P. R. China
| |
Collapse
|
29
|
Zhang X, Tang Y, Wang J, Yang M, Jiang J, Xue H, Wang Y, Zhang J, Wang X. Heat stress enhances the expression of METTL3 to mediate N6-methyladenosine modification of SOS2 and NLRP3 inflammasome activation in boar Sertoli cells. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137432. [PMID: 39884044 DOI: 10.1016/j.jhazmat.2025.137432] [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: 11/05/2024] [Revised: 01/14/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Heat stress negatively affects pig production by disrupting the immune homeostasis of Sertoli cells (SCs), which compromises sperm quality, culminating in male infertility. Herein, we aimed to study the mechanism by which the NLRP3 inflammasome is activated by heat stress through N6-methyladenosine (m6A) modification regulation in SCs. Initially, it was found that heat stress (44°C, 30 min) markedly activated ERK1/2 signaling, which subsequently promoted NLRP3 inflammasome activation and inflammatory cytokine release from SCs. Then, using an m6A dot-blot assay, m6A sequencing, and methylated RNA immunoprecipitation, we found that heat stress augmented the level of m6A modification in SCs, and METTL3 augmented the m6A modification of mRNA encoding SOS Ras/Rho guanine nucleotide exchange factor 2 (SOS2), a key activator of the ERK pathway. Furthermore, YTHDF1 recognized and bound to the m6A-modified SOS2 mRNA to enhance its translation efficiency, ultimately triggering ERK1/2 signaling activation. In vivo experiments demonstrated that heat stress-induced decline in semen quality in mice was associated with elevated levels of m6A modifications in the testis and NLRP3 inflammasome activation. However, the damage caused by heat stress could be attenuated by intraperitoneal injection of S-Adenosylhomocysteine (SAH), a specific methyltransferase inhibitor. Our results emphasize the critical roles of m6A in regulating NLRP3 inflammasome activation under heat stress, identifying a novel therapeutic avenue to address heat stress.
Collapse
Affiliation(s)
- Xuhua Zhang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing 400715, PR China
| | - Yan Tang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing 400715, PR China
| | - Jinxuan Wang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing 400715, PR China
| | - Mengyu Yang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing 400715, PR China
| | - Jing Jiang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing 400715, PR China
| | - Hongyan Xue
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing 400715, PR China
| | - Yi Wang
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Jiaojiao Zhang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing 400715, PR China.
| | - Xianzhong Wang
- Chongqing Key Laboratory of Forage & Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing 400715, PR China.
| |
Collapse
|
30
|
Liu D, Zhao Y, Dai J, Li R, Yuan J, Shen K, He Z, Gu S. Changes of N6-methyladenosine and ferroptosis in cadmium-induced reproductive toxicity of male mice fed a high fat and high sugar diet. Toxicology 2025; 516:154172. [PMID: 40334773 DOI: 10.1016/j.tox.2025.154172] [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: 01/10/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
Cadmium (Cd) and high-fat and high-sugar diet (HFHS) are risk factors contributing to the decline in male sperm quality. N6-methyladenosine (m6A) is essential in the processes of testicular development and spermatogenesis. Ferroptosis, a form of cell death that depends on iron, is prone to causing testicular dysfunction. However, the changes in m6A modification regulatory proteins and ferroptosis signaling molecules in male mice with sperm abnormalities resulting from combined exposure to Cd and HFHS remain incompletely elucidate. Our present data indicate that the combined treatment of Cd and HFHS significantly reduced sperm quality in comparison to those in single Cd or HFHS treatment. In addition, indicators related to ferroptosis in the combined treatment of Cd and HFHS group have also undergone significant changes. In detail, the contents of malondialdehyde (MDA) and Fe2+ as well as Slc7a11 expression were increased while Gclc expressions were reduced in the testicular tissue of Cd and HFHS combined treatment mice. Further detect results showed that the combined exposure to Cd and HFHS synergistically elevated the m6A modification levels alongside a downregulation of the Mettl3, Fto, Alkbh5 and Ythdc2 at the protein level when compared with those in single Cd or HFHS treatment. Altogether, it can be inferred that Cd and HFHS combined treatment may alter the levels of m6A modification regulatory proteins in testicular tissue, leading to increased Fe2+ and MDA production, thus activating the Slc7a11/Gpx4 signaling pathway, ultimately decreasing the sperm quality in mice, providing preliminary evidence for the occurrence of ferroptosis in testicular cells. Our findings may provide direction for the study of reproductive toxicity of cadmium and offer reference for the selection of molecular targets.
Collapse
Affiliation(s)
- De Liu
- School of Public Health, Dali University, Dali, Yunnan, China
| | - Yuan Zhao
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Jiao Dai
- Qujing Medical College, Qujing, Yunnan, China
| | - Rongxian Li
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Jiamin Yuan
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Kaiyan Shen
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Zuoshun He
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Shiyan Gu
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China.
| |
Collapse
|
31
|
Gunes S, Mahmutoglu AM, Hekim N. Epigenetics of nonobstructive azoospermia. Asian J Androl 2025; 27:311-321. [PMID: 39225008 DOI: 10.4103/aja202463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/04/2024] [Indexed: 09/04/2024] Open
Abstract
ABSTRACT Nonobstructive azoospermia (NOA) is a severe and heterogeneous form of male factor infertility caused by dysfunction of spermatogenesis. Although various factors are well defined in the disruption of spermatogenesis, not all aspects due to the heterogeneity of the disorder have been determined yet. In this review, we focus on the recent findings and summarize the current data on epigenetic mechanisms such as DNA methylation and different metabolites produced during methylation and demethylation and various types of small noncoding RNAs involved in the pathogenesis of different groups of NOA.
