1
|
García-Vílchez R, Guallar D. Interplay of transposable elements and ageing: epigenetic regulation and potential epitranscriptomic influence. Curr Opin Genet Dev 2025; 92:102331. [PMID: 40101544 DOI: 10.1016/j.gde.2025.102331] [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: 09/26/2024] [Revised: 02/16/2025] [Accepted: 02/20/2025] [Indexed: 03/20/2025]
Abstract
Transposable elements (TEs) are mobile elements, which have been crucial for mammalian genome evolution and function. Their activity, which influences genomic stability, gene expression and chromatin state, is tightly regulated by complex mechanisms. This review examines recent findings on TE regulation and the dynamics and connection during the ageing process. Here, we explore the interplay between chromatin state, DNA, RNA, and histone modifications in controlling TE activity, with a special emphasis in elucidating the emerging role of epitranscriptomic modifications in TE regulation. Additionally, we analyse the connection between TE activation and ageing, with the perspective for future research that could reveal novel targets for alleviating physiological and pathological ageing and age-related diseases.
Collapse
Affiliation(s)
- Raquel García-Vílchez
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Barcelona Avenue s/n, Santiago de Compostela, A Coruña 15782, Spain. https://twitter.com/@raquelgarcv
| | - Diana Guallar
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Barcelona Avenue s/n, Santiago de Compostela, A Coruña 15782, Spain.
| |
Collapse
|
2
|
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
|
3
|
Chensee G, Lee BS, Green ID, Tieng J, Song R, Pinello N, Lee Q, Mehravar M, Robinson DA, Wang M, Kavurma MM, Yu J, Wong JJ, Liu R. METTL14 promotes intimal hyperplasia through m6A-mediated control of vascular smooth muscle dedifferentiation genes. JCI Insight 2025; 10:e184444. [PMID: 40266881 DOI: 10.1172/jci.insight.184444] [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: 07/05/2024] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
Vascular smooth muscle cells (VSMCs) possess significant phenotypic plasticity, shifting between a contractile phenotype and a synthetic state for vascular repair/remodeling. Dysregulated VSMC transformation, marked by excessive proliferation and migration, primarily drives intimal hyperplasia. N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotes, plays a critical role in gene expression regulation; however, its impact on VSMC plasticity is not fully understood. We investigated the changes in m6A modification and its regulatory factors during VSMC phenotypic shifts and their influence on intimal hyperplasia. We demonstrate that METTL14, crucial for m6A deposition, significantly promoted VSMC dedifferentiation. METTL14 expression, initially negligible, was elevated in synthetic VSMC cultures, postinjury neointimal VSMCs, and human restenotic arteries. Reducing Mettl14 levels in mouse primary VSMCs decreased prosynthetic genes, suppressing their proliferation and migration. m6A-RIP-seq profiling showed key VSMC gene networks undergo altered m6A regulation in Mettl14-deficient cells. Mettl14 enhanced Klf4 and Serpine1 expression through increased m6A deposition. Local Mettl14 knockdown significantly curbed neointimal formation after arterial injury, and reducing Mettl14 in hyperplastic arteries halted further neointimal development. We show that Mettl14 is a pivotal regulator of VSMC dedifferentiation, influencing Klf4- and Serpine1-mediated phenotypic conversion. Inhibiting METTL14 is a viable strategy for preventing restenosis and halting restenotic occlusions.
Collapse
MESH Headings
- Animals
- Methyltransferases/metabolism
- Methyltransferases/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Kruppel-Like Factor 4
- Mice
- Hyperplasia/genetics
- Hyperplasia/pathology
- Humans
- Cell Dedifferentiation/genetics
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Cell Proliferation/genetics
- Tunica Intima/pathology
- Tunica Intima/metabolism
- Neointima/pathology
- Neointima/genetics
- Male
- Cells, Cultured
- Mice, Knockout
- Gene Expression Regulation
- Cell Movement/genetics
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Plasminogen Activator Inhibitor 1/metabolism
- Plasminogen Activator Inhibitor 1/genetics
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Grace Chensee
- Vascular Epigenetics Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Bob Sl Lee
- Vascular Epigenetics Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Immanuel D Green
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Jessica Tieng
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Renhua Song
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Natalia Pinello
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Quintin Lee
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Majid Mehravar
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - David A Robinson
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Centre for Peripheral Artery Disease, Heart Research Institute, Newtown, New South Wales, Australia
| | - Mian Wang
- Department of Vascular Surgery, National Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, the First Affiliated Hospital for Sun Yat-Sen University, Guangzhou, China
| | - Mary M Kavurma
- Centre for Peripheral Artery Disease, Heart Research Institute, Newtown, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Jun Yu
- Center for Metabolic Disease Research and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Justin Jl Wong
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Renjing Liu
- Vascular Epigenetics Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Zhang Q, Liu G, Jing L, Aghayants S, Xu F, Fan Y. The landscape of N 6-methyladenosine RNA methylation in skin diseases. Br J Dermatol 2025; 192:983-994. [PMID: 40059697 DOI: 10.1093/bjd/ljaf087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/15/2025] [Accepted: 03/05/2025] [Indexed: 05/20/2025]
Abstract
Skin diseases encompass a diverse range of conditions with significant psychological and physiological impacts. N6-methyladenosine (m6A) RNA methylation is a key epitranscriptomic modification that regulates gene expression by influencing RNA stability, splicing, translation, export and degradation. Recent studies have highlighted the crucial role of m6A modification in the pathogenesis and progression of various skin diseases. m6A modification affects critical biologic processes of the skin, such as inflammation, immune response and cellular ageing. This review systematically explores the landscape of m6A modification in nontumour skin diseases, elucidating its regulatory roles and therapeutic implications, including wound healing, scar and keloid, skin ageing, psoriasis, systemic lupus erythematosus, acne vulgaris, rosacea, chronic actinic dermatitis and scleroderma. The intricate mechanisms of m6A modification can lead to the development of novel diagnostic biomarkers and therapeutic strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guozhen Liu
- Department of Spinal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Li Jing
- School of Basic Medical Sciences, Ningxia Key Laboratory of Vascular Injury and Repair, Ningxia Medical University, Yinchuan, China
| | - Sis Aghayants
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangjing Xu
- Department of Critical Care Medicine, Yinchuan Hospital of Traditional Chinese Medicine, Affiliated to Ningxia Medical University, Yinchuan, China
| | - Yucheng Fan
- Department of Pathology, The First People's Hospital of Shizuishan, Affiliated to Ningxia Medical University, Shizuishan, China
| |
Collapse
|
5
|
Li X, Liu N. Advances in understanding LINE-1 regulation and function in the human genome. Trends Genet 2025:S0168-9525(25)00103-9. [PMID: 40382218 DOI: 10.1016/j.tig.2025.04.011] [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: 02/21/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025]
Abstract
LINE-1 (long interspersed nuclear element 1, L1) retrotransposons constitute ~17% of human DNA (~0.5 million genomic L1 copies) and exhibit context-dependent expression in different cell lines. Recent studies reveal that L1 is under multilayered control by diverse factors that either collaborate or compete with each other to ensure precise L1 activity. Remarkably, L1s have been co-opted as various transcription-dependent regulatory elements, such as promoters, enhancers, and topologically associating domain (TAD) boundaries, that regulate gene expression in zygotic genome activation, aging, cancer, and other disorders. This review highlights the regulation of L1 and its regulatory functions that influence disease and development.
Collapse
Affiliation(s)
- Xiufeng Li
- State Key Laboratory of Green Biomanufacturing, Tsinghua University-Peking University Joint Center for Life Sciences, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nian Liu
- State Key Laboratory of Green Biomanufacturing, Tsinghua University-Peking University Joint Center for Life Sciences, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
6
|
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
|
7
|
Wang L, Wang Y, Ding K, Li Z, Zhang Z, Li X, Song Y, Xie L, Chen Z. YTHDC1 promotes postnatal brown adipose tissue development and thermogenesis by stabilizing PPARγ. EMBO J 2025:10.1038/s44318-025-00460-x. [PMID: 40355558 DOI: 10.1038/s44318-025-00460-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Brown adipose tissue (BAT) plays a vital role in non-shivering thermogenesis and energy metabolism and is influenced by factors like environmental temperature, ageing, and obesity. However, the molecular mechanisms behind BAT development and thermogenesis are not fully understood. Our study identifies the m6A reader protein YTHDC1 as a crucial regulator of postnatal interscapular BAT development and energy metabolism in mice. YTHDC1 directly interacts with PPARγ through its intrinsically disordered region (IDR), thus protecting PPARγ from binding the E3 ubiquitin ligase ARIH2, and preventing its ubiquitin-mediated proteasomal degradation. Specifically, the ARIH2 RING2 domain is essential for PPARγ degradation, while PPARγ's A/B domain is necessary for their interaction. Deletion of Ythdc1 in BAT increases PPARγ degradation, impairing interscapular BAT development, thermogenesis, and overall energy expenditure. These findings reveal a novel mechanism by which YTHDC1 regulates BAT development and energy homeostasis independently of its m6A recognition function.
Collapse
Affiliation(s)
- Lihua Wang
- HIT Center for Life Sciences, School of Life Science and Technology, State Key Laboratory of Matter Behaviors in Space Environment, Frontier Science Center for Interaction between Space Environment and Matter, Zhengzhou Research Institute, Harbin Institute of Technology, Harbin, 150001, China
| | - Yuqin Wang
- HIT Center for Life Sciences, School of Life Science and Technology, State Key Laboratory of Matter Behaviors in Space Environment, Frontier Science Center for Interaction between Space Environment and Matter, Zhengzhou Research Institute, Harbin Institute of Technology, Harbin, 150001, China
- Department of Cardiovascular Surgery, Institute for Chronic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Kaixin Ding
- HIT Center for Life Sciences, School of Life Science and Technology, State Key Laboratory of Matter Behaviors in Space Environment, Frontier Science Center for Interaction between Space Environment and Matter, Zhengzhou Research Institute, Harbin Institute of Technology, Harbin, 150001, China
| | - Zhenzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, State Key Laboratory of Matter Behaviors in Space Environment, Frontier Science Center for Interaction between Space Environment and Matter, Zhengzhou Research Institute, Harbin Institute of Technology, Harbin, 150001, China
| | - Zhipeng Zhang
- HIT Center for Life Sciences, School of Life Science and Technology, State Key Laboratory of Matter Behaviors in Space Environment, Frontier Science Center for Interaction between Space Environment and Matter, Zhengzhou Research Institute, Harbin Institute of Technology, Harbin, 150001, China
| | - Xinzhi Li
- NHC Key Laboratory of Cell Transplantation, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yue Song
- HIT Center for Life Sciences, School of Life Science and Technology, State Key Laboratory of Matter Behaviors in Space Environment, Frontier Science Center for Interaction between Space Environment and Matter, Zhengzhou Research Institute, Harbin Institute of Technology, Harbin, 150001, China
| | - Liwei Xie
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, State Key Laboratory of Matter Behaviors in Space Environment, Frontier Science Center for Interaction between Space Environment and Matter, Zhengzhou Research Institute, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
8
|
Sun Y, Li J. Mechanistic insights into stem cell fate regulation via RNA methylation. Ageing Res Rev 2025; 107:102717. [PMID: 40054777 DOI: 10.1016/j.arr.2025.102717] [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: 11/13/2024] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 04/13/2025]
Abstract
Stem cells possess an extraordinary ability for self-renewal and differentiation, making them essential for tissue repair, regeneration, and anti-aging. RNA methylation is crucial in regulating stem cell fate by modulating gene expression. This review synthesizes current research on RNA methylation modifications, such as m6A, m7G, m5C, and m1A, and their impact on adult stem cell fate. It provides a comprehensive overview of the molecular machinery involved in RNA methylation, emphasizes the critical roles of these modifications in stem cell biology, reviews the latest advancements in sequencing technologies, and discusses potential crosstalk between RNA methylation and epigenetic mechanisms.
Collapse
Affiliation(s)
- Yushuang Sun
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jingting Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
9
|
Jiang D, Kejiou N, Qiu Y, Palazzo AF, Pennell M. Constraints on the optimization of gene product diversity. Mol Syst Biol 2025; 21:472-491. [PMID: 40210719 PMCID: PMC12048591 DOI: 10.1038/s44320-025-00095-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 02/27/2025] [Accepted: 03/13/2025] [Indexed: 04/12/2025] Open
Abstract
RNA and proteins can have diverse isoforms due to post-transcriptional and post-translational modifications. A fundamental question is whether these isoforms are mostly beneficial or the result of noisy molecular processes. To assess the plausibility of these explanations, we developed mathematical models depicting different regulatory architectures and investigated isoform evolution under multiple population genetic regimes. We found that factors beyond selection, such as effective population size and the number of cis-acting loci, significantly influence evolutionary outcomes. We found that sub-optimal phenotypes are more likely to evolve when populations are small and/or when the number of cis-loci is large. We also discovered that opposing selection on cis- and trans-acting loci can constrain adaptation, leading to a non-monotonic relationship between effective population size and optimization. More generally, our models provide a quantitative framework for developing statistical tests to analyze empirical data; as a demonstration of this, we analyzed A-to-I RNA editing levels in coleoids and found these to be largely consistent with non-adaptive explanations.
