1
|
Fan D, Shang Y, Cong Y, Jiao Y, Li N, Zhao H. Reciprocal regulation between m6 A modifications and non-coding RNAs: emerging roles in cancer therapeutic resistance. Discov Oncol 2025; 16:920. [PMID: 40413672 DOI: 10.1007/s12672-025-02641-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 05/09/2025] [Indexed: 05/27/2025] Open
Abstract
In recent years, the interplay between N6-methyladenosine (m6A) modifications and non-coding RNAs (ncRNAs) has emerged as a pivotal research area, owing to their crucial involvement in the pathophysiological mechanisms underlying various diseases. A significant hurdle in cancer therapy is therapeutic resistance, which frequently contributes to adverse patient outcomes. Recent investigations have underscored the vital role that interactions between m6A modifications and ncRNAs play in mediating cancer therapeutic resistance via the MAPK, PI3K/Akt/mTOR, Wnt/β-catenin, HIPPO, and NF-κB pathways. This review elucidates how these interactions drive tumor therapeutic resistance by modulating these pathways. By dissecting the regulatory dynamics between m6A and ncRNAs in the context of cancer therapeutic resistance, this review aims to deepen the understanding of m6A-ncRNA interaction in cancer therapeutic resistance and identify potential therapeutic targets to improve cancer treatment efficacy.
Collapse
Affiliation(s)
- Dan Fan
- Nanshan Class, The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Yan Shang
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China
| | - Yating Cong
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China
| | - Yanlin Jiao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China
| | - Na Li
- The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Hailong Zhao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
2
|
Han M, Fu ML, Zhu Y, Choi AA, Li E, Bezney J, Cai S, Miles L, Ma Y, Qi LS. Programmable control of spatial transcriptome in live cells and neurons. Nature 2025:10.1038/s41586-025-09020-z. [PMID: 40399675 DOI: 10.1038/s41586-025-09020-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/14/2025] [Indexed: 05/23/2025]
Abstract
Spatial RNA organization has a pivotal role in diverse cellular processes and diseases1-4. However, functional implications of the spatial transcriptome remain largely unexplored due to limited technologies for perturbing endogenous RNA within specific subcellular regions1,5. Here we present CRISPR-mediated transcriptome organization (CRISPR-TO), a system that harnesses RNA-guided, nuclease-dead dCas13 for programmable control of endogenous RNA localization in live cells. CRISPR-TO enables targeted localization of endogenous RNAs to diverse subcellular compartments, including the outer mitochondrial membrane, p-bodies, stress granules, telomeres and nuclear stress bodies, across various cell types. It allows for inducible and reversible bidirectional RNA transport along microtubules via motor proteins, facilitating real-time manipulation and monitoring of RNA localization dynamics in living cells. In primary cortical neurons, we demonstrate that repositioned mRNAs undergo local translation along neurites and at neurite tips, and co-transport with ribosomes, with β-actin mRNA localization enhancing the formation of dynamic filopodial protrusions and inhibiting axonal regeneration. CRISPR-TO-enabled screening in primary neurons identifies Stmn2 mRNA localization as a driver of neurite outgrowth. By enabling large-scale perturbation of the spatial transcriptome, CRISPR-TO bridges a critical gap left by sequencing and imaging technologies, offering a versatile platform for high-throughput functional interrogation of RNA localization in living cells and organisms.
Collapse
Affiliation(s)
- Mengting Han
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Maylin L Fu
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Yanyu Zhu
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Alexander A Choi
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Emmy Li
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Jon Bezney
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Sa Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Leann Miles
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Yitong Ma
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Lei S Qi
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Butterfield GL, Reisman SJ, Iglesias N, Gersbach CA. Gene regulation technologies for gene and cell therapy. Mol Ther 2025; 33:2104-2122. [PMID: 40195118 DOI: 10.1016/j.ymthe.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025] Open
Abstract
Gene therapy stands at the forefront of medical innovation, offering unique potential to treat the underlying causes of genetic disorders and broadly enable regenerative medicine. However, unregulated production of therapeutic genes can lead to decreased clinical utility due to various complications. Thus, many technologies for controlled gene expression are under development, including regulated transgenes, modulation of endogenous genes to leverage native biological regulation, mapping and repurposing of transcriptional regulatory networks, and engineered systems that dynamically react to cell state changes. Transformative therapies enabled by advances in tissue-specific promoters, inducible systems, and targeted delivery have already entered clinical testing and demonstrated significantly improved specificity and efficacy. This review highlights next-generation technologies under development to expand the reach of gene therapies by enabling precise modulation of gene expression. These technologies, including epigenome editing, antisense oligonucleotides, RNA editing, transcription factor-mediated reprogramming, and synthetic genetic circuits, have the potential to provide powerful control over cellular functions. Despite these remarkable achievements, challenges remain in optimizing delivery, minimizing off-target effects, and addressing regulatory hurdles. However, the ongoing integration of biological insights with engineering innovations promises to expand the potential for gene therapy, offering hope for treating not only rare genetic disorders but also complex multifactorial diseases.
Collapse
Affiliation(s)
- Gabriel L Butterfield
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Samuel J Reisman
- Department of Cell Biology, Duke University, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Nahid Iglesias
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA; Department of Cell Biology, Duke University, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
4
|
Briend M, Mathieu P. RNA epigenetic modifications: a new field of research in calcific aortic valve disease. Cardiovasc Res 2025; 121:8-9. [PMID: 39775749 DOI: 10.1093/cvr/cvae256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Affiliation(s)
- Mewen Briend
- Laboratory Pathobiology of Cardiovascular Diseases, Quebec Heart and Lung Institute, Laval University, 2725 Chemin Ste-Foy, Quebec, Canada G1V-4G5
| | - Patrick Mathieu
- Laboratory Pathobiology of Cardiovascular Diseases, Quebec Heart and Lung Institute, Laval University, 2725 Chemin Ste-Foy, Quebec, Canada G1V-4G5
- Department of Surgery, Laval University, 2325 Rue Université, Quebec, Canada G1V 0A6
| |
Collapse
|
5
|
Liu X, Qi Q, Xiong W, Zhang Y, Shen W, Xu X, Zhao Y, Li M, Zhou E, Tian T, Zhou X. Tailoring and reversing m6A editing with sequential RNA bioorthogonal chemistry. Nucleic Acids Res 2025; 53:gkaf283. [PMID: 40219967 PMCID: PMC11992675 DOI: 10.1093/nar/gkaf283] [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: 04/25/2024] [Revised: 03/23/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Many existing methods for post-transcriptional RNA modification rely on a single-step approach, limiting the ability to reversibly control m6A methylation at specific sites. Here, we address this challenge by developing a multi-step system that builds on the concept of sequential RNA bioorthogonal chemistry. Our strategy uses an azide-based reagent (NAI-N3) capable of both cleavage and ligation reactions, thereby allowing iterative and reversible modifications of RNA in living cells. By applying this approach in CRISPR (clustered regularly interspaced short palindromic repeats)-based frameworks, we demonstrate tailored editing of m6A marks at targeted RNA sites, overcoming the one-way restriction of conventional bioorthogonal methods. This sequential protocol not only broadens the scope for fine-tuned RNA regulation but also provides a versatile platform for exploring dynamic m6A function in genetic and epigenetic research.