Collapse
Affiliation(s)
- Sezgin Gunes
- Department of Medical Biology, Medical Faculty, Ondokuz Mayis University, Samsun 55139, Türkiye
| | - Asli Metin Mahmutoglu
- Department of Medical Biology, Medical Faculty, Yozgat Bozok University, Yozgat 66100, Türkiye
| | - Neslihan Hekim
- Department of Medical Biology, Medical Faculty, Ondokuz Mayis University, Samsun 55139, Türkiye
| |
Collapse
|
32
|
Shen Y, Wong SZH, Ma T, Zhang F, Wang Q, Kawaguchi R, Geschwind DH, Wang J, He C, Ming GL, Song H. m 6A deficiency impairs hypothalamic neurogenesis of feeding-related neurons in mice and human organoids and leads to adult obesity in mice. Cell Stem Cell 2025; 32:727-743.e8. [PMID: 40112816 DOI: 10.1016/j.stem.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/07/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
N6-methyladenosine (m6A), the most prevalent internal modification on mRNAs, plays important roles in the nervous system. Whether neurogenesis in the hypothalamus, a region critical for controlling appetite, is regulated by m6A signaling, especially in humans, remains unclear. Here, we showed that deletion of m6A writer Mettl14 in the mouse embryonic hypothalamus led to adult obesity, with impaired glucose-insulin homeostasis and increased energy intake. Mechanistically, deletion of Mettl14 leads to hypothalamic arcuate nucleus neurogenesis deficits with reduced generation of feeding-related neurons and dysregulation of neurogenesis-related m6A-tagged transcripts. Deletion of m6A writer Mettl3 or m6A reader Ythdc1 shared similar phenotypes. METTL14 or YTHDC1 knockdown also led to reduced generation of feeding-related neurons in human brain subregion-specific arcuate nucleus organoids. Our studies reveal a conserved role of m6A signaling in arcuate nucleus neurogenesis in mice and human organoids and shed light on the developmental basis of epitranscriptomic regulation of food intake and energy homeostasis.
Collapse
Affiliation(s)
- Yachen Shen
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel Zheng Hao Wong
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tong Ma
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qing Wang
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Chuan He
- Department of Chemistry, Howard Hughes Medical Institute, the University of Chicago, Chicago, IL, USA; Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, the University of Chicago, Chicago, IL, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Lai Y, Su B, Wang X, Zeng C, Chu H, Zhou L, Bing D. Transcriptomic analysis reveals the function of m6A regulators in aged cochlea. Braz J Otorhinolaryngol 2025; 91:101578. [PMID: 40198960 PMCID: PMC12005922 DOI: 10.1016/j.bjorl.2025.101578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/22/2025] [Accepted: 02/03/2025] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVE Presbycusis is a prevalent health issue among the elderly. Previous studies have shown mechanisms related to this condition, but the underlying mechanisms of presbycusis remain elusive. N6-methyladenosine (m6A) modification in regulating gene expression and cellular functions has been implicated in the development of various diseases. Nevertheless, the potential role of m6A regulators in presbycusis is still unclear. In this study, we aim to determine the expression of m6A regulators in the cochleae of young and old mice, and to investigate their potential role in aging. METHODS We sequenced the transcriptome from the cochleae of six young (2-mo) and six old mice (24-mo) bioinformatics analysis. Differential expression analysis and downstream functional analysis was performed to identify m6A regulators. Association of m6A regulators with protein-protein interaction and transcription factor-miRNA networks were constructed to explore their regulatory mechanisms. RESULTS ALKBH5 and YTHDC1 were found upregulated in the aged cochleae. They were strongly correlated with immune-related pathways, immune molecular subtypes, and immune infiltration levels in old mice, suggesting their potential involvement in immune-related mechanisms of presbycusis. Receiver Operating Characteristic (ROC) curve analysis demonstrated the high diagnostic potential molecules of AlkB Homolog 5 (ALKBH5) and YTHDC1. CONCLUSION This study has established a molecular foundation and introduce a novel perspective on the role of m6A regulators in presbycusis, emphasizing ALKBH5 and YTHDC1 as potential markers. LEVEL OF EVIDENCE Acknowledging methodological similarities with Level 3 (non-randomized controlled cohort or case-control studies) in clinical research, we reference Level 3 as a comparative framework, while recognizing the distinct differences between clinical and animal research settings.
Collapse
Affiliation(s)
- Yanbing Lai
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Bo Su
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Xiaodi Wang
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Chenghui Zeng
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Hanqi Chu
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Liangqiang Zhou
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Dan Bing
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China.
| |
Collapse
|
34
|
Kim JW, Tung HC, Yang B, Pant R, Guan X, Feng Y, Xie W. Heme-thiolate monooxygenase cytochrome P450 1B1, an old dog with many new tricks. Pharmacol Rev 2025; 77:100045. [PMID: 40054133 DOI: 10.1016/j.pharmr.2025.100045] [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: 09/15/2024] [Revised: 01/25/2025] [Accepted: 01/30/2025] [Indexed: 05/12/2025] Open
Abstract
Cytochrome P450 CYP1B1 is a heme-thiolate monooxygenase traditionally recognized for its xenobiotic functions and extrahepatic expressions. Recent studies have suggested that CYP1B1 is also expressed in hepatic stellate cells, immune cells, endothelial cells, and fibroblasts within the tumor microenvironment, as well as tumor cells themselves. CYP1B1 is responsible for the metabolism of a wide range of substrates, including xenobiotics such as drugs, environmental chemicals, and endobiotics such as steroids, retinol, and fatty acids. Consequently, CYP1B1 and its associated exogenous and endogenous metabolites have been critically implicated in the pathogenesis of many diseases. Understanding the mode of action of CYP1B1 in different pathophysiological conditions and developing pharmacological inhibitors that allow for systemic or cell type-specific modulation of CYP1B1 may pave the way for novel therapeutic opportunities. This review highlights the significant role of CYP1B1 in maintaining physiological homeostasis and provides a comprehensive discussion of recent advancements in our understanding of CYP1B1's involvement in the pathogenesis of diseases such as fibrosis, cancer, glaucoma, and metabolic disorders. Finally, the review emphasizes the therapeutic potential of targeting CYP1B1 for drug development, particularly in the treatment and prevention of cancers and liver fibrosis. SIGNIFICANCE STATEMENT: CYP1B1 plays a critical role in various physiological processes. Dysregulation or genetic mutations of the gene encoding this enzyme can lead to health complications and may increase the risk of diseases such as cancer and liver fibrosis. In this review, we summarize recent preclinical and clinical evidence that underscores the potential of CYP1B1 as a therapeutic target.