Collapse
Affiliation(s)
- Daohan Jiang
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
- Macroevolution Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, Japan
| | - Nevraj Kejiou
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Yi Qiu
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | | | - Matt Pennell
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA.
- Department of Computational Biology, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
10
|
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
|
11
|
Xu Z, Sun B, Wang W, Fan Y, Su J, Sun J, Gu X. Research progress on m6A and drug resistance in gastrointestinal tumors. Front Pharmacol 2025; 16:1565738. [PMID: 40356985 PMCID: PMC12066682 DOI: 10.3389/fphar.2025.1565738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Gastrointestinal (GI) tumors represent a significant global health burden and are among the leading causes of cancer-related mortality worldwide. their drug resistance is one of the major challenges in cancer therapy. In recent years, epigenetic modifications, especially N6-methyladenosine (m6A) RNA modifications, have become a hot research topic. m6A modification plays an important role in gene expression and cancer progression by regulating RNA splicing, translation, stability, and degradation, which are regulated by "writers," "erasers" and "readers." In GI tumors, resistance to chemotherapy, targeted therapy, and immunotherapy is closely associated with m6A RNA modification. Therefore, the molecular mechanism of m6A modification and its targeted drug development provide new therapeutic strategies for overcoming drug resistance and therapeutic efficacy in GI tumors. In this review, the biological functions of m6A were explored, the specific resistance mechanisms of m6A in different types of GI tumors were explored, new ideas and targets for future treatment resistance were identified, and the limitations of this field were highlighted.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiachun Sun
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Xinyu Gu
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
12
|
Wang W, Wang HT, Guo Y, Zhao Q, Lu JT, Cui ZM, Zhang X, Qiu LL, Wang XY, Wang TY, Jia YL. m6A modification profiles of the CHO cells with differential recombinant protein expression using MeRIP-seq/RNA-seq. Int J Biol Macromol 2025; 310:143429. [PMID: 40288720 DOI: 10.1016/j.ijbiomac.2025.143429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/24/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Chinese hamster ovary (CHO) cells remain the primary host system for recombinant therapeutic protein production. Enhancing transgene expression efficiency while maintaining stable production persists as a key challenge in CHO cell engineering. While N6-methyladenosine (m6A) modification - the most abundant RNA methylation - regulates RNA stability and translational efficiency, its role in modulating recombinant protein expression remains underexplored. In this study, through m6A-specific methylated RNA immunoprecipitation sequencing (MeRIP-seq) of high- (ADM-H) and low- (ADM-L) recombinant adalimumab (ADM)-producing CHO cell lines, we identified 668 differentially methylated peaks. Notably, m6A methylation patterns showed positive correlation with heavy chain (HC)/light chain (LC) expression levels between ADM-H and ADM-L cell lines. Differential expression of factors, such as Igf2bp2, Gli2, and Met correlated with PI3K-Akt and Hippo signaling pathways, suggesting m6A-mediated regulatory functions of recombinant protein expression in CHO cells. Furthermore, pharmacological inhibition of Gli2 or Met in cell culture effectively enhanced ADM production while suppressing target gene expression. These findings elucidate m6A's functional role in recombinant protein production and provide actionable strategies for CHO cell line optimization.
Collapse
Affiliation(s)
- Wen Wang
- School of Pharmacy, XinXiang Medical University, Xinxiang 453003, Henan, China; International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang 453003, Henan, China; Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Hai-Tong Wang
- School of Pharmacy, XinXiang Medical University, Xinxiang 453003, Henan, China; International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang 453003, Henan, China; Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yang Guo
- School of Pharmacy, XinXiang Medical University, Xinxiang 453003, Henan, China; International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang 453003, Henan, China; Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Qi Zhao
- School of Pharmacy, XinXiang Medical University, Xinxiang 453003, Henan, China
| | - Jiang-Tao Lu
- School of Pharmacy, XinXiang Medical University, Xinxiang 453003, Henan, China; International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang 453003, Henan, China; Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Zhao-Ming Cui
- School of Pharmacy, XinXiang Medical University, Xinxiang 453003, Henan, China; International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang 453003, Henan, China; Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Xi Zhang
- School of Pharmacy, XinXiang Medical University, Xinxiang 453003, Henan, China; International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang 453003, Henan, China; Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Le-Le Qiu
- School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Xiao-Yin Wang
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang 453003, Henan, China; Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang 453003, Henan, China; School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Tian-Yun Wang
- International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang 453003, Henan, China; Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang 453003, Henan, China; School of Basic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Yan-Long Jia
- School of Pharmacy, XinXiang Medical University, Xinxiang 453003, Henan, China; International Joint Laboratory of Recombinant Drug Protein Expression System, Xinxiang 453003, Henan, China; Henan Engineering Research Center for Biopharmaceutical Innovation, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| |
Collapse
|
13
|
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
|
14
|
Zhang W, Fu H, Huang Y, Zeng M, Ouyang X, Wang X, Ruan D, Ma L, Hu X, Guo J, Galardi JW, Dougan G, Yeung WSB, Li L, Liu J, Feschotte C, Liu P. METTL3-dependent m6A RNA methylation regulates transposable elements and represses human naïve pluripotency through transposable element-derived enhancers. Nucleic Acids Res 2025; 53:gkaf349. [PMID: 40298111 PMCID: PMC12038396 DOI: 10.1093/nar/gkaf349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 03/06/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025] Open
Abstract
N 6-methyladenosine (m6A) is the most prevalent messenger RNA modification with diverse regulatory roles in mammalian cells. While its functions are well-documented in mouse embryonic stem cells (mESCs), its role in human pluripotent stem cells (hPSCs) remains to be fully explored. METTL3 is the main enzyme responsible for m6A deposition. Here, using a METTL3 inducible knockout (iKO) system, we uncovered that, unlike in mESCs, METTL3 was indispensable for hPSC maintenance. Importantly, loss of METTL3 caused significant upregulation of pluripotency factors including naïve pluripotency genes and failure to exit pluripotency, thus impairing stem cell differentiation towards both embryonic and extraembryonic cell lineages. Mechanistically, METTL3 iKO in hPSCs promoted expression and enhancer activities of two primate-specific transposable elements (TEs), SVA_D and HERVK/LTR5_Hs. At SVA_D elements, loss of METTL3 leads to reduced H3K9me3 deposition. On the other hand, the activation of LTR5_Hs in the METTL3 iKO cells is accompanied by increased chromatin accessibility and binding pluripotency factors. The activated SVA_D and LTR5_Hs elements can act as enhancers and promote nearby naïve gene expression by directly interacting with their promoters. Together these findings reveal that METTL3-dependent m6A RNA methylation plays critical roles in suppressing TE expression and in regulating the human pluripotency network.
Collapse
Affiliation(s)
- Weiyu Zhang
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, United States
| | - Haifeng Fu
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yunying Huang
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ming Zeng
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Key laboratory for reproductive Medicine of Guangdong Province, The Third affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Xiangyu Ouyang
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiao Wang
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Degong Ruan
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Liyang Ma
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xinning Hu
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jilong Guo
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Justin W Galardi
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, United States
| | - Gordon Dougan
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Medicine, Jeffrey Cheah Biomedical Centre, Cambridge University, Puddicombe Way, Cambridge Biomedical Campus, Cambridge CB2 0AW, United Kingdom
| | - William S B Yeung
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong ‐ Shenzhen Hospital, Shenzhen 518000, China
| | - Lei Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Key laboratory for reproductive Medicine of Guangdong Province, The Third affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Jianqiao Liu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Key laboratory for reproductive Medicine of Guangdong Province, The Third affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Cedric Feschotte
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, United States
| | - Pentao Liu
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
15
|
Wei S, Tao HY, Duan Z, Wang Y. Environmental Exposure, Epitranscriptomic Perturbations, and Human Diseases. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:6387-6399. [PMID: 40126397 PMCID: PMC11978485 DOI: 10.1021/acs.est.5c00907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Epitranscriptomics is a rapidly evolving field, and it examines how chemical modifications on RNA regulate gene expression. Increasing lines of evidence support that exposure to various environmental agents can change substantially chemical modifications on RNA, thereby perturbing gene expression and contributing to disease development in humans. However, the molecular mechanisms through which environmental exposure impairs RNA modification-associated proteins ("reader", "writer", and "eraser" or RWE proteins) and alters the landscape of RNA modifications remain poorly understood. Here, we provide our perspectives on the current knowledge about how environmental exposure alters the epitranscriptome, where we focus on dynamic changes in RNA modifications and their regulatory proteins elicited by exposure to environmental agents. We discuss how these epitranscriptomic alterations may contribute to the development of human diseases, especially neurodegeneration and cancer. We also discuss the potential and technical challenges of harnessing RNA modifications as biomarkers for monitoring environmental exposure. Finally, we emphasize the need to integrate multiomics approaches to decipher the complex interplay between environmental exposure and the epitranscriptome and offer a forward-looking viewpoint on future research priorities that may inform public health interventions and environmental regulations.
Collapse
Affiliation(s)
- Songbo Wei
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Huan-Yu Tao
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Zheng Duan
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| |
Collapse
|
16
|
Zhao Y, He C. Acetylation of METTL3: A negative regulator of m 6A deposition on chromatin-associated regulatory RNAs. Mol Cell 2025; 85:1251-1252. [PMID: 40185074 DOI: 10.1016/j.molcel.2025.03.002] [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: 03/04/2025] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 04/07/2025]
Abstract
In this issue, Huang et al.1 report the acetylation of METTL3 as a negative regulator of m6A deposition of chromatin-associated regulatory RNAs from enhancers and promoters. This acetylation is mediated by p300 and positively regulated by PAK2.
Collapse
Affiliation(s)
- Yutao Zhao
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA; Howard Hughes Medical Institute, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
17
|
Li Z, Zhang C, Huang G, Zhang Z, Wang Q, Liu X, Qin Y, Zhou H, Hou A, He J, Li L, Hu X, Ding X. Deletion of Tfap2a in hepatocytes and macrophages promotes the progression of hepatocellular carcinoma by regulating SREBP1/FASN/ACC pathway and anti-inflammatory effect of IL10. Cell Death Dis 2025; 16:245. [PMID: 40180937 PMCID: PMC11968862 DOI: 10.1038/s41419-025-07500-8] [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: 11/12/2024] [Revised: 02/13/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025]
Abstract
The transcription factor AP-2α plays a crucial role in the control of tumor development and progression, and suppresses the proliferation and migration of hepatocellular carcinoma (HCC). However, the detailed function and mechanisms of AP-2α in the pathogenesis of HCC are still elusive. In the current study, we investigated the role of AP-2α regulation in liver injury-mediated HCC development. Downregulation of Tfap2a expression was found in the livers of DEN/CCl4-induced fibrosis and HCC mouse model. Hepatocyte (Alb-Cre), hepatic stellate cell (HSC) (Lrat-Cre) and macrophage (LysM-Cre) specific Tfap2a knockout mice were generated, respectively. Conditional knockout of Tfap2a was able to promote hepatic steatosis in Tfap2aΔHep and Tfap2aΔMΦ mice, but not in Tfap2aΔHSC mice fed with normal chow. Tfap2aΔHep and Tfap2aΔMΦ mice treated with DEN/CCl4 for 6 months increased tumor burden compared to Tfap2a flox controls. Tfap2a-deleted macrophages or hepatocytes could enhance lipid droplet (LD) accumulation in hepatocytes. Mechanistically, AP-2α binds to the promoter regions of SREBP1/ACC/FASN and inhibits hepatic lipid de novo synthesis. Deletion of Tfap2a in macrophages enhances polarization of M1 macrophages with increased iNOS expression but decreased CD206 expression, which resulted in increased pro-inflammatory cytokines and decreased anti-inflammatory factors, especially the hepatoprotective factor IL-10. The m6A modification writer WTAP could reduce the mRNA stability of AP-2α in a reader YTHDC1-dependent manner, whereas knockdown of WTAP or YTHDC1 enhances AP-2α expression and decreases lipid accumulation in HCC cells. Clinically, AP-2α expression negatively correlates with the expression of FASN, WTAP, YTHDC1 and the development of liver disease. Taken together, hepatocyte- or macrophage-specific deletion of Tfap2a promotes hepatic steatosis, fibrosis, and the development of HCC. These results suggest that AP-2α has been identified as a novel therapeutic target in fibrosis and inflammation-related HCC, exerting anti-lipogenesis, anti-inflammatory, and anti-tumor multi-roles.