Collapse
Affiliation(s)
- Xingyu Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Qianqian Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Wei Xiong
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Yuanyuan Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Wei Shen
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Xinyan Xu
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Yunting Zhao
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Ming Li
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Enyi Zhou
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Tian Tian
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| | - Xiang Zhou
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Hubei Province Key Laboratory of Allergy and Immunology, The Institute of Molecular Medicine, Wuhan University People's Hospital, Wuhan University, Wuhan 430072, Hubei, China
| |
Collapse
|
6
|
Cheng ECK, Lam JKC, Kwon SC. Cytosolic CRISPR RNAs for efficient application of RNA-targeting CRISPR-Cas systems. EMBO Rep 2025; 26:1891-1912. [PMID: 40011676 PMCID: PMC11976971 DOI: 10.1038/s44319-025-00399-4] [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: 09/16/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
Clustered regularly interspaced short palindromic repeats/CRISPR-associated protein (CRISPR/Cas) technologies have evolved rapidly over the past decade with the continuous discovery of new Cas systems. In particular, RNA-targeting CRISPR-Cas13 proteins are promising single-effector systems to regulate target mRNAs without altering genomic DNA, yet the current Cas13 systems are restrained by suboptimal efficiencies. Here, we show that U1 promoter-driven CRISPR RNAs (crRNAs) increase the efficiency of various applications, including RNA knockdown and editing, without modifying the Cas13 protein effector. We confirm that U1-driven crRNAs are exported into the cytoplasm, while conventional U6 promoter-driven crRNAs are mostly confined to the nucleus. Furthermore, we reveal that the end positions of crRNAs expressed by the U1 promoter are consistent regardless of guide sequences and lengths. We also demonstrate that U1-driven crRNAs, but not U6-driven crRNAs, can efficiently repress the translation of target genes in combination with catalytically inactive Cas13 proteins. Finally, we show that U1-driven crRNAs can counteract the inhibitory effect of miRNAs. Our simple and effective engineering enables unprecedented cytosolic RNA-targeting applications.
Collapse
Affiliation(s)
- Ezra C K Cheng
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Joe K C Lam
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - S Chul Kwon
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
7
|
Shen L, Yu H. RNA m 6A modification meets plant hormones. NATURE PLANTS 2025; 11:686-695. [PMID: 40155697 DOI: 10.1038/s41477-025-01947-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/19/2025] [Indexed: 04/01/2025]
Abstract
Plant hormones are essential signalling molecules that control and coordinate diverse physiological processes in plant development and adaptation to ever-fluctuating environments. This hormonal regulation of plant development and environmental responses has recently been shown to extensively involve the most widespread RNA modification, N6-methyladenosine (m6A). Here we discuss the current understanding of the crosstalk between m6A and plant hormones, focusing on their reciprocal regulation, where hormonal signals induce m6A reprogramming and m6A affects hormone biosynthesis and signalling cascades. We also highlight new insights into how m6A contributes to the hormonal control of plant development and stress responses. Furthermore, we discuss future prospects for unveiling the regulatory networks that orchestrate epitranscriptome-hormone interactions and harnessing the related knowledge accrued to enhance crop productivity and resilience in changing environments.
Collapse
Affiliation(s)
- Lisha Shen
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.
| | - Hao Yu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
8
|
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
|
9
|
Tang H, Han S, Jie Y, Jiang X, Zhang Y, Peng J, Wang F, Li X, Zhou X, Jiang W, Weng X. Enhanced or reversible RNA N6-methyladenosine editing by red/far-red light induction. Nucleic Acids Res 2025; 53:gkaf181. [PMID: 40103228 PMCID: PMC11915503 DOI: 10.1093/nar/gkaf181] [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: 06/03/2024] [Revised: 01/03/2025] [Accepted: 02/25/2025] [Indexed: 03/20/2025] Open
Abstract
The RNA N6-methyladenosine (m6A) modification is a critical regulator of various biological processes, but precise and dynamic control of m6A remains a challenge. In this work, we present a red/far-red light-inducible m6A editing system that enables efficient and reversible modulation of m6A levels with minimal off-target effects. By engineering the CRISPR dCas13 protein and sgRNA with two pairs of light-inducible heterodimerizing proteins, ΔphyA/FHY1 and Bphp1/PspR2, we achieved targeted recruitment of m6A effectors. This system significantly enhances m6A writing efficiency and allows dynamic regulation of m6A deposition and removal on specific transcripts, such as SOX2 and ACTB. Notably, reversible m6A editing was achieved through cyclic modulation at a single target site, demonstrating the ability to influence mRNA expression and modulate the differentiation state of human embryonic stem cells. This optogenetic platform offers a precise, versatile tool for cyclic and reversible m6A regulation, with broad implications for understanding RNA biology and its potential applications in research and medicine.
Collapse
Affiliation(s)
- Heng Tang
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Shaoqin Han
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan 430072, P.R. China
| | - Yang Jie
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China
| | - Xin Jiang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan 430072, P.R. China
| | - Yi Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Junran Peng
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan 430072, P.R. China
| | - Fang Wang
- Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiang Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Xiang Zhou
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan 430072, P.R. China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Xiaocheng Weng
- Department of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers of Ministry of Education, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan 430072, P.R. China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| |
Collapse
|
10
|
Kha T, Zhao Y, Zhu R. Site-Selective Modification and Labeling of Native RNA. Chemistry 2025; 31:e202404244. [PMID: 39865772 PMCID: PMC11855268 DOI: 10.1002/chem.202404244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 01/28/2025]
Abstract
Ribonucleic acid (RNA) plays a pivotal role in regulating biological processes within living systems, with modified nucleosides serving as critical modulators of various aspects of biological functions. Therefore, the development of efficient methodologies for late-stage, site-selective RNA modification is of considerable interest, as it facilitates the functional exploration of RNA chemical modifications and their implications for therapeutic applications. Precise RNA modification holds significant promise for the treatment of genetic diseases by enabling the correction of mutated nucleobases to their wild-type forms. Additionally, the site-selective incorporation of synthetic labeling groups into RNA provides invaluable tools for structural and functional studies, thereby uncovering previously hidden dimensions of RNA's role in biological systems. In this review, we provide a comprehensive overview of three principal approaches to site-selective, late-stage RNA modifications: enzyme-mediated strategies, catalytic nucleic acid-based techniques, and chemical methodologies. These approaches predominantly target the nucleobase or the 2'-hydroxyl (2'-OH) group of RNA nucleosides. We evaluate the advantages and limitations of each strategy and discuss future directions for advancing this field of research.