Collapse
Affiliation(s)
- Jong-Won Kim
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hung-Chun Tung
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bin Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rajat Pant
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiuchen Guan
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Ye Feng
- Department of Endocrinology and Metabolic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
35
|
Zhang R, Liu H, Bai B, Wang H. Quantification of Epigenetic DNA and RNA Modifications by UHPLC-MS/MS Technologies: New Concepts and New Improvements for the Special Collections. J Sep Sci 2025; 48:e70159. [PMID: 40344478 DOI: 10.1002/jssc.70159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 03/21/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025]
Abstract
Dynamic and reversible DNA and RNA modifications are essential for cell differentiation and development. Aberrant epigenetic modifications are closely associated with the occurrence and progression of diseases, serving as potential markers for cancer diagnosis and prognosis. Ultra-high-performance liquid chromatography coupled with tandem mass spectrometry (UHPLC-MS/MS) offers distinct advantages in the qualitative and quantitative analysis of various modifications due to its sensitivity, specificity, and accuracy. This review provides a comprehensive overview of the current knowledge regarding the liquid chromatography-mass spectrometry (LC-MS) analysis of DNA and RNA modifications, including analytical procedures, advancements, and biological applications, with a focus on tracing the source of (N6-2'-deoxy-adenosine) 6mdA in eukaryotes. Additionally, we examine the integration of UHPLC-MS/MS with other separation techniques to achieve accurate quantification of modifications in specific regions, certain fragments, and free nucleosides.
Collapse
Affiliation(s)
- Rui Zhang
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hailong Liu
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Biao Bai
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hailin Wang
- The State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| |
Collapse
|
36
|
Liu K, Wang X, Wang J, Wang S, Bai H, Dong W, Qiao L, Jin Q, Zhang Z, Luo GZ, Wang Z. N 6-methyladenosine modifications stabilize phosphate starvation response-related mRNAs in plant adaptation to nutrient-deficient stress. Nat Commun 2025; 16:4093. [PMID: 40312414 PMCID: PMC12045979 DOI: 10.1038/s41467-025-59331-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: 04/25/2024] [Accepted: 04/16/2025] [Indexed: 05/03/2025] Open
Abstract
N6-methyladenosine (m6A), an abundant internal mRNA modification, is induced by various stress conditions and post-transcriptionally regulates gene expression. However, how m6A modifications help plants respond to nutrient-deficiency stress remains unclear. Here, we profile high-confidence m6A modifications in Arabidopsis transcriptome-wide under normal and inorganic orthophosphate (Pi)-deficient conditions (-P). High-confidence m6A modifications are identified using synthetic modification-free RNA libraries for systematic calibration. Pi starvation induces widespread m6A modifications, mediated by the Pi starvation response (PSR) master regulator PHOSPHATE STARVATION RESPONSE1 (PHR1) and its family members. Many Pi starvation-induced (PSI) m6A modifications occur on PSR-related mRNAs, including PHR1. In addition, PHR1 proteins interact with the m6A writers MRNA ADENOSINE METHYLASE (MTA) and METHYLTRANSFERASE B (MTB) in nuclei under -P conditions. m6A modifications facilitate systemic PSR signaling, as reflected by the reduced Pi content and PSR signaling in a knockdown artificial miRNA line targeting MTA, which shows a global decrease in m6A. Transcriptome-wide mRNA decay analysis reveals that PSI-m6A increases the stability of PSR-related mRNAs, but not through alternative polyadenylation site shifts. Analysis of transgenic plants with mutations in m6A loci demonstrates that m6A stabilizes PHR1 transcripts via a positive feedback loop. Our findings indicate that PSI-m6A modifications facilitate PSR signaling by enhancing the stability of certain mRNAs, shedding light on the role of m6A modifications in nutrient stress responses in plants.