Collapse
Affiliation(s)
- Zhiwei Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Chun Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Guixiang Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Zixin Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Qinghao Wang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Xiran Liu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Yanling Qin
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Hao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Anyi Hou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Jun He
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, 410007, China
| | - Limin Li
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- College of Engineering and Design, Hunan Normal University, Changsha, 410081, China
| | - Xiang Hu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, China
| | - Xiaofeng Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China.
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
- Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
18
|
Huang X, Zhang J, Cun Y, Ye M, Ren Z, Guo W, Ma X, Liu J, Luo W, Sun X, Shao J, Wu Z, Zhu X, Wang J. Spatial control of m 6A deposition on enhancer and promoter RNAs through co-acetylation of METTL3 and H3K27 on chromatin. Mol Cell 2025; 85:1349-1365.e10. [PMID: 40101711 DOI: 10.1016/j.molcel.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/08/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Interaction between the N6-methyladenosine (m6A) methyltransferase METTL3 and METTL14 is critical for METTL3 to deposit m6A on various types of RNAs. It remains to be uncovered whether there is spatial control of m6A deposition on different types of RNAs. Here, through genome-wide CRISPR-Cas9 screening in the A549 cell line, we find that H3K27ac acetylase p300-mediated METTL3 acetylation suppresses the binding of METTL3 on H3K27ac-marked chromatin by inhibiting its interaction with METTL14. Consistently, p300 catalyzing the acetylation of METTL3 specifically occurs on H3K27ac-marked chromatin. Disruptive mutations on METTL3 acetylation sites selectively promote the m6A of chromatin-associated RNAs from p300-bound enhancers and promoters marked by H3K27ac, resulting in transcription inhibition of ferroptosis-inhibition-related genes. In addition, PAK2 promotes METTL3 acetylation by phosphorylating METTL3. Inhibition of PAK2 promotes ferroptosis in a manner that depends on the acetylation of METTL3. Our study reveals a spatial-selective way to specifically regulate the deposition of m6A on enhancer and promoter RNAs.
Collapse
Affiliation(s)
- Xiang Huang
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Medical College of Jiaying University, Meizhou 514031, China
| | - Jie Zhang
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Yixian Cun
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Meijun Ye
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhijun Ren
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Medical College of Jiaying University, Meizhou 514031, China
| | - Wenbing Guo
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China; Medical College of Jiaying University, Meizhou 514031, China
| | - Xiaojun Ma
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiayin Liu
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Weiwei Luo
- GeneMind Biosciences Company Limited, Shenzhen 518000, China
| | - Xiang Sun
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Jingwen Shao
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Zehong Wu
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaofeng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 518000, China
| | - Jinkai Wang
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
19
|
Yang L, Ma M, Gao Y, Liu J. Decoding N 6-methyladenosine's dynamic role in stem cell fate and early embryo development: insights into RNA-chromatin interactions. Curr Opin Genet Dev 2025; 91:102311. [PMID: 39908649 DOI: 10.1016/j.gde.2025.102311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
N6-methyladenosine (m6A), a reversible and dynamic RNA modification, plays pivotal roles in regulating stem cell pluripotency and early embryogenesis. Disruptions in m6A homeostasis lead to profound developmental defects, impairing processes such as stem cell self-renewal, lineage specification, oocyte maturation, zygotic genome activation, and maternal RNA degradation after fertilization. Beyond its well-recognized roles in mRNA transport, stability, and translation, recent studies have highlighted m6A's critical role in transcriptional regulation through intricate RNA-chromatin interactions, notably involving chromatin-associated regulatory RNAs (carRNAs) and retrotransposon RNAs. This review delves into the dynamic regulatory landscape of m6A, highlighting its critical interplay with chromatin modifications, and explores its broader implications in stem cell biology and early embryonic development.
Collapse
Affiliation(s)
- Lei Yang
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Mingli Ma
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yawei Gao
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Sycamore Research Institute of Life Sciences, Shanghai 201203, China.
| | - Jun Liu
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China.
| |
Collapse
|
20
|
Song X, Xu Y, Li M, Guan X, Liu H, Zhang J, Sun H, Ma C, Zhang L, Zhao X, Zheng X, Zhu D. SRSF4-Associated ca-circFOXP1 Regulates Hypoxia-Induced PASMC Proliferation by the Formation of R Loop With Host Gene. Arterioscler Thromb Vasc Biol 2025; 45:e118-e135. [PMID: 39973750 DOI: 10.1161/atvbaha.124.322026] [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: 11/14/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a rare and fatal disease, the pathological changes of which include pulmonary arterial smooth muscle cell (PASMC) proliferation, which is the pathological basis of pulmonary vascular remodeling. Studies have demonstrated that chromatin-associated circRNA can regulate a variety of biological processes. However, the role of chromatin-associated circRNA in the proliferation of PH remains largely unexplored. In this study, we aimed to identify the function and mechanism of chromatin-associated circRNA in PASMC proliferation in PH. METHODS The role of chromatin-associated circFOXP1 (ca-circFOXP1) was investigated in hypoxic mouse PASMCs and SuHX (Sugen5416+hypoxia) model mice through the use of antisense oligonucleotide knockdown and adeno-associated virus-mediated knockdown. Through bioinformatic sequence alignment, chromatin isolation by RNA purification, Cell Counting Kit 8, 5-ethynyl-2-deoxyuridine, Western blot, and other experiments, the function and mechanism of ca-circFOXP1 were verified. RESULTS The expression of ca-circFOXP1 was found to be significantly increased in SuHX model mice and hypoxic mouse PASMCs. Moreover, ca-circFOXP1 was found to regulate the level of the host protein FOXP1 (forkhead box protein 1) through the R loop, thereby influencing the phosphorylation activity of SMAD2 (SMAD family member 2) and, consequently, the proliferation of mouse PASMCs. It is noteworthy that the m6A modification was found to promote the formation of the R loop between ca-circFOXP1 and the host gene FOXP1, thereby regulating the expression of the host protein. Furthermore, we have identified that the splicing factor SRSF4 (serine/arginine rich splicing factor 4) can upregulate the expression of ca-circFOXP1 by splicing exons 6 and 9 of FOXP1 pre-mRNA. CONCLUSIONS The results demonstrated that the splicing factor SRSF4 upregulated the expression of ca-circFOXP1, and m6A methylation promoted R-loop formation between ca-circFOXP1 and host genes, regulated the level of host protein FOXP1, and then affected the phosphorylation activity of SMAD2, mediating PASMC proliferation, leading to pulmonary vascular remodeling. These results provide a theoretical basis for further study of the pathological mechanisms of hypoxic PH and may provide certain insights.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Disease Models, Animal
- Pulmonary Artery/pathology
- Pulmonary Artery/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Mice
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Serine-Arginine Splicing Factors/genetics
- Serine-Arginine Splicing Factors/metabolism
- Cells, Cultured
- Signal Transduction
- Smad2 Protein/metabolism
- Male
- Vascular Remodeling
- Mice, Inbred C57BL
- Hypoxia/metabolism
- Hypoxia/genetics
- Phosphorylation
- Cell Hypoxia
- Repressor Proteins
Collapse
Affiliation(s)
- Xinyue Song
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Ya Xu
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Mengnan Li
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Xiaoyu Guan
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Huiyu Liu
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Jingya Zhang
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Hanliang Sun
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Cui Ma
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- College of Medical Laboratory Science and Technology (C.M., L.Z., X. Zhao), Harbin Medical University (Daqing), P.R. China
| | - Lixin Zhang
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- College of Medical Laboratory Science and Technology (C.M., L.Z., X. Zhao), Harbin Medical University (Daqing), P.R. China
| | - Xijuan Zhao
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- College of Medical Laboratory Science and Technology (C.M., L.Z., X. Zhao), Harbin Medical University (Daqing), P.R. China
| | - Xiaodong Zheng
- Department of Genetic and Cell Biology (X. Zheng), Harbin Medical University (Daqing), P.R. China
| | - Daling Zhu
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Key Laboratory of Cardiovascular Medicine Research, Ministry of Education (D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| |
Collapse
|
21
|
Li K, Yi Y, Ling R, Zhang C, Zhang Z, Wang Y, Wang G, Chen J, Cheng M, Chen S. PCIF1 drives oesophageal squamous cell carcinoma progression via m6Am-mediated suppression of MTF2 translation. Clin Transl Med 2025; 15:e70286. [PMID: 40156159 PMCID: PMC11953057 DOI: 10.1002/ctm2.70286] [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: 12/02/2024] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 04/01/2025] Open
Abstract
Oesophageal squamous cell carcinoma (OSCC) represents a highly aggressive malignancy with limited therapeutic options and poor prognosis. This study uncovers PCIF1 as a critical driver of OSCC progression via m6Am RNA modification, leading to translational repression of the tumour suppressor MTF2. Our results demonstrate that PCIF1 selectively suppresses MTF2 translation, activating oncogenic pathways that promote OSCC growth. In vitro and in vivo models confirm that PCIF1 knockdown reduces OSCC progression, whereas MTF2 knockdown counteracts this effect, highlighting the importance of the PCIF1-MTF2 axis. Clinical analyses further reveal that high PCIF1 expression and low MTF2 expression correlate with advanced tumour stage, poor treatment response and decreased overall survival. Furthermore, in a preclinical mouse model, PCIF1 knockout enhanced the efficacy of anti-PD1 immunotherapy, reducing tumour burden and improving histological outcomes. Notably, flow cytometry analysis indicated that PCIF1 primarily exerts its effects through tumour-intrinsic mechanisms rather than direct modulation of the immune microenvironment, distinguishing its mode of action from PD1 blockade. These findings establish PCIF1 and MTF2 as promising prognostic markers and therapeutic targets for OSCC, offering new avenues for treatment strategies and patient stratification. KEY POINTS: PCIF1 promotes OSCC progression via m6Am methylation at the MTF2 mRNA 5' cap. m6Am methylation suppresses MTF2 translation, enhancing tumour cell proliferation and invasion. Targeting PCIF1 holds therapeutic potential for OSCC treatment.
Collapse
Affiliation(s)
- Kang Li
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yuxuan Yi
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Rongsong Ling
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Caihua Zhang
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhihui Zhang
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer MedicineGuangzhouChina
| | - Yue Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of StomatologySun Yat‐Sen UniversityGuangzhouChina
| | - Ganping Wang
- Department of Urology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jie Chen
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of StomatologySun Yat‐Sen UniversityGuangzhouChina
| | - Maosheng Cheng
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shuang Chen
- Otorhinolaryngology Hospital, Center for Translational MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
22
|
Hu B, Shi Y, Xiong F, Chen YT, Zhu X, Carrillo E, Wen X, Drolet N, Rajpurohit C, Xu X, Lee DF, Soto C, Zhong S, Jayaraman V, Zheng H, Li W. Rewired m6A methylation of promoter antisense RNAs in Alzheimer's disease regulates global gene transcription in the 3D nucleome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.22.644756. [PMID: 40196645 PMCID: PMC11974732 DOI: 10.1101/2025.03.22.644756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
N6-methyladenosine (m6A) is the most prevalent internal RNA modification that can impact mRNA expression post-transcriptionally. Recent progress indicates that m6A also acts on nuclear or chromatin-associated RNAs to impact transcriptional and epigenetic processes. However, the landscapes and functional roles of m6A in human brains and neurodegenerative diseases, including Alzheimer's disease (AD), have been under-explored. Here, we examined RNA m6A methylome using total RNA-seq and meRIP-seq in middle frontal cortex tissues of post-mortem human brains from individuals with AD and age-matched counterparts. Our results revealed AD-associated alteration of m6A methylation on both mRNAs and various noncoding RNAs. Notably, a series of promoter antisense RNAs (paRNAs) displayed cell-type-specific expression and changes in AD, including one produced adjacent to the MAPT locus that encodes the Tau protein. We found that MAPT-paRNA is enriched in neurons, and m6A positively controls its expression. In iPSC-derived human excitatory neurons, MAPT-paRNA promotes expression of hundreds of genes related to neuronal and synaptic functions, including a key AD resilience gene MEF2C, and plays a neuroprotective role against excitotoxicity. By examining RNA-DNA interactome in the three-dimensional (3D) nuclei of human brains, we demonstrated that brain paRNAs can interact with both cis- and trans-chromosomal target genes to impact their transcription. These data together reveal previously unexplored landscapes and functions of noncoding RNAs and m6A methylome in brain gene regulation, neuronal survival and AD pathogenesis.