Collapse
Affiliation(s)
- Tuan‐Khoa Kha
- Department of ChemistryNational University of SingaporeSingapore117544
| | - Yiran Zhao
- Department of ChemistryNational University of SingaporeSingapore117544
| | - Ru‐Yi Zhu
- Department of ChemistryNational University of SingaporeSingapore117544
| |
Collapse
|
11
|
Li P, Fang X, Huang D. Exploring m6A modifications in gastric cancer: from molecular mechanisms to clinical applications. Eur J Med Res 2025; 30:98. [PMID: 39940056 PMCID: PMC11823136 DOI: 10.1186/s40001-025-02353-5] [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] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
The significance of m6A modifications in several biological processes has been increasingly recognized, particularly in the context of cancer. For instance, m6A modifications in gastric cancer (GC) have been significantly implicated in tumor progression, metastasis, and treatment resistance. GC is characterized by the differential expression of m6A regulators. High expression writers such as METTL3 and WTAP are associated with poor prognosis and aggressive clinical features. Conversely, low expression of METTL14 is linked to worse clinical outcomes, whereas elevated levels of demethylases, such as FTO and ALKBH5, correlate with better survival rates. These m6A regulators influence several cellular biological functions, including proliferation, invasion, migration, glycolysis, and chemotherapy resistance, thereby affecting tumor growth and therapeutic outcomes. The assessment of m6A modification patterns and the expression profiles of m6A-related genes hold substantial potential for improving the clinical diagnosis and treatment of GC. In this review, we provide an updated and comprehensive summary of the role of m6A modifications in GC, emphasizing their molecular mechanisms, clinical significance, and translational applications in developing novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Penghui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China.
| | - Xiangjie Fang
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453100, Henan, China
| | - Di Huang
- Department of Child Health Care, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
12
|
Ge L, Pan F, Jia M, Pott DM, He H, Shan H, Lozano-Durán R, Wang A, Zhou X, Li F. RNA modifications in plant biotic interactions. PLANT COMMUNICATIONS 2025; 6:101232. [PMID: 39722456 PMCID: PMC11897454 DOI: 10.1016/j.xplc.2024.101232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
The chemical modifications of DNA and proteins are powerful mechanisms for regulating molecular and biological functions, influencing a wide array of signaling pathways in eukaryotes. Recent advancements in epitranscriptomics have shown that RNA modifications play crucial roles in diverse biological processes. Since their discovery in the 1970s, scientists have sought to decipher, identify, and elucidate the functions of these modifications across biological systems. Over the past decade, mounting evidence has demonstrated the importance of RNA modification pathways in plants, prompting significant efforts to decipher their physiological relevance. With the advent of high-resolution mapping techniques for RNA modifications and the gradual uncovering of their biological roles, our understanding of this additional layer of regulation is beginning to take shape. In this review, we summarize recent findings on the major RNA modifications identified in plants, with an emphasis on N6-methyladenosine (m6A), the most extensively studied modification. We discuss the functional significance of the effector components involved in m6A modification and its diverse roles in plant biotic interactions, including plant-virus, plant-bacterium, plant-fungus, and plant-insect relationships. Furthermore, we highlight new technological developments driving research progress in this field and outline key challenges that remain to be addressed.
Collapse
Affiliation(s)
- Linhao Ge
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Fuan Pan
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China; State Key Laboratory of Rice Biology, Institute of Biotechnology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Mingxuan Jia
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Delphine M Pott
- Department of Plant Biochemistry, Centre for Plant Molecular Biology (ZMBP), Eberhard Karls University, 72076 Tübingen, Germany
| | - Hao He
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Hongying Shan
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Rosa Lozano-Durán
- Department of Plant Biochemistry, Centre for Plant Molecular Biology (ZMBP), Eberhard Karls University, 72076 Tübingen, Germany
| | - Aiming Wang
- London Research and Development Centre, Agriculture and Agri-Food Canada, London, ON N5V 4T3, Canada
| | - Xueping Zhou
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China; State Key Laboratory of Rice Biology, Institute of Biotechnology, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Fangfang Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
13
|
Cai J, Shen L, Kang H, Xu T. RNA modifications in plant adaptation to abiotic stresses. PLANT COMMUNICATIONS 2025; 6:101229. [PMID: 39709520 PMCID: PMC11897461 DOI: 10.1016/j.xplc.2024.101229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/15/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
Epitranscriptomic chemical modifications of RNAs have emerged as potent regulatory mechanisms in the process of plant stress adaptation. Currently, over 170 distinct chemical modifications have been identified in mRNAs, tRNAs, rRNAs, microRNAs (miRNAs), and long noncoding RNAs (lncRNAs). Genetic and molecular studies have identified the genes responsible for addition and removal of chemical modifications from RNA molecules, which are known as "writers" and "erasers," respectively. N6-methyladenosine (m6A) is the most prevalent chemical modification identified in eukaryotic mRNAs. Recent studies have identified m6A writers and erasers across different plant species, including Arabidopsis (Arabidopsis thaliana), rice (Oryza sativa), cotton (Gossypium hirsutum), and tomato (Solanum lycopersicum). Accumulating discoveries have improved our understanding of the functions of RNA modifications in plant stress responses. This review highlights the latest research on RNA modification, emphasizing the biological and cellular roles of diverse chemical modifications of mRNAs, tRNAs, rRNAs, miRNAs, and lncRNAs in plant responses to environmental and hormonal signals. We also propose and discuss critical questions and future challenges for enhancing our understanding of the cellular and mechanistic roles of RNA modifications in plant stress responses. Integrating molecular insights into the regulatory roles of RNA modifications in stress responses with novel genome- and RNA-editing technologies will facilitate the breeding of stress-tolerant crops through precise engineering of RNA modifications.