Collapse
Affiliation(s)
- Kai Liu
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaojia Wang
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingyi Wang
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shuman Wang
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haiyan Bai
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Science, South China Normal University, Guangzhou, China
| | - Weiguo Dong
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lulu Qiao
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiongli Jin
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhonghui Zhang
- Guangdong Provincial Key Laboratory of Biotechnology for Plant Development, School of Life Science, South China Normal University, Guangzhou, China
| | - Guan-Zheng Luo
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhiye Wang
- State Key Laboratory of Plant Environmental Resilience, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
37
|
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; 42:1613-1624. [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] [MESH Headings] [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
|
38
|
Zuidhof HR, Müller C, Kortman G, Wardenaar R, Stepanova E, Loayza‐Puch F, Calkhoven CF. The m6A demethylase FTO promotes C/EBPβ-LIP translation to perform oncogenic functions in breast cancer cells. FEBS J 2025; 292:2688-2709. [PMID: 40022434 PMCID: PMC12103066 DOI: 10.1111/febs.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/04/2024] [Accepted: 12/30/2024] [Indexed: 03/03/2025]
Abstract
N6-methyladenosine (m6A) is a prevalent posttranscriptional mRNA modification involved in the regulation of transcript turnover, translation, and other aspects of RNA fate. The modification is mediated by multicomponent methyltransferase complexes (so-called writers) and is reversed through the action of the m6A-demethylases fat mass and obesity-associated (FTO) and alkB homolog 5 (ALKBH5) (so-called erasers). FTO promotes cell proliferation, colony formation and metastasis in models of triple-negative breast cancer (TNBC). However, little is known about genome-wide or specific downstream regulation by FTO. Here, we examined changes in the genome-wide transcriptome and translatome following FTO knockdown in TNBC cells. Unexpectedly, FTO knockdown had a limited effect on the translatome, while transcriptome analysis revealed that genes related to extracellular matrix (ECM) and epithelial-mesenchymal transition (EMT) are regulated through yet unidentified mechanisms. Differential translation of CEBPB mRNA into the C/EBPβ transcription factor isoform C/EBPβ-LIP is known to act in a pro-oncogenic manner in TNBC cells through regulation of EMT genes. Here we show that FTO is required for efficient C/EBPβ-LIP expression, suggesting that FTO has oncogenic functions through regulation of C/EBPβ-LIP.
Collapse
Affiliation(s)
- Hidde R. Zuidhof
- European Institute for the Biology of Ageing (ERIBA)University Medical Center Groningen, University of GroningenThe Netherlands
| | - Christine Müller
- European Institute for the Biology of Ageing (ERIBA)University Medical Center Groningen, University of GroningenThe Netherlands
| | - Gertrud Kortman
- European Institute for the Biology of Ageing (ERIBA)University Medical Center Groningen, University of GroningenThe Netherlands
| | - René Wardenaar
- European Institute for the Biology of Ageing (ERIBA)University Medical Center Groningen, University of GroningenThe Netherlands
| | - Ekaterina Stepanova
- Translational Control and MetabolismGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Fabricio Loayza‐Puch
- Translational Control and MetabolismGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Cornelis F. Calkhoven
- European Institute for the Biology of Ageing (ERIBA)University Medical Center Groningen, University of GroningenThe Netherlands
| |
Collapse
|
39
|
Zhang Y, Diao HT, Leng MY, Wu YZ, Huang BY, Li X, Tang WY, Wu KL, Tan HL, Wang L, Lu W, Xiong A, Shao XQ, Liang HH, Guo J. YTHDF3-mediated FLCN/cPLA2 axis improves cardiac fibrosis via suppressing lysosomal function. Acta Pharmacol Sin 2025; 46:1262-1274. [PMID: 39806064 PMCID: PMC12032138 DOI: 10.1038/s41401-024-01425-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/07/2024] [Indexed: 01/16/2025]
Abstract
Cardiac fibrosis characterized by aberrant activation of cardiac fibroblasts impairs cardiac contractile and diastolic functions, inducing the progression of the disease towards its terminal phase, resulting in the onset of heart failure. Therefore, the inhibition of cardiac fibrosis has become a promising treatment for cardiac diseases. The ovarian follicle-stimulating hormone folliculin (FLCN) plays a significant role in various biological processes, such as lysosome function, mitochondrial synthesis, angiogenesis, ciliogenesis and autophagy. Severe heart failure was observed in FLCN knockout mice. In this study, we investigated the role of FLCN in cardiac fibrosis and its potential mechanisms. The mice were subjected to transverse aortic constriction (TAC) surgery. Myocardial fibrosis developed in the mice 8 weeks after surgery. We showed that the protein and mRNA expression levels of FLCN were significantly decreased in TAC mice. Similar results were observed in primary mouse cardiac fibroblasts treated with Ang-II, an in vitro cardiac fibrosis model, suggesting that FLCN is involved in the pathological process of cardiac fibrosis. We demonstrated that overexpression of FLCN inhibited lysosome function in cardiac fibroblasts. Furthermore, overexpression of FLCN protected the heart from TAC-induced pathological cardiac fibrosis. We revealed that FLCN bound to the cPLA2 protein, increased its activity, regulated lysosomal function, and promoted membrane permeabilisation in cardiac fibroblasts during cardiac fibrosis. Knockdown of cPLA2 blocked the antifibrotic effect of FLCN in cardiac fibrosis. In addition, we found that the reduced expression of FLCN in cardiac fibrosis resulted from the modulation of YTHDF3-regulated m6A methylation of FLCN mRNA. The overexpression of YTHDF3 alleviated the production of collagens and improved cardiac structure and function in TAC mice. YTHDF3 inhibited proliferation and differentiation and regulated lysosomal function in mouse cardiac fibroblasts, whereas these effects were abolished by FLCN knockdown. We conclude that FLCN undergoes YTHDF3-regulated m6A modification and interacts with cPLA2 to improve lysosomal function in cardiac fibroblasts, highlighting its role in myocardial fibrosis therapy. These results suggest that FLCN and YTHDF3 could serve as potential therapeutic targets for cardiac fibroblast treatment.