Collapse
Affiliation(s)
- Benxia Hu
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Yuqiang Shi
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Feng Xiong
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Yi-Ting Chen
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Xiaoyu Zhu
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Elisa Carrillo
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Xingzhao Wen
- Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Nathan Drolet
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Chetan Rajpurohit
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, USA
| | - Dung-Fang Lee
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Claudio Soto
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Sheng Zhong
- Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Vasanthi Jayaraman
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, USA
| | - Wenbo Li
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center and UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
23
|
Krishnan S, Roy A, Wong L, Gromiha M. DRLiPS: a novel method for prediction of druggable RNA-small molecule binding pockets using machine learning. Nucleic Acids Res 2025; 53:gkaf239. [PMID: 40173014 PMCID: PMC11963762 DOI: 10.1093/nar/gkaf239] [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: 10/02/2024] [Revised: 02/16/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
Ribonucleic Acid (RNA) is the central conduit for information transfer in the cell. Identifying potential RNA targets in disease conditions is a challenging task, given the vast repertoire of functional non-coding RNAs in a human cell. A potential druggable target must satisfy several criteria, including disease association, cellular accessibility, binding pockets for drug-like molecules, and minimal cross-reactivity. While several methods exist for prediction of druggable proteins, they cannot be repurposed for RNAs due to fundamental differences in their binding modality. Taking all these constraints into account, a new structure-based model, Druggable RNA-Ligand binding Pocket Selector (DRLiPS), is developed here to predict binding site-level druggability of any given RNA target. A novel strategy for sampling negative binding sites in RNA structures using three parallel approaches is demonstrated here to improve model specificity: backbone motif search, exhaustive pocket prediction, and blind docking. An external blind test dataset has also been curated to showcase the model's generalizability to both experimental and modelled apo state RNA structures. DRLiPS has achieved an F1-score of 0.70, precision of 0.61, specificity of 0.89, and recall of 0.73 on this external test dataset, outperforming two existing methods, DrugPred_RNA and RNACavityMiner. Further analysis indicates that the features selected for model-building generalize well to both apo and holo states with a backbone RMSD tolerance of 3 Å. It can also predict the effect of binding site single point mutations on druggability, which can aid in optimizing synthetic RNA aptamers for small molecule recognition. The DRLiPS model is freely accessible at https://web.iitm.ac.in/bioinfo2/DRLiPS/.
Collapse
Affiliation(s)
- Sowmya Ramaswamy Krishnan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
- TCS Research (Life Sciences division), Tata Consultancy Services, Hyderabad 500081, India
| | - Arijit Roy
- TCS Research (Life Sciences division), Tata Consultancy Services, Hyderabad 500081, India
| | - Limsoon Wong
- Department of Computer Science, National University of Singapore, 117417, Singapore
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
- Department of Computer Science, National University of Singapore, 117417, Singapore
| |
Collapse
|
24
|
Zhang L, Wang J, Tang Z, Lin Z, Su R, Hu N, Tang Y, Ge G, Fan J, Tong MH, Xue Y, Zhou Y, Cheng H. The nuclear exosome co-factor MTR4 shapes the transcriptome for meiotic initiation. Nat Commun 2025; 16:2605. [PMID: 40097464 PMCID: PMC11914058 DOI: 10.1038/s41467-025-57898-0] [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: 02/08/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
Nuclear RNA decay has emerged as a mechanism for post-transcriptional gene regulation in cultured cells. However, whether this process occurs in animals and holds biological relevance remains largely unexplored. Here, we demonstrate that MTR4, the central cofactor of the nuclear RNA exosome, is essential for embryogenesis and spermatogenesis. Embryonic development of Mtr4 knockout mice arrests at 6.5 day. Germ cell-specific knockout of Mtr4 results in male infertility with a specific and severe defect in meiotic initiation. During the pre-meiotic stage, MTR4/exosome represses meiotic genes, which are typically shorter in size and possess fewer introns, through RNA degradation. Concurrently, it ensures the expression of mitotic genes generally exhibiting the opposite features. Consistent with these regulation rules, mature replication-dependent histone mRNAs and polyadenylated retrotransposon RNAs were identified as MTR4/exosome targets in germ cells. In addition, MTR4 regulates alternative splicing of many meiotic genes. Together, our work underscores the importance of nuclear RNA degradation in regulating germline transcriptome, ensuring the appropriate gene expression program for the transition from mitosis to meiosis during spermatogenesis.
Collapse
Affiliation(s)
- Li Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jianshu Wang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhidong Tang
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, RNA Institute, Wuhan University, Wuhan, China
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Zhen Lin
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ruibao Su
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Naijing Hu
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yao Tang
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, RNA Institute, Wuhan University, Wuhan, China
| | - Gaoxiang Ge
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jing Fan
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ming-Han Tong
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yuanchao Xue
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Yu Zhou
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, RNA Institute, Wuhan University, Wuhan, China.
| | - Hong Cheng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
25
|
Liu Y, Liu J, Ren R, Xin Z, Luo Y, Chen Y, Huang C, Liu Y, Yang T, Wang X. Short-term and long-term high-fat diet promote metabolic disorder through reprogramming mRNA m 6A in white adipose tissue by gut microbiota. MICROBIOME 2025; 13:75. [PMID: 40091072 PMCID: PMC11912683 DOI: 10.1186/s40168-025-02047-4] [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] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Although short-term high-fat diet (S-HFD) and long-term high-fat diet (L-HFD) induce metabolic disorder, the underlying epigenetic mechanism is still unclear. RESULTS Here, we found that both 4 days of S-HFD and 10 weeks of L-HFD increased mRNA m6A level in epididymal white adipose tissue (eWAT) and impaired metabolic health. Interestingly, S-HFD activated transposable elements (TEs), especially endogenous retroviruses (ERVs) in eWAT, while L-HFD activated long interspersed elements (LINEs). Subsequently, we demonstrated that both S-HFD and L-HFD increased m6A level of Ehmt2 and decreased EHMT2 protein expression and H3K9me2 level, accounting for activation of ERVs and LINEs. Overexpression of EHMT2 in eWAT or inhibition of ERVs and LINEs by antiviral therapy improved metabolic health under HFD feeding. Notably, we found that both short-term and long-term HFD feeding increased Fimicutes/Bacteroidota ratio and decreased the gut microbiome health index. Fecal microbiota transplantation (FMT) experiments demonstrated that gut microbiota from S-HFD and L-HFD was responsible for increased m6A level in eWAT, resulting in glucose intolerance and insulin insensitivity. Furthermore, we identified that both S-HFD and L-HFD increased the abundance of the gut microbial metabolite homogentisic acid (HGA), and HGA level was positively correlated with unclassified_f__Lachnospiraceae which was both increased in S-HFD and L-HFD feeding mice. Administration of HGA increased the m6A level of Ehmt2 and decreased the EHMT2 protein expression and H3K9me2 level in eWAT, leading to metabolic disorder in mice. CONCLUSIONS Together, this study reveals a novel mechanism that S-HFD and L-HFD induce metabolism disorder through gut microbiota-HGA-m6A-Ehmt2-ERV/LINE signaling. These findings may provide a novel insight for prevention and treatment of metabolism disorder upon short-term or long-term dietary fat intake. Video Abstract.
Collapse
Affiliation(s)
- Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Ruiti Ren
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Zimeng Xin
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Yaojun Luo
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Chaoqun Huang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Yuxi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Tongyudan Yang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou, China.
- Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China.
- Zhejiang Key Laboratory of nutrition and breeding for high-quality animal products, Hangzhou, Zhejiang, China.
| |
Collapse
|
26
|
Gao M, Wu Y, Zhang L, Zhou Y, Liu H, Zhang W, Wang S, Cui Y. YTHDC1 negatively regulates UBE3A to influence RAD51 ubiquitination and inhibit apoptosis in colorectal cancer cells. Sci Rep 2025; 15:8857. [PMID: 40087295 PMCID: PMC11909180 DOI: 10.1038/s41598-025-92925-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
YTHDC1, a key protein in the m6A-related regulatory network within cells, is involved in multiple cellular processes such as chromatin-related regulation, RNA splicing, and nuclear export. Understanding its role in colorectal cancer (CRC) development and DNA damage repair is critical for the advancement of treatment strategies. Our study found that YTHDC1 was highly expressed in high-malignancy CRC tissues compared with low-malignancy ones. Upon silencing YTHDC1, we observed a pronounced suppression of the proliferation of CRC cell lines, accompanied by a substantial increase in cell apoptosis. Furthermore, we identified RAD51 as a crucial downstream target of YTHDC1. Knocking down YTHDC1 led to a notable decrease in RAD51 protein levels, and silencing RAD51 also inhibited cancer cell proliferation. Interestingly, RNA-sequencing data indicated that the YTHDC1 deletion did not affect RAD51 transcription. However, Western blot revealed that this deletion increased the ubiquitination of RAD51, likely due to the upregulated E3 ligase UBE3A. Ubiquitination experiments subsequently confirmed that RAD51 is indeed one of the substrates of UBE3A. In summary, our study provides novel insights into how YTHDC1 modulates the expression of RAD51 through post-translational modifications. These findings offer valuable information that may potentially contribute to the development of more effective therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Mingwei Gao
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yueguang Wu
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Li Zhang
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yan Zhou
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huijuan Liu
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Weimin Zhang
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shubin Wang
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China.
| | - Yongping Cui
- Cancer Institute, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China.
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
27
|
Du Q, Wang Q, Yang C, Wang Y, Yuan H, Zhang B, Ji H, Fu S, Xue C. Investigating the efficacy of immune checkpoint inhibitors in clear cell renal cell carcinoma based on methylation cross talk scoring. Medicine (Baltimore) 2025; 104:e41795. [PMID: 40101071 PMCID: PMC11922407 DOI: 10.1097/md.0000000000041795] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
Methylation processes in different molecular contexts (DNA, RNA, and histones) are controlled by different regulatory factors and serve as critical determinants in cancer development. However, the mechanistic links between these epigenetic modifications during malignant transformation, metastasis, disease relapse, and therapeutic resistance remain incompletely understood. In this research, we investigated the transcriptional and genetic alterations of regulators associated with 3 major types of methylation modifications in clear cell renal cell carcinoma. Utilizing ChIP/MeRIP-seq and 450K methylation array data, we identified genes regulated by multiple methylation modifications and constructed a scoring model to quantify the methylation patterns for each patient. Our findings indicate that patients with a low score may be more likely to respond to immunotherapy, whereas patients with a high score may be more sensitive to targeted therapy, such as RITA, Pazopanib, Irlotinib, SU-11274, BRD-K16762525, and FCCP. In conclusion, the score model can serve as a valuable biomarker to guide clinical selection of immunotherapy and targeted drugs and help to improve personalized clear cell renal cell carcinoma treatment.
Collapse
Affiliation(s)
- Qinglong Du
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qiyuan Wang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chen Yang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yiping Wang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Huiyang Yuan
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Bing Zhang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Hong Ji
- Department of Pathology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Shuai Fu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Chunlei Xue
- Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| |
Collapse
|
28
|
Xu L, Shen T, Li Y, Wu X. The Role of M 6A Modification in Autoimmunity: Emerging Mechanisms and Therapeutic Implications. Clin Rev Allergy Immunol 2025; 68:29. [PMID: 40085180 DOI: 10.1007/s12016-025-09041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
N6-methyladenosine (m6A), a prevalent and essential RNA modification, serves a key function in driving autoimmune disease pathogenesis. By modulating immune cell development, activation, migration, and polarization, as well as inflammatory pathways, m6A is crucial in forming innate defenses and adaptive immunity. This article provides a comprehensive overview of m6A modification features and reveals how its dysregulation affects the intensity and persistence of immune responses, disrupts immune tolerance, exacerbates tissue damage, and promotes the development of autoimmunity. Specific examples include its contributions to systemic autoimmune disorders like lupus and rheumatoid arthritis, as well as conditions that targeting specific organs like multiple sclerosis and type 1 diabetes. Furthermore, this review explores the therapeutic promise of target m6A-related enzymes ("writers," "erasers," and "readers") and summarizes recent advances in intervention strategies. By focusing on the mechanistic and therapeutic implications of m6A modification, this review sheds light on its role as a promising tool for both diagnosis and treatment in autoimmune disorders, laying the foundation for advancements in customized medicine.