Collapse
Affiliation(s)
- Jing Cai
- Jiangsu International Joint Center of Genomics, Jiangsu Key Laboratory of Comparative Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China
| | - Ling Shen
- Jiangsu International Joint Center of Genomics, Jiangsu Key Laboratory of Comparative Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China
| | - Hunseung Kang
- Jiangsu International Joint Center of Genomics, Jiangsu Key Laboratory of Comparative Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China; Department of Applied Biology, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, South Korea.
| | - Tao Xu
- Jiangsu International Joint Center of Genomics, Jiangsu Key Laboratory of Comparative Genomics, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China.
| |
Collapse
|
14
|
Garcia-Oliveira AL, Ortiz R, Sarsu F, Rasmussen SK, Agre P, Asfaw A, Kante M, Chander S. The importance of genotyping within the climate-smart plant breeding value chain - integrative tools for genetic enhancement programs. FRONTIERS IN PLANT SCIENCE 2025; 15:1518123. [PMID: 39980758 PMCID: PMC11839310 DOI: 10.3389/fpls.2024.1518123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 11/25/2024] [Indexed: 02/22/2025]
Abstract
The challenges faced by today's agronomists, plant breeders, and their managers encompass adapting sustainably to climate variability while working with limited budgets. Besides, managers are dealing with a multitude of issues with different organizations working on similar initiatives and projects, leading to a lack of a sustainable impact on smallholder farmers. To transform the current food systems as a more sustainable and resilient model efficient solutions are needed to deliver and convey results. Challenges such as logistics, labour, infrastructure, and equity, must be addressed alongside adapting to increasingly unstable climate conditions which affect the life cycle of transboundary pathogens and pests. In this context, transforming food systems go far beyond just farmers and plant breeders and it requires substantial contributions from industry, global finances, transportation, energy, education, and country developmental sectors including legislators. As a result, a holistic approach is essential for achieving sustainable and resilient food systems to sustain a global population anticipated to reach 9.7 billion by 2050 and 11.2 billion by 2100. As of 2021, nearly 193 million individuals were affected by food insecurity, 40 million more than in 2020. Meanwhile, the digital world is rapidly advancing with the digital economy estimated at about 20% of the global gross domestic product, suggesting that digital technologies are increasingly accessible even in areas affected by food insecurity. Leveraging these technologies can facilitate the development of climate-smart cultivars that adapt effectively to climate variation, meet consumer preferences, and address human and livestock nutritional needs. Most economically important traits in crops are controlled by multiple loci often with recessive alleles. Considering particularly Africa, this continent has several agro-climatic zones, hence crops need to be adapted to these. Therefore, targeting specific loci using modern tools offers a precise and efficient approach. This review article aims to address how these new technologies can provide a better support to smallholder farmers.
Collapse
Affiliation(s)
- Ana Luísa Garcia-Oliveira
- Genetic Resources Program, Alliance Bioversity International and International Center for Tropical Agriculture (CIAT), Cali, Colombia
| | - Rodomiro Ortiz
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Alnarp, Sweden
| | - Fatma Sarsu
- Plant Breeding and Genetics Section, Joint FAO/IAEA Center, International Atomic Energy Agency, Vienna, Austria
| | | | - Paterne Agre
- Yam Breeding Unit, International Institute of Tropical Agriculture, Ibadan, Nigeria
| | - Asrat Asfaw
- Yam Breeding Unit, International Institute of Tropical Agriculture, Ibadan, Nigeria
| | - Moctar Kante
- Genetics, Genomics, and Crop Improvement Division, International Potato Center, Lima, Peru
| | - Subhash Chander
- Oilseeds Section, Department of Genetics & Plant Breeding, CCS Haryana Agricultural University, Hisar, India
| |
Collapse
|
15
|
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
|
16
|
Li G, Cheng Y, Yu J, Zhu Y, Ma H, Zhou Y, Pu Z, Zhu G, Yuan Y, Zhang Z, Zhou X, Tian K, Qiao J, Hu X, Chen XX, Ji Q, Huang X, Ma B, Yao Y. Compact RNA editors with natural miniature Cas13j nucleases. Nat Chem Biol 2025; 21:280-290. [PMID: 39300230 DOI: 10.1038/s41589-024-01729-8] [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: 07/11/2023] [Accepted: 08/15/2024] [Indexed: 09/22/2024]
Abstract
Clustered regularly interspaced short palindromic repeats-Cas13 effectors are used for RNA editing but the adeno-associated virus (AAV) packaging limitations because of their big sizes hinder their therapeutic application. Here we report the identification of the Cas13j family, with LepCas13j (529 aa) and ChiCas13j (424 aa) being the smallest and most highly efficient variants for RNA interference. The miniaturized Cas13j proteins enable the development of compact RNA base editors. Chi-RESCUE-S, by fusing dChiCas13j with hADAR2dd, demonstrates high efficiency and specificity in A-to-G and C-to-U conversions. Importantly, this system is compatible with single-AAV packaging without the need for protein sequence truncation. It successfully corrected pathogenic mutations, such as APOC3D65N and SCN9AR896Q, to the wild-type forms. In addition, we developed an optimized system, Chi-RESCUE-S-mini3, which pioneered efficient in vivo C-to-U RNA editing of PCSK9 in mice through single-AAV delivery, resulting in reduced total cholesterol levels. These results highlight the potential of Cas13j to treat human diseases.
Collapse
Affiliation(s)
- Guo Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.
- Xianghu Laboratory, Hangzhou, China.
| | - Yaxian Cheng
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Jingwen Yu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Agricultural Resources and Environment, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China
| | - Yunfei Zhu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
| | - Hongru Ma
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Yuqiao Zhou
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Zhongji Pu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
- Xianghu Laboratory, Hangzhou, China
| | - Guanglin Zhu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | | | - Ziyue Zhang
- Zhejiang Institute of Tianjin University, Shaoxing, China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
| | - Xinzhi Zhou
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Kairen Tian
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Zhejiang Institute of Tianjin University, Shaoxing, China
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Zhejiang Institute of Tianjin University, Shaoxing, China
| | - Xiaoxiang Hu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
| | - Xue-Xin Chen
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Quanjiang Ji
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | | | - Bin Ma
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
- Zhejiang Provincial Key Laboratory of Agricultural Resources and Environment, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China.