Collapse
Affiliation(s)
- Yue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Hong-Tao Diao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ming-Yang Leng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ying-Zi Wu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Bing-Ying Huang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xu Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wen-Yue Tang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Kai-Li Wu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Hui-Ling Tan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Liang Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wen Lu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ao Xiong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiao-Qi Shao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Hai-Hai Liang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
40
|
Gu X, Dai X, Sun H, Lian Y, Huang X, Shen B, Zhang P. METTL16 and YTHDC1 Regulate Spermatogonial Differentiation via m6A. Cell Prolif 2025; 58:e13782. [PMID: 39614650 PMCID: PMC12099213 DOI: 10.1111/cpr.13782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/11/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
Spermatogenesis is a highly unique and intricate process, finely regulated at multiple levels, including post-transcriptional regulation. N6-methyladenosine (m6A), the most prevalent internal modification in eukaryotic mRNA, plays a significant role in transcriptional regulation during spermatogenesis. Previous research indicated extensive m6A modification at each stage of spermatogenesis, but depletion of Mettl3 and/or Mettl14 in spermatogenic cells with Stra8-Cre did not reveal any detectable abnormalities up to the stage of elongating spermatids. This suggests the involvement of other methyltransferases in the regulation of m6A modification during spermatogonial differentiation and meiosis. As a METTL3/14-independent m6A methyltransferase, METTL16 remains insufficiently studied in its roles during spermatogenesis. We report that male mice with Mettl16vasa-cre exhibited significantly smaller testes, accompanied by a progressive loss of spermatogonia after birth. Additionally, the deletion of Mettl16 in A1 spermatogonia using Stra8-Cre results in a blockade in spermatogonial differentiation. Given YTHDC1's specific recognition for METTL16 target genes, we further investigated the role of YTHDC1 using Ythdc1-sKO mouse model. Our results indicate that Ythdc1Stra8-cre also impairs spermatogonial differentiation, similar to the effects observed in Mettl16Stra8-cre mice. RNA-seq and m6A-seq analyses revealed that deletion of either Mettl6 or Ythdc1 disrupted the gene expression related to chromosome organisation and segregation, ultimately leading to male infertility. Collectively, this study underscores the essential roles of the m6A writer METTL16 and its reader YTHDC1 in the differentiation of spermatogonia.
Collapse
Affiliation(s)
- Xueying Gu
- State Key Laboratory of Reproductive Medicine and Offspring HealthWomen's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical UniversityNanjingChina
| | - Xinyuan Dai
- State Key Laboratory of Reproductive Medicine and Offspring HealthWomen's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical UniversityNanjingChina
| | - Haifeng Sun
- State Key Laboratory of Reproductive Medicine and Offspring HealthWomen's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical UniversityNanjingChina
| | - Yilong Lian
- State Key Laboratory of Reproductive Medicine and Offspring HealthWomen's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical UniversityNanjingChina
| | | | - Bin Shen
- State Key Laboratory of Reproductive Medicine and Offspring HealthWomen's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical UniversityNanjingChina
| | - Pengfei Zhang
- State Key Laboratory of Reproductive Medicine and Offspring HealthWomen's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical UniversityNanjingChina
- Zhejiang LabHangzhouChina
| |
Collapse
|
41
|
Penning A, Fuks F. The importance of physiological and disease contexts in capturing mRNA modifications. Nat Struct Mol Biol 2025; 32:780-789. [PMID: 40383817 DOI: 10.1038/s41594-025-01548-y] [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: 01/21/2022] [Accepted: 03/18/2025] [Indexed: 05/20/2025]
Abstract
The variety of modifications decorating various RNA species has prompted researchers to study messenger RNA (mRNA) modifications that are likely to have, like N6-methyladenosine (m6A), important biological functions. Yet tackling these modifications has proved more complicated than anticipated. In this Perspective, we discuss two major obstacles to progress in epitranscriptomic research: the low abundance of most mRNA modification and the nonspecificity of many mRNA modifiers. We then shift our focus to the removal of mRNA modifications and their upstream regulation, emphasizing the context-dependent nature of epitranscriptomic regulation. We illustrate how specific modifications, such as N1-methyladenosine (m1A) and pseudouridine, are enriched in distinct environments, most notably within mitochondria and in certain physiopathological conditions. By focusing on biological settings in which non-m6A modifications are more abundant, we could deepen our understanding of their precise roles in gene regulation.
Collapse
Affiliation(s)
- Audrey Penning
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Centre, Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - François Fuks
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Centre, Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium.
| |
Collapse
|
42
|
Yen YP, Lung TH, Liau ES, Wu CC, Huang GL, Hsu FY, Chang M, Yang ZD, Huang CY, Zheng Z, Zhao W, Hung JH, He C, Nie Q, Chen JA. The motor neuron m6A repertoire governs neuronal homeostasis and FTO inhibition mitigates ALS symptom manifestation. Nat Commun 2025; 16:4063. [PMID: 40307231 PMCID: PMC12043976 DOI: 10.1038/s41467-025-59117-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/10/2025] [Indexed: 05/02/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a swiftly progressive and fatal neurodegenerative ailment marked by the degenerative motor neurons (MNs). Why MNs are specifically susceptible in predominantly sporadic cases remains enigmatic. Here, we demonstrated N6-methyladenosine (m6A), an RNA modification catalyzed by the METTL3/METTL14 methyltransferase complex, as a pivotal contributor to ALS pathogenesis. By conditional knockout Mettl14 in murine MNs, we recapitulate almost the full spectrum of ALS disease characteristics. Mechanistically, pervasive m6A hypomethylation triggers dysregulated expression of high-risk genes associated with ALS and an unforeseen reduction of chromatin accessibility in MNs. Additionally, we observed diminished m6A levels in induced pluripotent stem cell derived MNs (iPSC~MNs) from familial and sporadic ALS patients. Restoring m6A equilibrium via a small molecule or gene therapy significantly preserves MNs from degeneration and mitigates motor impairments in ALS iPSC~MNs and murine models. Our study presents a substantial stride towards identifying pioneering efficacious ALS therapies via RNA modifications.