Collapse
Affiliation(s)
- Liyun Xu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Tian Shen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yongzhen Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Xiaochuan Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
29
|
Yang Y, Huang Y, Wang T, Li S, Jiang J, Chen S, Chen F, Wang L. mRNA m 6A regulates gene expression via H3K4me3 shift in 5' UTR. Genome Biol 2025; 26:54. [PMID: 40075435 PMCID: PMC11900566 DOI: 10.1186/s13059-025-03515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) is a prevalent and conserved RNA modification in eukaryotes. While its roles in the 3' untranslated regions (3' UTR) are well-studied, its role in the 5' UTR and its relationship with histone modifications remain underexplored. RESULTS We demonstrate that m6A methylation in the 5' UTR of mRNA triggers a downstream shift in H3K4me3 modification. This regulatory mechanism is conserved in Arabidopsis, rice, and chrysanthemum. The observed shift in H3K4me3 is genetically controlled by m6A modifiers and influences gene expression. MTA, the m6A methylase, preferentially binds to phosphorylated serine 5 (Ser5P)-CTD of RNA Pol II during transcription, leading to the displacement of ATX1, the H3K4me3 methylase. This dynamic binding of MTA and ATX1 to RNA Pol II ultimately results in the shift of H3K4me3 modification. Genetic evidence demonstrates that m6A in the 5' UTR controls H3K4me3 shift, thereby affecting SEDOHEPTULOSE-BISPHOSPHATASE expression and leaf senescence. CONCLUSIONS Our study provides new insights into the roles of m6A modification and its crosstalk with histone modification in 5' UTRs, shedding light on the mechanism of m6A-mediated gene expression regulation.
Collapse
Affiliation(s)
- Yuna Yang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Key Laboratory of Flower Biology and Germplasm Innovation, Ministry of Agriculture and Rural Affairs, College of Horticulture, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- Zhongshan Biological Breeding Laboratory, No.50 Zhongling Street, Nanjing, Jiangsu, 210014, China
| | - Yuqing Huang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Key Laboratory of Flower Biology and Germplasm Innovation, Ministry of Agriculture and Rural Affairs, College of Horticulture, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- Zhongshan Biological Breeding Laboratory, No.50 Zhongling Street, Nanjing, Jiangsu, 210014, China
| | - Tian Wang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Key Laboratory of Flower Biology and Germplasm Innovation, Ministry of Agriculture and Rural Affairs, College of Horticulture, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- Zhongshan Biological Breeding Laboratory, No.50 Zhongling Street, Nanjing, Jiangsu, 210014, China
| | - Song Li
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Key Laboratory of Flower Biology and Germplasm Innovation, Ministry of Agriculture and Rural Affairs, College of Horticulture, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- Zhongshan Biological Breeding Laboratory, No.50 Zhongling Street, Nanjing, Jiangsu, 210014, China
| | - Jiafu Jiang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Key Laboratory of Flower Biology and Germplasm Innovation, Ministry of Agriculture and Rural Affairs, College of Horticulture, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- Zhongshan Biological Breeding Laboratory, No.50 Zhongling Street, Nanjing, Jiangsu, 210014, China
| | - Sumei Chen
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Key Laboratory of Flower Biology and Germplasm Innovation, Ministry of Agriculture and Rural Affairs, College of Horticulture, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- Zhongshan Biological Breeding Laboratory, No.50 Zhongling Street, Nanjing, Jiangsu, 210014, China
| | - Fadi Chen
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Key Laboratory of Flower Biology and Germplasm Innovation, Ministry of Agriculture and Rural Affairs, College of Horticulture, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- Zhongshan Biological Breeding Laboratory, No.50 Zhongling Street, Nanjing, Jiangsu, 210014, China
| | - Likai Wang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Key Laboratory of Flower Biology and Germplasm Innovation, Ministry of Agriculture and Rural Affairs, College of Horticulture, Nanjing Agricultural University, Nanjing, 210095, P.R. China.
- Zhongshan Biological Breeding Laboratory, No.50 Zhongling Street, Nanjing, Jiangsu, 210014, China.
| |
Collapse
|
30
|
Chen W, Zhang J, Ma W, Liu N, Wu T. METTL3-Mediated m6A Modification Regulates the Polycomb Repressive Complex 1 Components BMI1 and RNF2 in Hepatocellular Carcinoma Cells. Mol Cancer Res 2025; 23:190-201. [PMID: 39625677 PMCID: PMC11873720 DOI: 10.1158/1541-7786.mcr-24-0362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/15/2024] [Accepted: 11/27/2024] [Indexed: 03/04/2025]
Abstract
Methyltransferase-like 3 (METTL3) is a primary RNA methyltransferase that catalyzes N6-methyladenosine (m6A) modification. The current study aims to further delineate the effect and mechanism of METTL3 in hepatocellular carcinoma (HCC). By using a murine model of hepatocellular cancer development induced via hydrodynamic tail vein injection, we showed that METTL3 enhanced HCC development. In cultured human HCC cell lines (Huh7 and PLC/PRF/5), we observed that stable knockdown of METTL3 by short hairpin RNA significantly decreased tumor cell proliferation, colony formation, and invasion, in vitro. When Huh7 and PLC/PRF/5 cells with short hairpin RNA knockdown of METTL3 were inoculated into the livers of SCID mice, we found that METTL3 knockdown significantly inhibited the growth of HCC xenograft tumors. These findings establish METTL3 as an important oncogene in HCC. Through m6A sequencing, RNA sequencing, and subsequent validation studies, we identified BMI1 and RNF2, two key components of the polycomb repressive complex 1, as direct downstream targets of METTL3-mediated m6A modification in HCC cells. Our data indicated that METTL3 catalyzed m6A modification of BMI1 and RNF2 mRNAs which led to increased mRNA stability via the m6A reader proteins IGF2BP1/2/3. Furthermore, we showed that the METTL3 inhibitor, STM2457, significantly inhibited HCC cell growth in vitro and in mice. Collectively, this study provides novel evidence that METTL3 promotes HCC development and progression through m6A modification of BMI1 and RNF2. Our findings suggest that the METTL3-m6A-BMI1/RNF2 signaling axis may represent a new therapeutic target for the treatment of HCC. Implications: The METTL3-m6A-BMI1/RNF2 signaling axis promotes HCC development and progression.
Collapse
Affiliation(s)
- Weina Chen
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jinqiang Zhang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Wenbo Ma
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Nianli Liu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
31
|
Shao Y, Ma J, Zhang S, Xu Y, Yu H. NERD-dependent m 6A modification of the nascent FLC transcript regulates flowering time in Arabidopsis. NATURE PLANTS 2025; 11:468-482. [PMID: 40087542 DOI: 10.1038/s41477-025-01945-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/19/2025] [Indexed: 03/17/2025]
Abstract
N6-methyladenosine (m6A) is the most prevalent internal modification on messenger RNA. Although recent studies have shown m6A effects on determining the fate of mRNA through modulating various aspects of plant mRNA metabolism, whether and how m6A affects gene transcription in plants remains elusive. Here we show that NEEDED FOR RDR2-INDEPENDENT DNA METHYLATION (NERD), a plant-specific protein, is an essential component of the m6A methyltransferase complex required for regulating the transcription of a central floral repressor FLOWERING LOCUS C (FLC) in Arabidopsis. NERD interacts with and stabilizes the two core methyltransferases, mRNA adenosine methylases A and B, to promote m6A modification of nascent RNA, conferring an overall negative effect on gene transcription. At the FLC locus, NERD-mediated m6A modification on the nascent transcript negatively affects H3K36me3 deposition and FLC transcription through NERD interaction with the H3K36me3 methyltransferase SET DOMAIN GROUP 8. Collectively, our findings reveal that NERD mediates the crosstalk between epitranscriptomic and epigenetic regulation of FLC to modulate flowering in Arabidopsis.
Collapse
Affiliation(s)
- Yanlin Shao
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
| | - Jinqi Ma
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Songyao Zhang
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Yifeng Xu
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Hao Yu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
32
|
Zhang H, Lu L, Yi C, Jiang T, Lu Y, Yang X, Zhong K, Zhou J, Li J, Xie G, Chen Z, Jiang Z, Asadikaram G, Peng Y, Zhou D, Wang H. BRD4 regulates m 6A of ESPL1 mRNA via interaction with ALKBH5 to modulate breast cancer progression. Acta Pharm Sin B 2025; 15:1552-1570. [PMID: 40370540 PMCID: PMC12069253 DOI: 10.1016/j.apsb.2024.12.037] [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: 06/25/2024] [Revised: 09/18/2024] [Accepted: 11/15/2024] [Indexed: 05/16/2025] Open
Abstract
The interaction between m6A-methylated RNA and chromatin modification remains largely unknown. We found that targeted inhibition of bromodomain-containing protein 4 (BRD4) by siRNA or its inhibitor (JQ1) significantly decreases mRNA m6A levels and suppresses the malignancy of breast cancer (BC) cells via increased expression of demethylase AlkB homolog 5 (ALKBH5). Mechanistically, inhibition of BRD4 increases the mRNA stability of ALKBH5 via enhanced binding between its 3' untranslated regions (3'UTRs) with RNA-binding protein RALY. Further, BRD4 serves as a scaffold for ubiquitin enzymes tripartite motif containing-21 (TRIM21) and ALKBH5, resulting in the ubiquitination and degradation of ALKBH5 protein. JQ1-increased ALKBH5 then demethylates mRNA of extra spindle pole bodies like 1 (ESPL1) and reduces binding between ESPL1 mRNA and m6A reader insulin like growth factor 2 mRNA binding protein 3 (IGF2BP3), leading to decay of ESPL1 mRNA. Animal and clinical studies confirm a critical role of BRD4/ALKBH5/ESPL1 pathway in BC progression. Further, our study sheds light on the crosstalks between histone modification and RNA methylation.
Collapse
Affiliation(s)
- Haisheng Zhang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Linlin Lu
- Institute of Medical Sciences, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Cheng Yi
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tao Jiang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- School of Life Sciences, Zhuhai College of Science and Technology, Zhuhai 519041, China
| | - Yunqing Lu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xianyuan Yang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ke Zhong
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiawang Zhou
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiexin Li
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Guoyou Xie
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhuojia Chen
- Sun Yat-sen University Cancer Center State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Zongpei Jiang
- Department of Nephrology, the Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
| | - Gholamreza Asadikaram
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Medical University Campus, Kerman 7616914115, Iran
| | - Yanxi Peng
- School of Public Health, Xiangnan University, Chenzhou 423000, China
| | - Dan Zhou
- Department of Breast Surgery, the First People's Hospital of Foshan, Foshan 528100, China
| | - Hongsheng Wang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery; State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
33
|
Ru W, Cheng J, Gao Y, Yang K, Qi A, Zhang X, Qi X, Lan X, Liu W, Huang B, Chen H. METTL3-mediated m 6A modification regulates muscle development by promoting TM4SF1 mRNA degradation in P-body via YTHDF2. Int J Biol Macromol 2025; 295:139576. [PMID: 39778834 DOI: 10.1016/j.ijbiomac.2025.139576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/06/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
N6-methyladenosine (m6A), a well-known post-transcriptional modification, is implicated in diverse cellular and physiological processes. However, much remains unknown regarding the precise role and mechanism of m6A modification on muscle development. In this study, we make observation that the levels of m6A and METTL3 are markedly elevated during the differentiation phase (DM) compared to the growth phase (GM) in both C2C12 and bovine myoblasts. Notably, deletion of METTL3 decreased m6A levels, and promoted myoblast proliferation, inhibited myoblast differentiation in vitro. By performing m6A sequencing in both GM and DM myoblast, we further identified that TM4SF1 is involved in m6A -regulated muscle development. Mechanistically, METTL3 increases m6A-modified TM4SF1 transcripts, and subsequently YTHDF2 promotes TM4SF1 mRNA degradation in P-body through liquid-liquid phase separation (LLPS). Additionally, the rescue experiments in vivo showed that overexpressing METTL3 could rescue the attenuated myogenesis induced by TM4SF1 overexpression during muscle regeneration in mice. Collectively, our findings shed light on a regulatory mechanism by which m6A modulates muscle development and raise a new model for m6A-mediated mRNA degradation within P-bodies.