| | - Yuan Yao
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
17
|
Bosmeny MS, Pater AA, Zhang L, Sha BE, Lyu Z, Larkai L, Damha MJ, Mamede JI, Gagnon KT. An HIV-1 Reference Epitranscriptome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635805. [PMID: 39975020 PMCID: PMC11838527 DOI: 10.1101/2025.01.30.635805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Post-transcriptional chemical modifications to RNA, or the epitranscriptome, play important roles in RNA metabolism, gene regulation, and human disease, including viral pathogenesis. Modifications to the RNA viral genome and transcripts of human immunodeficiency virus 1 (HIV-1) have been reported, including methylation of adenosine (m6A) and cytosine (m5C), acetylation of cytosine, pseudouridylation (psi), and conversion of adenosine to inosine, and their effects on virus and host biology have been investigated. However, diverse experimental approaches have been used, making clear correlations across studies difficult to assess. To address this need, we propose the establishment of a reference HIV-1 epitranscriptome. We sequenced the model NL4-3 HIV-1 genome from infected Jurkat CD4+ T cells cells using the latest nanopore chemistry, custom RNA preparation methods, and commercial base-calling algorithms. This resulted in a reproducible sense and preliminary antisense HIV-1 epitranscriptome where m6A, m5C, psi, ands inosine could be identified by multiplexed base-calling. Multiplexed base-calling miscalled modifications due to sequence and neighboring modification contexts, which we demonstrate can be corrected with synthetic HIV-1 RNA fragments. We validate m6A modification sites with a small molecule inhibitor of methyltransferase-like 3 (METTL3), STM2457. We conclude that modifications do not change substantially under combination antiretroviral therapy (cART) treatment or in primary CD4+ T cells. Samples from patients living with HIV reveal conservation of certain modifications, such as m6A. Our approach and reference data offer a straightforward benchmark that can be adopted to help advance rigor, reproducibility, and uniformity across future HIV-1 epitranscriptomics studies.
Collapse
Affiliation(s)
- Michael S. Bosmeny
- Dept. of Biochemistry, Wake Forest University, School of Medicine, Winston-Salem, North Carolina, USA, 27101
- Equally contributing authors
| | - Adrian A. Pater
- Dept. of Biochemistry, Wake Forest University, School of Medicine, Winston-Salem, North Carolina, USA, 27101
- Equally contributing authors
| | - Li Zhang
- Equally contributing authors
- Dept. of Microbial Pathogens and Immunity, Rush University, Chicago, Illinois, USA, 60612
| | - Beverly E. Sha
- Division of Infectious Diseases, Rush University Medical Center, Chicago, Illinois, USA, 60612
| | - Zidi Lyu
- Dept. of Chemistry, McGill University, Montreal, Canada, H3A, 0G3
| | - Lydia Larkai
- Dept. of Biochemistry, Wake Forest University, School of Medicine, Winston-Salem, North Carolina, USA, 27101
| | - Masad J. Damha
- Dept. of Chemistry, McGill University, Montreal, Canada, H3A, 0G3
| | - Joao I. Mamede
- Dept. of Microbial Pathogens and Immunity, Rush University, Chicago, Illinois, USA, 60612
| | - Keith T. Gagnon
- Dept. of Biochemistry, Wake Forest University, School of Medicine, Winston-Salem, North Carolina, USA, 27101
| |
Collapse
|
18
|
Su-Tobon Q, Fan J, Goldstein M, Feeney K, Ren H, Autissier P, Wang P, Huang Y, Mohanty U, Niu J. CRISPR-Hybrid: A CRISPR-Mediated Intracellular Directed Evolution Platform for RNA Aptamers. Nat Commun 2025; 16:595. [PMID: 39799111 PMCID: PMC11724954 DOI: 10.1038/s41467-025-55957-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
Recent advances in gene editing and precise regulation of gene expression based on CRISPR technologies have provided powerful tools for the understanding and manipulation of gene functions. Fusing RNA aptamers to the sgRNA of CRISPR can recruit cognate RNA-binding protein (RBP) effectors to target genomic sites, and the expression of sgRNA containing different RNA aptamers permit simultaneous multiplexed and multifunctional gene regulations. Here, we report an intracellular directed evolution platform for RNA aptamers against intracellularly expressed RBPs. We optimize a bacterial CRISPR-hybrid system coupled with FACS, and identified high affinity RNA aptamers orthogonal to existing aptamer-RBP pairs. Application of orthogonal aptamer-RBP pairs in multiplexed CRISPR allows effective simultaneous transcriptional activation and repression of endogenous genes in mammalian cells.
Collapse
Affiliation(s)
- Qiwen Su-Tobon
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Jiayi Fan
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | | | - Kevin Feeney
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Hongyuan Ren
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | | | - Peiyi Wang
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Yingzi Huang
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Udayan Mohanty
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA
| | - Jia Niu
- Department of Chemistry, Boston College, Chestnut Hill, MA, USA.
| |
Collapse
|
19
|
Wang PX, Zhu L, Xiang M, Zhang R, Zheng X, Zheng Z, Li K. FTO Alleviates Hepatic Ischemia-Reperfusion Injury by Regulating Apoptosis and Autophagy. Gastroenterol Res Pract 2025; 2025:5587859. [PMID: 39811145 PMCID: PMC11730018 DOI: 10.1155/grp/5587859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/07/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Objective: Despite N6-methyladenosine (m6A) being closely involved in various pathophysiological processes, its potential role in liver injury is largely unknown. We designed the current research to study the potential role of fat mass and obesity-associated protein (FTO), an m6A demethylase, on hepatic ischemia-reperfusion injury (IRI). Methods: Wild-type mice injected with an adeno-associated virus carrying fat mass and obesity-associated protein (AAV-FTO) or adeno-associated virus carrying green fluorescent protein (GFP) (AAV-GFP) were subjected to a hepatic IRI model in vivo. Hematoxylin-eosin staining was performed to observe IRI. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining was used to observe the cell apoptosis. Reverse transcription polymerase chain reaction (RT-PCR) was used to observe the expression of FTO. The protein levels of FTO, apoptosis, or autophagy-associated signaling proteins were detected by western blot. Reactive oxygen species (ROS) levels were determined by flow cytometry, and immunohistochemistry was used to detect the FTO and LC3-II expression. For in vitro experiments, cultured hepatocytes were subjected to hypoxia/reoxygenation (H/R) stimulation. Monodansylcadaverine (MDC) staining was used to visualize autophagic vesicles. Results: In the present study, we showed that FTO was involved in hepatic IRI, apoptosis, and autophagy. Specifically, the expression level of FTO was significantly reduced in the hepatic IRI. Besides, increasing FTO expression (AAV-FTO) ameliorated the hepatic IRI in animal models, accompanied by decreased apoptosis and autophagy. Furthermore, the FTO inhibitor (FB23-2) aggravated autophagy in hepatocytes upon H/R-induced damage. Conclusion: FTO could act as a protective effector during hepatic IRI, associated with decreased apoptosis and autophagy. FTO-mediated m6A demethylation modification may be an important therapeutic target for hepatic IRI.