Collapse
Affiliation(s)
- Ya-Ping Yen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| | | | - Ee Shan Liau
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chuan-Che Wu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Guan-Lin Huang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Fang-Yu Hsu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Mien Chang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Zheng-Dao Yang
- Department of Computer Science, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan
| | - Chia-Yi Huang
- Department of Computer Science, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan
| | - Zhong Zheng
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
| | - Wei Zhao
- Department of Mathematics, NSF-Simons Center for Multiscale Cell Fate Research, Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jui-Hung Hung
- Department of Computer Science, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan
| | - Chuan He
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, Chicago, IL, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Qing Nie
- Department of Mathematics, NSF-Simons Center for Multiscale Cell Fate Research, Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jun-An Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
43
|
Liu L, Ge D, Lin Y, Han Z, Zhao H, Cao L, Wu X, Ma G. Epigenetic regulation in oogenesis and fetal development: insights into m6A modifications. Front Immunol 2025; 16:1516473. [PMID: 40356909 PMCID: PMC12066277 DOI: 10.3389/fimmu.2025.1516473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
The unique physiological structure of women has led to a variety of diseases that have attracted the attention of many people in recent years. Disturbances in the reproductive system microenvironment lead to the progression of various female tumours and pregnancy disorders. Numerous studies have shown that epigenetic modifications crucially influence both oogenesis and foetal development. m6A, a modification at the mRNA level, consists of three parts, namely, writers, erasers, and readers, which are involved in several biological functions, such as the nucleation and stabilisation of mRNAs, thereby regulating the development of reproductive system diseases. In this manuscript, we delineate the constituents of m6A, their biological roles, and advancements in understanding m6A within the maternal-foetal immunological context. In addition, we summarise the mechanism of m6A in gynaecological diseases and provide a new perspective for targeting m6A to delay the progression of reproductive system diseases in clinical practice.
Collapse
Affiliation(s)
- Lusheng Liu
- Department of Acupuncture and Moxibustion, Shanghai Traditional Chinese Medicine (TCM)-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Clinical Medical College of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danxia Ge
- Department of Critical Care Medicine, Traditional Chinese Medicine Hospital of, Ningbo, Zhejiang, China
| | - Yumeng Lin
- Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- Department of Acupuncture and Moxibustion, Shanghai Traditional Chinese Medicine (TCM)-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Heng Zhao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liqin Cao
- Department of Gynecology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xi Wu
- Department of Gynecology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guizhi Ma
- Department of Acupuncture and Moxibustion, Shanghai Traditional Chinese Medicine (TCM)-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
44
|
Zhou Y, Cao P, Zhu Q. The regulatory role of m6A in cancer metastasis. Front Cell Dev Biol 2025; 13:1539678. [PMID: 40356596 PMCID: PMC12066624 DOI: 10.3389/fcell.2025.1539678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Metastasis remains a primary cause of cancer-related mortality, with its intricate mechanisms continuing to be uncovered through advancing research. Among the various regulatory processes involved, RNA modification has emerged as a critical epitranscriptomic mechanism influencing cancer metastasis. N6-methyladenosine (m6A), recognized as one of the most prevalent and functionally significant RNA modifications, plays a central role in the regulation of RNA metabolism. In this review, we explore the multifaceted role of m6A in the different stages of cancer metastasis, including epithelial-mesenchymal transition, invasion, migration, and colonization. In addition to summarizing the current state of our understanding, we offer insights into how m6A modifications modulate key oncogenic pathways, highlighting the implications of recent discoveries for therapeutic interventions. Furthermore, we critically assess the limitations of previous studies and propose areas for future research, including the potential for targeting m6A as a novel approach in anti-metastatic therapies. Our analysis provides a comprehensive understanding of the regulatory landscape of m6A in metastasis, offering directions for continued exploration in this rapidly evolving field.
Collapse
Affiliation(s)
- Ying Zhou
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Cao
- Department of Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Yan S, Wen W, Mo Z, Gu S, Chen Z. Epitranscriptomic Role of m6A in Obesity-Associated Disorders and Cancer Metabolic Reprogramming. Genes (Basel) 2025; 16:498. [PMID: 40428320 PMCID: PMC12111259 DOI: 10.3390/genes16050498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
The global rise in obesity and its associated metabolic disorders underscores the need for a deeper investigation into their underlying molecular mechanisms. While genetic factors are well-established contributors, recent research has increasingly focused on epigenetic regulators, particularly N6-methyladenosine (m6A)-the most prevalent internal RNA modification in eukaryotes. This post-transcriptional modification plays a crucial role in RNA metabolism by regulating mRNA stability, splicing, nuclear export, and translation efficiency. Notably, emerging evidence implicates m6A in both adipogenesis and metabolic dysregulation. In this review, we systematically examine three key dimensions: (1) the molecular mechanisms of m6A modification, including writers, erasers, and readers, in obesity; (2) dysregulated m6A patterns in obesity-related pathologies, such as type 2 diabetes (T2D), insulin resistance, metabolic dysfunction-associated steatotic liver disease (MASLD), and the glycolysis in cancer cells; and (3) the therapeutic potential of targeting m6A and the regulators. By critically assessing recent advancements, we highlight m6A's dual role as both a metabolic sensor and a disease modulator, offering novel insights into potential strategies for combating obesity-related metabolic syndromes.
Collapse
Affiliation(s)
- Sujun Yan
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Bin Sheng Road, Binjiang District, Hangzhou 310051, China; (S.Y.); (W.W.); (Z.M.); (S.G.)
| | - Weijing Wen
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Bin Sheng Road, Binjiang District, Hangzhou 310051, China; (S.Y.); (W.W.); (Z.M.); (S.G.)