Collapse
Affiliation(s)
- Wenxiu Ru
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jie Cheng
- Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yuee Gao
- Yunnan Academy of Grassland and Animal Science, Kunming 650212, China
| | - Kai Yang
- Yunnan Academy of Grassland and Animal Science, Kunming 650212, China
| | - Ao Qi
- Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaoyan Zhang
- Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xinglei Qi
- Bureau of Animal Husbandry of Biyang County, Biyang, Henan 463700, China
| | - Xianyong Lan
- Key laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, college of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wujun Liu
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China.
| | - Bizhi Huang
- Yunnan Academy of Grassland and Animal Science, Kunming 650212, China.
| | - Hong Chen
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China.
| |
Collapse
|
34
|
Quarto G, Li Greci A, Bizet M, Penning A, Primac I, Murisier F, Garcia-Martinez L, Borges RL, Gao Q, Cingaram PKR, Calonne E, Hassabi B, Hubert C, Herpoel A, Putmans P, Mies F, Martin J, Van der Linden L, Dube G, Kumar P, Soin R, Kumar A, Misra A, Lan J, Paque M, Gupta YK, Blomme A, Close P, Estève PO, Caine EA, Riching KM, Gueydan C, Daniels DL, Pradhan S, Shiekhattar R, David Y, Morey L, Jeschke J, Deplus R, Collignon E, Fuks F. Fine-tuning of gene expression through the Mettl3-Mettl14-Dnmt1 axis controls ESC differentiation. Cell 2025; 188:998-1018.e26. [PMID: 39826545 DOI: 10.1016/j.cell.2024.12.009] [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: 04/11/2023] [Revised: 10/29/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025]
Abstract
The marking of DNA, histones, and RNA is central to gene expression regulation in development and disease. Recent evidence links N6-methyladenosine (m6A), installed on RNA by the METTL3-METTL14 methyltransferase complex, to histone modifications, but the link between m6A and DNA methylation remains scarcely explored. This study shows that METTL3-METTL14 recruits the DNA methyltransferase DNMT1 to chromatin for gene-body methylation. We identify a set of genes whose expression is fine-tuned by both gene-body 5mC, which promotes transcription, and m6A, which destabilizes transcripts. We demonstrate that METTL3-METTL14-dependent 5mC and m6A are both essential for the differentiation of embryonic stem cells into embryoid bodies and that the upregulation of key differentiation genes during early differentiation depends on the dynamic balance between increased 5mC and decreased m6A. Our findings add a surprising dimension to our understanding of how epigenetics and epitranscriptomics combine to regulate gene expression and impact development and likely other biological processes.
Collapse
Affiliation(s)
- Giuseppe Quarto
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Andrea Li Greci
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Martin Bizet
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Audrey Penning
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Irina Primac
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Frédéric Murisier
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Liliana Garcia-Martinez
- Sylvester Comprehensive Cancer Center, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rodrigo L Borges
- Sylvester Comprehensive Cancer Center, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Qingzeng Gao
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Pradeep K R Cingaram
- Sylvester Comprehensive Cancer Center, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Emilie Calonne
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Bouchra Hassabi
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Céline Hubert
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Adèle Herpoel
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Pascale Putmans
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Frédérique Mies
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Jérôme Martin
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Louis Van der Linden
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Gaurav Dube
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Pankaj Kumar
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Romuald Soin
- Laboratory of Molecular Biology of the Gene, Department of Molecular Biology, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Abhay Kumar
- Greehey Children's Cancer Research Institute and Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Anurag Misra
- Greehey Children's Cancer Research Institute and Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jie Lan
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Morgane Paque
- Laboratory of Cancer Signaling, GIGA-Institute, University of Liège, Liège, Belgium; WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Yogesh K Gupta
- Greehey Children's Cancer Research Institute and Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Arnaud Blomme
- Laboratory of Cancer Signaling, GIGA-Institute, University of Liège, Liège, Belgium; WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Pierre Close
- Laboratory of Cancer Signaling, GIGA-Institute, University of Liège, Liège, Belgium; WELBIO Department, WEL Research Institute, Wavre, Belgium
| | | | | | | | - Cyril Gueydan
- Laboratory of Molecular Biology of the Gene, Department of Molecular Biology, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | | | | | - Ramin Shiekhattar
- Sylvester Comprehensive Cancer Center, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Yael David
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Lluis Morey
- Sylvester Comprehensive Cancer Center, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jana Jeschke
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Rachel Deplus
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - Evelyne Collignon
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
| | - François Fuks
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Université libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium.
| |
Collapse
|
35
|
Deng X, Fan G. Tuning up gene transcription via direct crosstalk of DNA and RNA methylation. Mol Cell 2025; 85:674-676. [PMID: 39983671 DOI: 10.1016/j.molcel.2025.01.030] [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: 01/30/2025] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/23/2025]
Abstract
In a recent Cell study,1 Quarto et al. uncovered a mechanism by which the METTL3-METTL14-DNMT1 axis fine-tunes gene expression during embryonic stem cell (ESC) differentiation. This work highlights the interplay between epigenetics and epitranscriptomics, shedding light on how methylation of DNA and RNA coordinately regulates a subset of differentiation genes.
Collapse
Affiliation(s)
- Xiaoqian Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; The Scintillon Institute, 6868 Nancy Ridge Drive, San Diego, CA 92121, USA.
| |
Collapse
|
36
|
Wang L, Weichselbaum RR, He C. N 6-methyladenosine reader YTHDF2 in cell state transition and antitumor immunity. RNA (NEW YORK, N.Y.) 2025; 31:395-401. [PMID: 39719324 PMCID: PMC11874973 DOI: 10.1261/rna.080259.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Recent studies have revealed that the YTHDF family proteins bind preferentially to the N 6-methyladenosine (m6A)-modified mRNA and regulate the functions of these RNAs in different cell types. YTHDF2, the first identified m6A reader in mammals, has garnered significant attention because of its profound effect to regulate the m6A epitranscriptome in multiple biological processes. Here, we review current knowledge on the mechanisms by which YTHDF2 exerts its functions and discuss recent advances that underscore the multifaceted role of YTHDF2 in development, stem cell expansion, and immune evasion. We also highlight potential therapeutic interventions targeting the m6A/YTHDF2 axis to improve the response to current antitumor therapies.
Collapse
Affiliation(s)
- Liangliang Wang
- The Laboratory of Microbiome and Microecological Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois 60637, USA
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois 60637, USA
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, Illinois 60637, USA
- Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
37
|
Hu X, Li J, Ding C, Jiang J, Xia W, Lu J, Su H, Zhang M, Li H, Liu JL, Lin Y, Meng Q, Huang B. Mettl3/Eed/Ythdc1 regulatory axis controls endometrial receptivity and function. Commun Biol 2025; 8:215. [PMID: 39934221 PMCID: PMC11814224 DOI: 10.1038/s42003-025-07667-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: 08/10/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
The regulatory mechanism between N6-methyladenosine (m6A) RNA methylation and histone modification in endometrial receptivity remains poorly understood. In this study, we depict that RIF induced m6A and Mettl3 level restrain, affecting H3K27me3 modification and chromatin accessibility. We show that Mettl3 deletion in the endometrium alters mRNA m6A methylation via Eed interaction. This reduces m6A recognized by Ythdc1, which recruits Eed to suppress H3K27me3 modification co-transcriptionally. The reduction of H3K27me3 disrupts chromatin accessibility and impairs transcription of genes critical for endometrial receptivity. Collectively, these results shed light on a Mettl3-Eed-m6A-Ythdc1 axis that links m6A and histone modification in regulating local chromatin state and gene expression, advancing our understanding of the epigenetic crosstalk between RNA and DNA modification in infertility disease.
Collapse
Affiliation(s)
- Xiujuan Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Jincheng Li
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Chenyue Ding
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Jing Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Wenjuan Xia
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Jiafeng Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Huapeng Su
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Ming Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Hong Li
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China
| | - Ji-Long Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| | - Yuan Lin
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Department of Maternal, Child and Adolescent Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Qingxia Meng
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China.
| | - Boxian Huang
- State Key Laboratory of Reproductive Medicine and Offspring Health (Suzhou Centre), Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, China.
| |
Collapse
|
38
|
Pilala KM, Panoutsopoulou K, Papadimitriou MA, Soureas K, Scorilas A, Avgeris M. Exploring the methyl-verse: Dynamic interplay of epigenome and m6A epitranscriptome. Mol Ther 2025; 33:447-464. [PMID: 39659016 PMCID: PMC11852398 DOI: 10.1016/j.ymthe.2024.12.003] [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: 07/23/2024] [Revised: 11/19/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024] Open
Abstract
The orchestration of dynamic epigenetic and epitranscriptomic modifications is pivotal for the fine-tuning of gene expression. However, these modifications are traditionally examined independently. Recent compelling studies have disclosed an interesting communication and interplay between m6A RNA methylation (m6A epitranscriptome) and epigenetic modifications, enabling the formation of feedback circuits and cooperative networks. Intriguingly, the interaction between m6A and DNA methylation machinery, coupled with the crosstalk between m6A RNA and histone modifications shape the transcriptional profile and translational efficiency. Moreover, m6A modifications interact also with non-coding RNAs, modulating their stability, abundance, and regulatory functions. In the light of these findings, m6A imprinting acts as a versatile checkpoint, linking epigenetic and epitranscriptomic layers toward a multilayer and time-dependent control of gene expression and cellular homeostasis. The scope of the present review is to decipher the m6A-coordinated circuits with DNA imprinting, chromatin architecture, and non-coding RNAs networks in normal physiology and carcinogenesis. Ultimately, we summarize the development of innovative CRISPR-dCas engineering platforms fused with m6A catalytic components (m6A writers or erasers) to achieve transcript-specific editing of m6A epitranscriptomes that can create new insights in modern RNA therapeutics.
Collapse
Affiliation(s)
- Katerina-Marina Pilala
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantina Panoutsopoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Alexandra Papadimitriou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Soureas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece; Laboratory of Clinical Biochemistry - Molecular Diagnostics, Second Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Margaritis Avgeris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece; Laboratory of Clinical Biochemistry - Molecular Diagnostics, Second Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, Greece.
| |
Collapse
|
39
|
Tang W, Kong X, He S, Deng J, Mao M, Peng S, Song C. WTAP Regulates SOX1 Expression to Affect the Tumorigenicity of Colorectal Cancer via an m 6A-YTHDF2-Dependent Manner. Dig Dis Sci 2025; 70:598-611. [PMID: 39681745 PMCID: PMC11839869 DOI: 10.1007/s10620-024-08780-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND Wilms tumor 1-associated protein (WTAP) plays a critical role in various cancers, including colorectal cancer (CRC). However, the biological function and molecular mechanisms of WTAP in CRC remain to be elucidated. METHODS We determined the expression of WTAP and its correlation with unfavorable prognosis of CRC using RNA-seq and the UALCAN dataset. And we investigated the effects of WTAP on CRC cells using cell proliferation assay, colony formation, cell migration and invasion, and subcutaneous xenograft experiments. We then knockdown of WTAP to identify candidate targets of WTAP. Moreover, the mRNA stability of SRY-box transcription factor 1 (SOX1) was assessed by overexpressing YTHDF2. Finally, we investigated the regulatory mechanism of WTAP in CRC by MeRIP assay, RNA pulldown, dual-luciferase reporter assay, and RIP assay. RESULTS We demonstrated that CRC patients with a high expression of WTAP have a risk prognosis. Additionally, WTAP expression can serve as a predictor of survival in CRC. WTAP promoted the proliferation and tumor growth of CRC cells. Moreover, WTAP has been recognized as the upstream regulator of SOX1. WTAP regulated the m6A modification, resulting in the post-transcriptional inhibition of SOX1. YTHDF2 plays a role in promoting mRNA degradation. Then, SOX1 can hinder the progression of CRC. Furthermore, WTAP can regulate the proliferation, migration, and invasion of CRC cells by SOX1 via an m6A-YTHDF2-dependent manner. CONCLUSION Our findings demonstrate that WTAP-mediated m6A modification facilitated the progression of CRC through the YTHDF2-SOX1 axis and could serve as a potential therapeutic targeting for CRC.