Collapse
Affiliation(s)
- Pi-Xiao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Xiang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rixin Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolin Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Li
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Shen L. Epitranscriptomic regulation through phase separation in plants. TRENDS IN PLANT SCIENCE 2024:S1360-1385(24)00313-3. [PMID: 39706711 DOI: 10.1016/j.tplants.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/23/2024]
Abstract
Epitranscriptomic regulation has emerged as a crucial layer of gene control where RNA modifications, particularly N6-methyladenosine (m6A), introduce complexity and versatility to gene regulation. Increasing evidence suggests that epitranscriptomic regulation through phase separation plays critical roles in mediating RNA metabolism during plant development and stress responses. m6A-associated biomolecular condensates formed via phase separation act as dynamic cellular hotspots where m6A effectors, RNAs, and other regulatory proteins coalesce to facilitate RNA regulation. Moreover, m6A modulates condensate assembly. Herein, I summarize the current understanding of how m6A- and m6A effector-mediated formation of biomolecular condensates mediates plant development and stress adaptation. I also discuss several working models for m6A-associated biomolecular condensates and highlight the prospects for future research on epitranscriptomic regulation through phase separation.
Collapse
Affiliation(s)
- Lisha Shen
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, 117604, Singapore; Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543, Singapore.
| |
Collapse
|
21
|
Otonari K, Asami Y, Ogata K, Ishihama Y, Futaki S, Imanishi M. Highly sequence-specific, timing-controllable m 6A demethylation by modulating RNA-binding affinity of m 6A erasers. Chem Commun (Camb) 2024; 61:69-72. [PMID: 39499124 DOI: 10.1039/d4cc04070h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Recent advancements in tools using programmable RNA binding proteins and m6A-erasers enable sequence-selective and timing-controllable m6A demethylation. However, off-target effects are still a concern. This study addresses the problem by reducing the RNA-binding ability of m6A-erasers. The modulated m6A-erasers achieved sequence-specific and timing-controllable m6A demethylation with minimal off-target activity.
Collapse
Affiliation(s)
- Kenko Otonari
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| | - Yuri Asami
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| | - Kosuke Ogata
- National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Science, Kyoto University, Kyoto 606-8501, Japan
- National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Shiroh Futaki
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| | - Miki Imanishi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan.
| |
Collapse
|
22
|
Uddin MB, Wang Z, Yang C. Epitranscriptomic RNA m 6A Modification in Cancer Therapy Resistance: Challenges and Unrealized Opportunities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 12:e2403936. [PMID: 39661414 PMCID: PMC11775542 DOI: 10.1002/advs.202403936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/24/2024] [Indexed: 12/12/2024]
Abstract
Significant advances in the development of new cancer therapies have given rise to multiple novel therapeutic options in chemotherapy, radiotherapy, immunotherapy, and targeted therapies. Although the development of resistance is often reported along with temporary disease remission, there is often tumor recurrence of an even more aggressive nature. Resistance to currently available anticancer drugs results in poor overall and disease-free survival rates for cancer patients. There are multiple mechanisms through which tumor cells develop resistance to therapeutic agents. To date, efforts to overcome resistance have only achieved limited success. Epitranscriptomics, especially related to m6A RNA modification dysregulation in cancer, is an emerging mechanism for cancer therapy resistance. Here, recent studies regarding the contributions of m6A modification and its regulatory proteins to the development of resistance to different cancer therapies are comprehensively reviewed. The promise and potential limitations of targeting these entities to overcome resistance to various anticancer therapies are also discussed.
Collapse
Affiliation(s)
- Mohammad Burhan Uddin
- Department of Pharmaceutical SciencesNorth South UniversityBashundharaDhaka1229Bangladesh
| | - Zhishan Wang
- Stony Brook Cancer CenterStony Brook UniversityStony BrookNY11794USA
| | - Chengfeng Yang
- Stony Brook Cancer CenterStony Brook UniversityStony BrookNY11794USA
- Department of PathologyRenaissance School of MedicineStony Brook UniversityStony BrookNY11794USA
| |
Collapse
|
23
|
Zhou Z, Wang YQ, Zheng XN, Zhang XH, Ji LY, Han JY, Zuo ZC, Mo WL, Zhang L. Optimizing ABA-based chemically induced proximity for enhanced intracellular transcriptional activation and modification response to ABA. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2650-2663. [PMID: 39172347 DOI: 10.1007/s11427-024-2707-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Abscisic acid (ABA)-based chemically induced proximity (CIP) is primarily mediated by the interaction of the ABA receptor pyrabactin resistance 1-like 1 (PYL1) and the 2C-type protein phosphatase ABI1, which confers ABA-induced proximity to their fusion proteins, and offers precise temporal control of a wide array of biological processes. However, broad application of ABA-based CIP has been limited by ABA response intensity. In this study, we demonstrated that ABA-induced interaction between another ABA receptor pyrabactin resistance 1 (PYR1) and ABI1 exhibited higher ABA response intensity than that between PYL1 and ABI1 in HEK293T cells. We engineered PYR1-ABI1 and PYL1-ABI1 into ABA-induced transcriptional activation tools in mammalian cells by integration with CRISPR/dCas9 and found that the tool based on PYR1-ABI1 demonstrated better ABA response intensity than that based on PYL1-ABI1 for both exogenous and endogenous genes in mammalian cells. We further achieved ABA-induced RNA m6A modification installation and erasure by combining ABA-induced PYR1-ABI1 interaction with CRISPR/dCas13, successfully inhibiting tumor cell proliferation. We subsequently improved the interaction of PYR1-ABI1 through phage-assisted continuous evolution (PACE), successfully generating a PYR1 mutant (PYR1m) whose interaction with ABI1 exhibited a higher ABA response intensity than that of the wild-type. In addition, we tested the transcriptional activation tool based on PYRm-ABI1 and found that it also showed a higher ABA response intensity than that of the wild type. These results demonstrate that we have developed a novel ABA-based CIP and further improved upon it using PACE, providing a new approach for the modification of other CIP systems.