- School of Public Health, Health Science Center, Ningbo University, 818 Feng Hua Road, Jiangbei District, Ningbo 315211, China
| | - Zhe Mo
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Bin Sheng Road, Binjiang District, Hangzhou 310051, China; (S.Y.); (W.W.); (Z.M.); (S.G.)
| | - Simeng Gu
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Bin Sheng Road, Binjiang District, Hangzhou 310051, China; (S.Y.); (W.W.); (Z.M.); (S.G.)
| | - Zhijian Chen
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Bin Sheng Road, Binjiang District, Hangzhou 310051, China; (S.Y.); (W.W.); (Z.M.); (S.G.)
| |
Collapse
|
46
|
Wu H, Lai GQ, Cheng R, Huang H, Wang J, Liu Z, Gao J, Zhou H, Li C, Yang CG, Liu H. Discovery of Covalent and Cell-Active ALKBH5 Inhibitors with Potent Antileukemia Effects In Vivo. Angew Chem Int Ed Engl 2025; 64:e202424928. [PMID: 39976119 DOI: 10.1002/anie.202424928] [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: 12/19/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 02/21/2025]
Abstract
The N6-methyladenosine (m6A) demethylase ALKBH5 is the only other identified m6A eraser except for FTO, and dysregulated ALKBH5 functions were closely associated with leukemogenesis. However, the development of ALKBH5 inhibitors is slow compared to FTO inhibitors. Inspired by a non-catalytic C200-covalent strategy, a series of maleimide derivatives were designed and synthesized as potent and covalent ALKBH5 inhibitors in this work. The analog 18 l exhibited excellent inhibitory effects on ALKBH5 (IC50=0.62 μM), and exerted a strong antiproliferative effect on NB4 cells with IC50 of 0.63 μM. The Kd value of 18 l binding to ALKBH5 was 804 nM, while no binding was observed with FTO. This result indicated that 18 l was a highly selective inhibitor of ALKBH5 rather than FTO. Additionally, proteomic experiments showed that 18 l directly targeted ALKBH5 in cells and altered m6A levels on mRNA, blocked the related downstream signal pathways, promoted differentiation, and induced apoptosis. Furthermore, 18 l exerted excellent in vivo antitumor activity with TGITV values of 66.3 % at 1 mg/kg in NB4 tumor xenograft models.
Collapse
Affiliation(s)
- Hengbo Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Gan-Qiang Lai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Ruixiang Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Hui Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ju Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Zeyu Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Jing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hu Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Chunpu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Cai-Guang Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| |
Collapse
|
47
|
Artika IM, Arianti R, Demény MÁ, Kristóf E. RNA modifications and their role in gene expression. Front Mol Biosci 2025; 12:1537861. [PMID: 40351534 PMCID: PMC12061695 DOI: 10.3389/fmolb.2025.1537861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 04/02/2025] [Indexed: 05/14/2025] Open
Abstract
Post-transcriptional RNA modifications have recently emerged as critical regulators of gene expression programs. Understanding normal tissue development and disease susceptibility requires knowledge of the various cellular mechanisms which control gene expression in multicellular organisms. Research into how different RNA modifications such as in N6-methyladenosine (m6A), inosine (I), 5-methylcytosine (m5C), pseudouridine (Ψ), 5-hydroxymethylcytosine (hm5C), N1-methyladenosine (m1A), N6,2'-O-dimethyladenosine (m6Am), 2'-O-methylation (Nm), N7-methylguanosine (m7G) etc. affect the expression of genes could be valuable. This review highlights the current understanding of RNA modification, methods used to study RNA modification, types of RNA modification, and molecular mechanisms underlying RNA modification. The role of RNA modification in modulating gene expression in both physiological and diseased states is discussed. The potential applications of RNA modification in therapeutic development are elucidated.
Collapse
Affiliation(s)
- I. Made Artika
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Bogor, Indonesia
| | - Rini Arianti
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Universitas Muhammadiyah Bangka Belitung, Pangkalpinang, Indonesia
| | - Máté Á. Demény
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Endre Kristóf
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
48
|
Yang L, Du X, Hu Y, Wu Z, Bao W. Insight into mechanism of ALKBH5-mediated N 6-methyladenosine (m 6A) regulating porcine epidemic diarrhea virus infection in IPEC-J2 cells. Int J Biol Macromol 2025; 310:143501. [PMID: 40280045 DOI: 10.1016/j.ijbiomac.2025.143501] [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: 02/25/2025] [Revised: 03/29/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Porcine epidemic diarrhea (PED), induced by porcine epidemic diarrhea virus (PEDV) infection, represents a highly contagious swine disease that causes substantial economic losses in the global pig industry. As the most prevalent RNA modification, N6-methyladenosine (m6A) has emerged as a crucial epitranscriptomic regulator of host-pathogen interactions, although its functional significance in PEDV-infected porcine intestinal epithelial cells (IPEC-J2) remains to be fully elucidated. This study drew m6A maps before and after ALKBH5 interfering with IPEC-J2 cells, revealing the mechanism by which ALKBH5 mediated m6A regulates PEDV infection in IPEC-J2 cells. This study employs an in vitro model of PEDV-induced IPEC-J2 cell damage to investigate the regulatory mechanisms of m6A methylation in host antiviral responses. Utilizing a comprehensive multi-omics approach, including MeRIP-seq, RNA-seq, qPCR, Western blot, MeRIP-qPCR, and RIP-qPCR, we systematically uncovered the critical role of m6A methylation in antiviral defense. Our findings reveal that ALKBH5, a key demethylase during PEDV infection, plays a pivotal role in modulating m6A modification levels. Specifically, gene silencing of ALKBH5 significantly upregulates m6A modification but downregulates expression of the antiviral effector genes IFIT3 and HERC5. Mechanistically, we demonstrate that the YTHDC2 protein selectively recognizes m6A modification sites within IFIT3 and HERC5 transcripts, thereby mediating ALKBH5-dependent regulation of mRNA stability and protein expression. Functional analyses further reveal that IFIT3 activates the IRF3/TBK1 signaling axis, while HERC5 enhances antiviral responses by modulating the key effector molecule ISG15. Together, these findings establish a synergistic antiviral mechanism in IPEC-J2 cells. This study is the first to elucidate the novel ALKBH5-YTHDC2-m6A molecular axis, which orchestrates host antiviral immunity through a dual-pathway regulatory mode. These findings provide a theoretical foundation for understanding the role of RNA epigenetic modifications in enterovirus infections and offer a molecular basis for developing m6A-targeted strategies to prevent and control porcine infectious gastroenteritis.