Collapse
Affiliation(s)
- Wei Tang
- Department of Oncology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha City, Hunan Province, China
| | - Xian Kong
- Department of Oncology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha City, Hunan Province, China
| | - Shoushu He
- Department of Oncology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha City, Hunan Province, China
| | - Jing Deng
- Centre for Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, No. 283, Tongzipo Road, Yuelu District, Changsha City, Hunan Province, China
| | - Min Mao
- Centre for Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, No. 283, Tongzipo Road, Yuelu District, Changsha City, Hunan Province, China
| | - Siyuan Peng
- Centre for Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, No. 283, Tongzipo Road, Yuelu District, Changsha City, Hunan Province, China
| | - Cheng Song
- Centre for Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, No. 283, Tongzipo Road, Yuelu District, Changsha City, Hunan Province, China.
| |
Collapse
|
40
|
Mangoni D, Mazzetti A, Ansaloni F, Simi A, Tartaglia GG, Pandolfini L, Gustincich S, Sanges R. From the genome's perspective: Bearing somatic retrotransposition to leverage the regulatory potential of L1 RNAs. Bioessays 2025; 47:e2400125. [PMID: 39520370 PMCID: PMC11755705 DOI: 10.1002/bies.202400125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Transposable elements (TEs) are mobile genomic elements constituting a big fraction of eukaryotic genomes. They ignite an evolutionary arms race with host genomes, which in turn evolve strategies to restrict their activity. Despite being tightly repressed, TEs display precisely regulated expression patterns during specific stages of mammalian development, suggesting potential benefits for the host. Among TEs, the long interspersed nuclear element (LINE-1 or L1) has been found to be active in neurons. This activity prompted extensive research into its possible role in cognition. So far, no specific cause-effect relationship between L1 retrotransposition and brain functions has been conclusively identified. Nevertheless, accumulating evidence suggests that interactions between L1 RNAs and RNA/DNA binding proteins encode specific messages that cells utilize to activate or repress entire transcriptional programs. We summarize recent findings highlighting the activity of L1 RNAs at the non-coding level during early embryonic and brain development. We propose a hypothesis suggesting a mutualistic relationship between L1 mRNAs and the host cell. In this scenario, cells tolerate a certain rate of retrotransposition to leverage the regulatory effects of L1s as non-coding RNAs on potentiating their mitotic potential. In turn, L1s benefit from the cell's proliferative state to increase their chance to mobilize.
Collapse
Affiliation(s)
- Damiano Mangoni
- Center for Human Technologies, Non‐Coding RNAs and RNA‐Based TherapeuticsIstituto Italiano di Tecnologia (IIT)GenovaItaly
| | - Aurora Mazzetti
- Area of NeuroscienceInternational School for Advanced Studies (SISSA)TriesteItaly
| | - Federico Ansaloni
- Center for Human Technologies, Non‐Coding RNAs and RNA‐Based TherapeuticsIstituto Italiano di Tecnologia (IIT)GenovaItaly
| | - Alessandro Simi
- Center for Human Technologies, Non‐Coding RNAs and RNA‐Based TherapeuticsIstituto Italiano di Tecnologia (IIT)GenovaItaly
| | - Gian Gaetano Tartaglia
- Center for Human Technologies, RNA Systems BiologyIstituto Italiano di Tecnologia (IIT)GenovaItaly
| | - Luca Pandolfini
- Center for Human Technologies, Non‐Coding RNAs and RNA‐Based TherapeuticsIstituto Italiano di Tecnologia (IIT)GenovaItaly
| | - Stefano Gustincich
- Center for Human Technologies, Non‐Coding RNAs and RNA‐Based TherapeuticsIstituto Italiano di Tecnologia (IIT)GenovaItaly
| | - Remo Sanges
- Center for Human Technologies, Non‐Coding RNAs and RNA‐Based TherapeuticsIstituto Italiano di Tecnologia (IIT)GenovaItaly
- Area of NeuroscienceInternational School for Advanced Studies (SISSA)TriesteItaly
| |
Collapse
|
41
|
Lee ES, Smith HW, Wang YE, Ihn SS, Scalize de Oliveira L, Kejiou NS, Liang YL, Nabeel-Shah S, Jomphe RY, Pu S, Greenblatt JF, Palazzo AF. N-6-methyladenosine (m6A) promotes the nuclear retention of mRNAs with intact 5' splice site motifs. Life Sci Alliance 2025; 8:e202403142. [PMID: 39626965 PMCID: PMC11629677 DOI: 10.26508/lsa.202403142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
In humans, misprocessed mRNAs containing intact 5' Splice Site (5'SS) motifs are nuclear retained and targeted for decay by ZFC3H1, a component of the Poly(A) Exosome Targeting complex, and U1-70K, a component of the U1 snRNP. In S. pombe, the ZFC3H1 homolog, Red1, binds to the YTH domain-containing protein Mmi1 and targets certain RNA transcripts to nuclear foci for nuclear retention and decay. Here we show that YTHDC1 and YTHDC2, two YTH domain-containing proteins that bind to N-6-methyladenosine (m6A) modified RNAs, interact with ZFC3H1 and U1-70K, and are required for the nuclear retention of mRNAs with intact 5'SS motifs. Disruption of m6A deposition inhibits both the nuclear retention of these transcripts and their accumulation in YTHDC1-enriched foci that are adjacent to nuclear speckles. Endogenous RNAs with intact 5'SS motifs, such as intronic poly-adenylated transcripts, tend to be m6A-modified at low levels. Thus, the m6A modification acts on a conserved quality control mechanism that targets misprocessed mRNAs for nuclear retention and decay.
Collapse
Affiliation(s)
- Eliza S Lee
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Harrison W Smith
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Yifan E Wang
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Sean Sj Ihn
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | | | - Nevraj S Kejiou
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Yijing L Liang
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Canada
| | - Syed Nabeel-Shah
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Robert Y Jomphe
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Cell Biology Program, Hospital for Sick Children, Toronto, Canada
| | - Shuye Pu
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Jack F Greenblatt
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | | |
Collapse
|
42
|
Su Z, Lan J, Wang Y, Ma N, Yang J, Liang D, Zeng H, Yang M. Lactylation-driven ALKBH5 diminishes macrophage NLRP3 inflammasome activation in patients with G6PT deficiency. J Allergy Clin Immunol 2025:S0091-6749(25)00117-4. [PMID: 39900266 DOI: 10.1016/j.jaci.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Neutropenia represents an important clinical problem in patients with glycogen storage disease type Ib, characterized by genetic deficiency in glucose-6-phosphate translocase (G6PT/SLC37A4). However, the role of G6PT in macrophages has not been elucidated. OBJECTIVE We sought to investigate the function of G6PT in macrophage inflammation. METHODS Functional assays (including immunoblotting, real-time quantitative PCR, flow cytometry, immunofluorescence staining, and enzyme-linked immunosorbent assay) and RNA sequencing were performed. RESULTS We find that macrophages from patients deficient in G6PT exhibited diminished NLRP3 inflammasome activation. Mechanistically, deficiency of G6PT promotes glycolysis and lactate production in macrophages. Lactate accumulation potently induces ALKBH5 upregulation via H3K18 lactylation. ALKBH5 decreases m6A modification on NLRP3 messenger RNA, attenuating its transcript stability and thus inhibiting inflammasome activation. Further, treating G6PT-deficient macrophages with an inhibitor of the lactate dehydrogenase to lower their lactate levels restores NLRP3 inflammasome activation and rescues bacterial handling defect. CONCLUSION These findings reveal a previously unknown pathogenic mechanism of lactylation-driven defective NLRP3 inflammasome signaling and subsequent impaired antimicrobial activity as driving factors in these inflammatory disorders, indicating that glycolysis/lactate/histone lactylation cascade may be a potential therapeutic target for glycogen storage disease type Ib.
Collapse
Affiliation(s)
- Zexiong Su
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China; Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiaoli Lan
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying Wang
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ni Ma
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jing Yang
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Danxia Liang
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hanshi Zeng
- Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Min Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China; Department of Pediatrics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
43
|
Chen W, Liu Y, Liu J, Chen Y, Wang X. Acute exercise promotes WAT browning by remodeling mRNA m 6A methylation. Life Sci 2025; 361:123269. [PMID: 39581460 DOI: 10.1016/j.lfs.2024.123269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/30/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
AIMS Regular exercise promotes the beiging and metabolic adaptations of white adipose tissue (WAT) through the cumulative transcriptional responses that occur after each exercise session. However, the effects of a single bout of acute exercise and the role of N6-methyladenosine (m6A) in these adaptations remain unclear. We aim to investigate this further. MATERIALS AND METHODS We constructed mouse models for chronic (8 weeks of running) and acute (single 1-hour run) exercise to study the effects on white adipose tissue (WAT) metabolism and beiging through metabolic phenotyping and transcriptome sequencing. Additionally, we explored the impact of acute exercise on WAT m6A modification and target genes, combining m6A regulators with cell models to elucidate the role of m6A in WAT exercise adaptation. KEY FINDINGS Here, we reveal that upregulated m6A modification after acute exercise induces the formation of glycolytic beige fat (g-beige fat) in WAT. Mechanistically, the metabolite β-hydroxybutyrate (BHBA) secreted after acute exercise upregulates m6A modification in WAT. This enhances m6A-dependent translation of the histone acetyltransferase CREBBP, promoting the transcription of key beiging genes by increasing chromatin accessibility. Pharmacologically elevating circulating BHBA mimics the metabolic response induced by acute exercise, upregulating m6A modification and its downstream signals. Additionally, BHBA exhibits long-term effects, improving metabolic homeostasis in obesity by promoting thermogenesis in WAT. SIGNIFICANCE Our results reveal the role of metabolites in WAT metabolic adaptation through m6A-mediated chromatin accessibility after acute exercise, providing a novel therapeutic target for regulating WAT metabolism from a nutritional epigenetics perspective.
Collapse
Affiliation(s)
- Wei Chen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Youhua Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Jiaqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Yushi Chen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
| | - Xinxia Wang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China.
| |
Collapse
|
44
|
Zhang H, Yi C, Li J, Lu Y, Wang H, Tao L, Zhou J, Tan Y, Li J, Chen Z, Asadikaram G, Cao J, Peng J, Li W, He J, Wang H. N6-methyladenosine RNA modification regulates the transcription of SLC7A11 through KDM6B and GATA3 to modulate ferroptosis. J Biomed Sci 2025; 32:8. [PMID: 39800682 PMCID: PMC11726933 DOI: 10.1186/s12929-024-01100-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: 07/22/2024] [Accepted: 11/12/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Recent studies indicate that N6-methyladenosine (m6A) RNA modification may regulate ferroptosis in cancer cells, while its molecular mechanisms require further investigation. METHODS Liquid Chromatography-Tandem Mass Spectrometry (HPLC/MS/MS) was used to detect changes in m6A levels in cells. Transmission electron microscopy and flow cytometry were used to detect mitochondrial reactive oxygen species (ROS). RNA sequencing (RNA-seq) was employed to analyze the factors regulating ferroptosis. Chromatin immunoprecipitation (ChIP) was used to assess the binding of regulatory factors to the SLC7A11 promoter, and a Dual-Luciferase reporter assay measured promoter activity of SLC7A11. The dm6ACRISPR system was utilized for the demethylation of specific transcripts. The Cancer Genome Atlas Program (TCGA) database and immunohistochemistry validated the role of the METTL3/SLC7A11 axis in cancer progression. RESULTS The m6A methyltransferase METTL3 was upregulated during cancer cell ferroptosis and facilitated erastin-induced ferroptosis by enhancing mitochondrial ROS. Mechanistic studies showed that METTL3 negatively regulated the transcription and promoter activity of SLC7A11. Specifically, METTL3 induced H3K27 trimethylation of the SLC7A11 promoter by suppressing the mRNA stability of H3K27 demethylases KDM6B. Furthermore, METTL3 suppressed the expression of GATA3, which regulated SLC7A11 transcription by binding to the putative site at - 597 to - 590 of the SLC7A11 promoter. METTL3 decreased the precursor mRNA stability of GATA3 through m6A/YTHDF2-dependent recruitment of the 3'-5' exoribonuclease Dis3L2. Targeted demethylation of KDM6B and GATA3 m6A using the dm6ACRISPR system significantly increased the expression of SLC7A11. Moreover, the transcription factor YY1 was responsible for erastin-induced upregulation of METTL3 by binding to its promoter-proximal site. In vivo and clinical data supported the positive roles of the METTL3/SLC7A11 axis in tumor growth and progression. CONCLUSIONS METTL3 regulated the transcription of SLC7A11 through GATA3 and KDM6B to modulate ferroptosis in an m6A-dependent manner. This study provides a novel potential strategy and experimental support for the future treatment of cancer.
Collapse
Affiliation(s)
- Haisheng Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Cheng Yi
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jianing Li
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yunqing Lu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Haoran Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Lijun Tao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jiawang Zhou
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yonghuang Tan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Jiexin Li
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Zhuojia Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Gholamreza Asadikaram
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Medical University Campus, Kerman, Iran
| | - Jie Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, 510180, China
| | - Jianxin Peng
- Department of Hepatobiliary Surgery, Guangdong Province Traditional Chinese Medical Hospital, Guangzhou, 510120, China
| | - Wanglin Li
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, 510180, China.