Collapse
Affiliation(s)
- Zeng Zhou
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Basic Forestry and Proteomics Research Center, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yue-Qi Wang
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China
| | - Xu-Nan Zheng
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China
| | - Xiao-Hong Zhang
- Basic Forestry and Proteomics Research Center, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Lu-Yao Ji
- Basic Forestry and Proteomics Research Center, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Jun-You Han
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China
| | - Ze-Cheng Zuo
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China.
| | - Wei-Liang Mo
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China.
| | - Li Zhang
- Jilin Province Engineering Laboratory of Plant Genetic Improvement, College of Plant Science, Jilin University, Changchun, 130062, China.
| |
Collapse
|
24
|
Diao L, Xie S, Xu W, Zhang H, Hou Y, Hu Y, Liang X, Liang J, Zhang Q, Xiao Z. CRISPR/Cas13 sgRNA-Mediated RNA-RNA Interaction Mapping in Live Cells with APOBEC RNA Editing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409004. [PMID: 39392366 PMCID: PMC11615753 DOI: 10.1002/advs.202409004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/18/2024] [Indexed: 10/12/2024]
Abstract
Current research on long non-coding RNA (lncRNA) has predominantly focused on identifying their protein partners and genomic binding sites, leaving their RNA partners largely unknown. To address this gap, the study has developed a method called sarID (sgRNA scaffold assisted RNA-RNA interaction detection), which integrates Cas13-based RNA targeting, sgRNA engineering, and proximity RNA editing to investigate lncRNA-RNA interactomes. By applying sarID to the lncRNA NEAT1, over one thousand previously unidentified binding transcripts are discovered. sarID is further expanded to investigate binders of XIST, MALAT1, NBR2, and DANCR, demonstrating its broad applicability in identifying lncRNA-RNA interactions. The findings suggest that lncRNAs may regulate gene expression by interacting with mRNAs, expanding their roles beyond known functions as protein scaffolds, miRNA sponges, or guides for epigenetic modulators. sarID has the potential to be adapted for studying other specific RNAs, providing a novel immunoprecipitation-free method for uncovering RNA partners and facilitating the exploration of the RNA-RNA interactome.
Collapse
Affiliation(s)
- Li‐Ting Diao
- Biotherapy Center, The Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630P. R. China
| | - Shu‐Juan Xie
- Institute of VaccineThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630P. R. China
| | - Wan‐Yi Xu
- Biotherapy Center, The Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630P. R. China
| | | | - Ya‐Rui Hou
- Biotherapy Center, The Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630P. R. China
| | - Yan‐Xia Hu
- Biotherapy Center, The Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630P. R. China
| | | | | | - Qi Zhang
- Biotherapy Center, The Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630P. R. China
- Institute of VaccineThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630P. R. China
| | - Zhen‐Dong Xiao
- Biotherapy Center, The Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease ResearchThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630P. R. China
| |
Collapse
|
25
|
Wen J, Zhu Q, Liu Y, Gou LT. RNA modifications: emerging players in the regulation of reproduction and development. Acta Biochim Biophys Sin (Shanghai) 2024; 57:33-58. [PMID: 39574165 PMCID: PMC11802351 DOI: 10.3724/abbs.2024201] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 11/05/2024] [Indexed: 01/25/2025] Open
Abstract
The intricate world of RNA modifications, collectively termed the epitranscriptome, covers over 170 identified modifications and impacts RNA metabolism and, consequently, almost all biological processes. In this review, we focus on the regulatory roles and biological functions of a panel of dominant RNA modifications (including m 6A, m 5C, Ψ, ac 4C, m 1A, and m 7G) on three RNA types-mRNA, tRNA, and rRNA-in mammalian development, particularly in the context of reproduction as well as embryonic development. We discuss in detail how those modifications, along with their regulatory proteins, affect RNA processing, structure, localization, stability, and translation efficiency. We also highlight the associations among dysfunctions in RNA modification-related proteins, abnormal modification deposition and various diseases, emphasizing the roles of RNA modifications in critical developmental processes such as stem cell self-renewal and cell fate transition. Elucidating the molecular mechanisms by which RNA modifications influence diverse developmental processes holds promise for developing innovative strategies to manage developmental disorders. Finally, we outline several unexplored areas in the field of RNA modification that warrant further investigation.
Collapse
Affiliation(s)
- Junfei Wen
- Key Laboratory of RNA InnovationScience and EngineeringShanghai Key Laboratory of Molecular AndrologyCAS Center for Excellence in Molecular. Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Qifan Zhu
- Key Laboratory of RNA InnovationScience and EngineeringShanghai Key Laboratory of Molecular AndrologyCAS Center for Excellence in Molecular. Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yong Liu
- Key Laboratory of RNA InnovationScience and EngineeringShanghai Key Laboratory of Molecular AndrologyCAS Center for Excellence in Molecular. Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
| | - Lan-Tao Gou
- Key Laboratory of RNA InnovationScience and EngineeringShanghai Key Laboratory of Molecular AndrologyCAS Center for Excellence in Molecular. Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
26
|
Chen C, Qi LS. Precision Transcriptome Editing. ACS Synth Biol 2024; 13:3487-3496. [PMID: 39435985 PMCID: PMC12050085 DOI: 10.1021/acssynbio.4c00183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Manipulating RNA species in mammalian cells has emerged as an important strategy for precise gene expression control. Here we review recent advances in precision transcriptome editing with a focus on tools that engineer specific transcripts for abundance, translation, base editing, alternative isoforms, and chemical modifications. While some of these methods have demonstrated efficiency in therapeutically relevant cellular or in vivo models, most require further study on their clinical safety and efficacy. Precision transcriptome engineering holds great potential for both mechanistic study of RNA biology and future gene and cell-based therapeutic applications.
Collapse
Affiliation(s)
- Crystal Chen
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | - Lei S. Qi
- Department of Bioengineering, Stanford University, Stanford, CA 94305
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305
- Chan Zuckerberg Biohub – San Francisco, San Francisco, CA 94158
| |
Collapse
|
27
|
Kaur P, Sharma P, Bhatia P, Singh M. Current insights on m6A RNA modification in acute leukemia: therapeutic targets and future prospects. Front Oncol 2024; 14:1445794. [PMID: 39600630 PMCID: PMC11590065 DOI: 10.3389/fonc.2024.1445794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 10/08/2024] [Indexed: 11/29/2024] Open
Abstract
RNA modification is the critical mechanism for regulating post-transcriptional processes. There are more than 150 RNA modifications reported so far, among which N6-Methyladenosine is the most prevalent one. M6A RNA modification complex consists of 'writers', 'readers' and 'erasers' which together in a group catalyze, recognize and regulate the methylation process of RNA and thereby regulate the stability and translation of mRNA. The discovery of erasers also known as demethylases, revolutionized the research on RNA modifications as it revealed that this modification is reversible. Since then, various studies have focused on discovering the role of m6A modification in various diseases especially cancers. Aberrant expression of these 'readers', 'writers', and 'erasers' is found to be altered in various cancers resulting in disturbance of cellular homeostasis. Acute leukemias are the most common cancer found in pediatric patients and account for 20% of adult cases. Dysregulation of the RNA modifying complex have been reported in development and progression of hematopoietic malignancies. Further, targeting m6A modification is the new approach for cancer immunotherapy and is being explored extensively. This review provides detailed information about current information on the role of m6A RNA modification in acute leukemia and their therapeutic potential.