Collapse
Affiliation(s)
- Li Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China.
| | - Xiaomei Du
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Yueqing Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Zhengchang Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China; Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, Jiangsu, PR China.
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China; Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, Jiangsu, PR China.
| |
Collapse
|
49
|
Liu S, Chen W, Chen J, Liu T, Deng M, Xia L, Li Z, Shi J, Li Y, Peng Y, Ren Q, Miao Z, Wu G, Cao X, Xiao S, Zhang J, Zhong M, Wang L, Xia L. m 6A deficiency impairs uterine spiral artery remodeling to induce preeclampsia-like symptoms via FGF2. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2846-4. [PMID: 40304921 DOI: 10.1007/s11427-024-2846-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/30/2024] [Indexed: 05/02/2025]
Abstract
Failures in uterine spiral artery remodeling can lead to placental defects and subsequent preeclampsia, a leading cause of fetal and maternal mortality during pregnancy. N6-methyladenosine (m6A), the most abundant mRNA modification, is dysregulated in samples with preeclampsia. However, whether and how m6A regulates uterine spiral artery remodeling and leads to subsequent preeclampsia in vivo remains unexplored. In this study, we generated two m6A deficiency mouse models: one with a trophoblast-specific knockout of the m6A methyltransferase gene Mettl3, and another with a methyltransferase enzyme mutation. Using these models, we demonstrated that m6A deficiency impaired extravillous trophoblasts (EVTs) infiltration into the uterine spiral arteries, and the remodeling of the spiral arteries in vivo. We further showed that m6A inhibition induced preeclampsia-like symptoms. Mechanistically, we revealed that the m6A modification of FGF2 mRNA, which encodes a secreted peptide implicated in preeclampsia, facilitated its expression. Notably, administration of the FGF2 peptide largely restored EVTs invasion and uterine spiral artery remodeling in m6A-deficient mice. Our findings underscore the importance of m6A in facilitating uterine spiral artery remodeling and prove the pathological mechanisms in vivo, suggesting a new therapeutic approach for preeclampsia caused by m6A deficiency.
Collapse
Affiliation(s)
- Sun Liu
- Department of Pediatric Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenqian Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiaqi Chen
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tianqi Liu
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mingqiang Deng
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Linjian Xia
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zengguang Li
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junfang Shi
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yuan Li
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - You Peng
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qihuan Ren
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ziteng Miao
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guangjin Wu
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Cao
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shan Xiao
- Department of Pediatric Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Zhanjiang Key Laboratory of Zebrafish Model for Development and Diseases, Guangdong Medical University, Zhanjiang, 524001, China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Liping Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Reproductive Medicine Centre, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| | - Laixin Xia
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Department of Obstetrics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
50
|
Miller C, Ealy A, Gregory A, Janarthanam C, Albers W, Richardson G, Jin H, Zenitsky G, Anantharam V, Kanthasamy A, Kanthasamy AG. Pathological α-synuclein dysregulates epitranscriptomic writer METTL3 to drive neuroinflammation in microglia. Cell Rep 2025; 44:115618. [PMID: 40279247 DOI: 10.1016/j.celrep.2025.115618] [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: 10/03/2024] [Revised: 02/11/2025] [Accepted: 04/04/2025] [Indexed: 04/27/2025] Open
Abstract
Recent reports suggest dysregulation of the N6-methyladenosine (m6A) RNA modification may contribute to the pathology of neurodegenerative diseases. Herein, we show the m6A methyltransferase complex including METTL3-the catalytic component of the nuclear-localized complex-is robustly upregulated in human microglia and astrocytes exposed to αSynf and Mn. Subcellular localization studies reveal METTL3 was predominantly cytoplasmic following Mn insult but remained nuclear following αSynf stimulation in activated microglia. Functional analysis revealed METTL3 and downstream m6A readers, including YTHDF2 and IGF2BP1-3, may regulate the proinflammatory secretome of activated microglia. Notably, methyltransferase activity and m6A abundance were significantly increased following Mn and αSynf treatment. METTL3 in Mn and αSynfin vivo models of neuroinflammation, along with human postmortem tissues from Alzheimer's disease (AD), Parkinson's disease (PD), and dementia with Lewy bodies (DLB) patients, was significantly upregulated. This was further confirmed by single-cell RNA sequencing (scRNA-seq) analysis. Overall, we demonstrate the m6A writer METTL3 may function as a major regulator of chronic neuroinflammation in synucleinopathies.
Collapse
Affiliation(s)
- Cameron Miller
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Alyssa Ealy
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA
| | - Amanda Gregory
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA
| | - Chelva Janarthanam
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA
| | - William Albers
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Biology, The University of Georgia, Athens, GA 30602, USA
| | - Gabriel Richardson
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA
| | - Gary Zenitsky
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA
| | - Arthi Kanthasamy
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Anumantha G Kanthasamy
- Isakson Center for Neurological Disease Research, The University of Georgia, Athens, GA 30602, USA; Department of Biology, The University of Georgia, Athens, GA 30602, USA; Department of Biochemistry and Molecular Biology, The University of Georgia, Athens, GA 30602, USA; Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|