- Huadu District People's Hospital of Guangzhou, Guangzhou, 510800, China.
| | - Junming He
- Department of Hepatobiliary Surgery, Guangdong Province Traditional Chinese Medical Hospital, Guangzhou, 510120, China.
| | - Hongsheng Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| |
Collapse
|
45
|
Yan R, Qi M, Zhang P, Shen B, Yin J, Chen C, Tian S, Chen L, Huang X, Wang H, Gao S, Wu Y, Gao Y. Core factor of NEXT complex, ZCCHC8, governs the silencing of LINE1 during spermatogenesis. Natl Sci Rev 2025; 12:nwae407. [PMID: 39758125 PMCID: PMC11697976 DOI: 10.1093/nsr/nwae407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 01/07/2025] Open
Abstract
The overactivation of transposable elements (TEs) is a significant threat to male reproduction, particularly during the delicate process of spermatogenesis. Here, we report that zinc finger protein ZCCHC8-a key component of the nuclear exosome targeting (NEXT) complex that is involved in ribonucleic acid (RNA) surveillance-is required for TE silencing during spermatogenesis. Loss of ZCCHC8 results in delayed meiotic progression and reduced production of round spermatids (RS). We observed that young long-interspersed nuclear element (LINE1, L1) subfamilies that are targeted by ZCCHC8 were upregulated in both spermatogonial stem cells (SSC) and pachytene spermatocytes (PS) of Zcchc8 null testes. Further study found that a reduced H3K9me3 modification in SSC and elevated H3 lysine 4 trimethylation in the PS of Zcchc8 KO mice occurred upon young L1, especially L1Md_A, which may have contributed to impairment of the chromatin condensation from PS to RS during spermatogenesis. This study highlights the crucial role of RNA surveillance-mediated chromatin repression by the NEXT complex during spermatogenesis.
Collapse
Affiliation(s)
- Rushuang Yan
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Meijie Qi
- Reproductive and Genetic Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | | | - Bin Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Jiqing Yin
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Chuan Chen
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Silin Tian
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Lin Chen
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | | | - Hong Wang
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Shaorong Gao
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, China
| | - You Wu
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, China
| | - Yawei Gao
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, China
| |
Collapse
|
46
|
Liu Z, Li D, Deng M, Zhou L, Wang J, Liu L, Mao W, Lu H, Xu X, Wang F, Wan Y. METTL3 improves the development of somatic cell nuclear transfer embryos through AURKB and H3S10ph in goats. Int J Biol Macromol 2025; 286:138546. [PMID: 39653227 DOI: 10.1016/j.ijbiomac.2024.138546] [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: 07/02/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Developmental abnormalities are more common in somatic cell nuclear transfer (SCNT) embryos due to epigenetic barriers that occur during the maternal-to-zygotic transition (MZT). N6-methyladenosine (m6A) is an RNA epigenetic modification that plays a significant role in numerous biological processes. However, the relationship between m6A and SCNT embryonic development is largely unexplored. In the present study, we found that the low expression of m6A methyltransferase-like 3 (METTL3) was associated with developmental arrest before zygotic genome activation (ZGA) in goat SCNT embryos and that karyokinesis defects were evident during their development. Notably, we demonstrated that METTL3 overexpression rescued the karyokinesis abnormalities, enhanced embryonic development and elevated the blastocyst formation rate. Further experiments revealed that METTL3 could mitigate the defects of maternal mRNA degradation, enhance the translation of Aurora kinase B (AURKB) and increase the phosphorylation of serine 10 on histone H3 (H3S10ph) to ensure the normal karyokinesis in SCNT embryos before ZGA in goats. Overall, our study highlights the essential role of METTL3 in enhancing the development of goat SCNT embryos. These findings indicate that METTL3 is critical for optimal SCNT efficiency and advance our understanding of m6A's role in embryonic development.
Collapse
Affiliation(s)
- Zifei Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University; Changzhou Key Laboratory of Maternal and Child Health Medicine, Changzhou 213003, Jiangsu, China
| | - Dongxu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Mingtian Deng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Lei Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jingang Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Liang Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Weijia Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Honghui Lu
- Department of Animal Husbandry & Veterinary, Haimen distinct, Nantong 226000, China
| | - Xinsong Xu
- Department of Animal Husbandry & Veterinary, Haimen distinct, Nantong 226000, China
| | - Feng Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yongjie Wan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
47
|
Wang W, Jin Y, Xie Z, He M, Li J, Wang Z, Ma S, Zhang W, Tong J. When animal viruses meet N 6-methyladenosine (m 6A) modifications: for better or worse? Vet Res 2024; 55:171. [PMID: 39695760 DOI: 10.1186/s13567-024-01424-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/07/2024] [Indexed: 12/20/2024] Open
Abstract
N6-methyladenosine (m6A) is a prevalent and dynamic RNA modification, critical in regulating gene expression. Recent research has shed light on its significance in the life cycle of viruses, especially animal viruses. Depending on the context, these modifications can either enhance or inhibit the replication of viruses. However, research on m6A modifications in animal virus genomes and the impact of viral infection on the host cell m6A landscape has been hindered due to the difficulty of detecting m6A sites at a single-nucleotide level. This article summarises the methods for detecting m6A in RNA. It then discusses the progress of research into m6A modification within animal viruses' infections, such as influenza A virus, porcine epidemic diarrhoea virus, porcine reproductive, and respiratory syndrome virus. Finally, the review explores how m6A modification affects the following three aspects of the replication of animal RNA viruses: the regulation of viral genomic RNA function, the alteration of the m6A landscape in cells after viral infection, and the modulation of antiviral immunity through m6A modification. Research on m6A modifications in viral RNA sheds light on virus-host interactions at a molecular level. Understanding the impact of m6A on viral replication can help identify new targets for antiviral drug development and may uncover novel regulatory pathways that could potentially enhance antiviral immune responses.
Collapse
Affiliation(s)
- Wenjing Wang
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Yufei Jin
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Ziyun Xie
- College of Veterinary Medicine, Hebei Agriculture University, Baoding, 071001, China
| | - Mei He
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Jing Li
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Zihan Wang
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Saiya Ma
- College of Life Sciences, Hebei University, Baoding, 071002, China
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China
| | - Wuchao Zhang
- College of Veterinary Medicine, Hebei Agriculture University, Baoding, 071001, China.
| | - Jie Tong
- College of Life Sciences, Hebei University, Baoding, 071002, China.
- School of Life Sciences and Green Development, Hebei University, Baoding, 071002, China.
| |
Collapse
|
48
|
Zhao Y, Li J, Dian M, Bie Y, Peng Z, Zhou Y, Zhou B, Hao W, Wang X. Role of N6-methyladenosine methylation in nasopharyngeal carcinoma: current insights and future prospective. Cell Death Discov 2024; 10:490. [PMID: 39695216 DOI: 10.1038/s41420-024-02266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a distinct type of head and neck squamous cell carcinoma prevalent in Southern China, Southeast Asia, and North Africa. Despite advances in treatment options, the prognosis for advanced NPC remains poor, underscoring the urgent need to explore its underlying mechanisms and develop novel therapeutic strategies. Epigenetic alterations have been shown to play a key role in NPC progression. Recent studies indicate that dysregulation of RNA modifications in NPC specifically affects tumor-related transcripts, influencing various oncogenic processes. This review provides a comprehensive overview of altered RNA modifications and their regulators in NPC, with a focus on m6A and its regulatory mechanisms. We discuss how m6A RNA modification influences gene expression and affects NPC initiation and progression at the molecular level, analyzing its impact on cancer-related biological functions. Understanding these modifications could reveal new biomarkers and therapeutic targets for NPC, offering promising directions for future research and precision medicine.
Collapse
Affiliation(s)
- YaYan Zhao
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie Li
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - MeiJuan Dian
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - YaNan Bie
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - ZhiTao Peng
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - BingQian Zhou
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - WeiChao Hao
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - XiCheng Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- Cancer Research Institute of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
49
|
Xia R, Yin X, Huang J, Chen K, Ma J, Wei Z, Su J, Blake N, Rigden DJ, Meng J, Song B. Interpretable deep cross networks unveiled common signatures of dysregulated epitranscriptomes across 12 cancer types. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102376. [PMID: 39618823 PMCID: PMC11605186 DOI: 10.1016/j.omtn.2024.102376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 10/25/2024] [Indexed: 01/12/2025]
Abstract
Cancer is a complex and multifaceted group of diseases characterized by uncontrolled cell growth that leads to the formation of malignant tumors. Recent studies suggest that N6-methyladenosine (m6A) RNA methylation plays pivotal roles in cancer pathology by influencing various cellular processes. However, the degree to which these mechanisms are shared across different cancer types remains unclear. In this study, we analyze an expansive array of 167 m6A epitranscriptome profiles covering 12 distinct cancer types and their originating normal tissues. We trained 12 distinct, cancer type-specific interpretable deep cross network models, which successfully distinguish between specific pairs of normal and cancer m6A contexts using integrated information from both the sequences and curated genomic knowledge. Interestingly, cross-cancer type testing indicated the existence of shared genomic patterns across various cancers at the epitranscriptome level. A pan-cancer model was subsequently developed to identify these shared patterns that could not be observed in a single cancer type. Our analysis uncovered, for the first time, a common epitranscriptome signature shared across multiple cancer types, particularly associated with RNA hybridization process and aberrant splicing. This highlights the importance of a comprehensive understanding of the pan-cancer epitranscriptome and holding potential implications in the development of RNA methylation-based therapeutics for various cancers.
Collapse
Affiliation(s)
- Rong Xia
- Department of Public Health, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Department of Biological Sciences, School of Science, Suzhou Key Laboratory of Cancer Biology and Chronic Disease, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
- School of AI and Advanced Computing, XJTLU Entrepreneur College (Taicang), Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Xiangyu Yin
- Department of Biological Sciences, School of Science, Suzhou Key Laboratory of Cancer Biology and Chronic Disease, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L7 8TX Liverpool, UK
| | - Jiaming Huang
- Department of Biological Sciences, School of Science, Suzhou Key Laboratory of Cancer Biology and Chronic Disease, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Kunqi Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350004, China
| | - Jiongming Ma
- Department of Biological Sciences, School of Science, Suzhou Key Laboratory of Cancer Biology and Chronic Disease, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Zhen Wei
- Department of Biological Sciences, School of Science, Suzhou Key Laboratory of Cancer Biology and Chronic Disease, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, L7 8TX Liverpool, UK
| | - Jionglong Su
- School of AI and Advanced Computing, XJTLU Entrepreneur College (Taicang), Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Neil Blake
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L7 8TX Liverpool, UK
| | - Daniel J. Rigden
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L7 8TX Liverpool, UK
| | - Jia Meng
- Institute of Biomedical Research, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei Provincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
- Department of Biological Sciences, School of Science, Suzhou Key Laboratory of Cancer Biology and Chronic Disease, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L7 8TX Liverpool, UK
| | - Bowen Song
- Department of Public Health, School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
50
|
Chen Y, Liang R, Li Y, Jiang L, Ma D, Luo Q, Song G. Chromatin accessibility: biological functions, molecular mechanisms and therapeutic application. Signal Transduct Target Ther 2024; 9:340. [PMID: 39627201 PMCID: PMC11615378 DOI: 10.1038/s41392-024-02030-9] [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: 03/11/2024] [Revised: 08/04/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024] Open
Abstract
The dynamic regulation of chromatin accessibility is one of the prominent characteristics of eukaryotic genome. The inaccessible regions are mainly located in heterochromatin, which is multilevel compressed and access restricted. The remaining accessible loci are generally located in the euchromatin, which have less nucleosome occupancy and higher regulatory activity. The opening of chromatin is the most important prerequisite for DNA transcription, replication, and damage repair, which is regulated by genetic, epigenetic, environmental, and other factors, playing a vital role in multiple biological progresses. Currently, based on the susceptibility difference of occupied or free DNA to enzymatic cleavage, solubility, methylation, and transposition, there are many methods to detect chromatin accessibility both in bulk and single-cell level. Through combining with high-throughput sequencing, the genome-wide chromatin accessibility landscape of many tissues and cells types also have been constructed. The chromatin accessibility feature is distinct in different tissues and biological states. Research on the regulation network of chromatin accessibility is crucial for uncovering the secret of various biological processes. In this review, we comprehensively introduced the major functions and mechanisms of chromatin accessibility variation in different physiological and pathological processes, meanwhile, the targeted therapies based on chromatin dynamics regulation are also summarized.
Collapse
Affiliation(s)
- Yang Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Rui Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Yong Li
- Hepatobiliary Pancreatic Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lingli Jiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Di Ma
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China.
| |
Collapse
|