Collapse
Affiliation(s)
| | | | | | - Minu Singh
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical
Education and Research, Chandigarh, India
| |
Collapse
|
28
|
Ochiai Y, Clifton B, Le Coz M, Terenzio M, Laurino P. SUPREM: an engineered non-site-specific m6A RNA methyltransferase with highly improved efficiency. Nucleic Acids Res 2024; 52:12158-12172. [PMID: 39417589 PMCID: PMC11551740 DOI: 10.1093/nar/gkae887] [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: 09/05/2023] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
N 6-Methyladenine (m6A) RNA methylation plays a key role in RNA processing and translational regulation, influencing both normal physiological and pathological processes. Yet, current techniques for studying RNA methylation struggle to isolate the effects of individual m6A modifications. Engineering of RNA methyltransferases (RNA MTases) could enable development of improved synthetic biology tools to manipulate RNA methylation, but it is challenging due to limited understanding of structure-function relationships in RNA MTases. Herein, using ancestral sequence reconstruction, we explore the sequence space of the bacterial DNA methyltransferase EcoGII (M.EcoGII), a promising target for protein engineering due to its lack of sequence specificity and its residual activity on RNA. We thereby created an efficient non-specific RNA MTase termed SUPer RNA EcoGII Methyltransferase (SUPREM), which exhibits 8-fold higher expression levels, 7°C higher thermostability and 12-fold greater m6A RNA methylation activity compared with M.EcoGII. Immunofluorescent staining and quantitative liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis confirmed SUPREM's higher RNA methylation activity compared with M.EcoGII in mammalian cells. Additionally, Nanopore direct RNA sequencing highlighted that SUPREM is capable of methylating a larger number of RNA methylation sites than M.EcoGII. Through phylogenetic and mutational analysis, we identified a critical residue for the enhanced RNA methylation activity of SUPREM. Collectively, our findings indicate that SUPREM holds promise as a versatile tool for in vivo RNA methylation and labeling.
Collapse
Affiliation(s)
- Yoshiki Ochiai
- Protein Engineering and Evolution Unit, Okinawa Institute of Science and Technology Graduate University (OIST), 1919-1 Tancha, Onna, Kunigami District, Okinawa 904-0495, Japan
| | - Ben E Clifton
- Protein Engineering and Evolution Unit, Okinawa Institute of Science and Technology Graduate University (OIST), 1919-1 Tancha, Onna, Kunigami District, Okinawa 904-0495, Japan
| | - Madeleine Le Coz
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University (OIST), 1919-1 Tancha, Onna, Kunigami District, Okinawa 904-0495, Japan
| | - Marco Terenzio
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University (OIST), 1919-1 Tancha, Onna, Kunigami District, Okinawa 904-0495, Japan
| | - Paola Laurino
- Protein Engineering and Evolution Unit, Okinawa Institute of Science and Technology Graduate University (OIST), 1919-1 Tancha, Onna, Kunigami District, Okinawa 904-0495, Japan
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita-shi, Osaka 565-0871, Japan
| |
Collapse
|
29
|
Tan L, Zhu C, Zhang X, Fu J, Huang T, Zhang W, Zhang W. Mitochondrial RNA methylation in cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189213. [PMID: 39521292 DOI: 10.1016/j.bbcan.2024.189213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/08/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Mitochondria have a complete and independent genetic system with necessary biological energy for cancer occurrence and persistence. Mitochondrial RNA (mt-RNA) methylation, as a frontier in epigenetics, has linked to cancer progression with growing evidences. This review has comprehensively summarized detailed mechanisms of mt-RNA methylation in regulating cancer proliferation, metastasis, and immune infiltration from the mt-RNA methylation sites, biological significance, and its methyltransferases. The mt-RNA methylation also plays a very significant role via epigenetic crosstalk between nucleus and mitochondria. Importantly, the unique structures and functional characteristics of mt-RNA methyltransferases and the potential targeting treatment drugs for cancer are also analyzed. Revealing human mt-RNA methylation regulatory system and the relationship with cancer will contribute to identifying potential biomarkers and therapeutic targets for precise prevention, detection, intervention and treatment in the future.
Collapse
Affiliation(s)
- Luyi Tan
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Chenyu Zhu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Xinyu Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Jiaqi Fu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Tingting Huang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Wenji Zhang
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510640, PR China.
| | - Wenjuan Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, PR China.
| |
Collapse
|
30
|
Chen Z, Zhang J, Wang J, Tong H, Pan W, Ma F, Wu Q, Dai J. N6-methyladenosine RNA modification promotes Severe Fever with Thrombocytopenia Syndrome Virus infection. PLoS Pathog 2024; 20:e1012725. [PMID: 39585899 PMCID: PMC11627400 DOI: 10.1371/journal.ppat.1012725] [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: 05/06/2024] [Revised: 12/09/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024] Open
Abstract
Severe Fever with Thrombocytopenia Syndrome Virus (SFTSV), a novel bunyavirus primarily transmitted by Haemaphysalis longicornis, induces severe disease with a high mortality rate. N6-methyladenosine (m6A) is a prevalent internal chemical modification in eukaryotic mRNA that has been reported to regulate viral infection. However, the role of m6A modification during SFTSV infection remains elusive. We here reported that SFTSV RNAs bear m6A modification during infection. Manipulating the expressions or activities of host m6A regulators significantly impacted SFTSV infection. Mechanistically, SFTSV recruited m6A regulators through the nucleoprotein to modulate the m6A modification of viral RNA, eventually resulting in enhanced infection by promoting viral mRNA translation efficiency and/or genome RNA stability. m6A mutations in the S genome diminished virus particle production, while m6A mutations in the G transcript impaired the replication of recombinant vesicular stomatitis virus (rVSV) expressing G protein in vitro and in vivo. Interestingly, m6A modification was evolutionarily conserved and facilitated SFTSV infection in primary tick cells. These findings may open an avenue for the development of m6A-targeted anti-SFTSV vaccines, drugs, and innovative strategies for the prevention and control of tick-borne disease.
Collapse
Affiliation(s)
- Zhiqiang Chen
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinyu Zhang
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jun Wang
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Hao Tong
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wen Pan
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Feng Ma
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Qihan Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jianfeng Dai
- Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, The Forth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
31
|
Hu W, Kumar A, Ahmed SF, Qi S, Ma DKG, Chen H, Singh GJ, Casan JML, Haber M, Voskoboinik I, McKay MR, Trapani JA, Ekert PG, Fareh M. Single-base tiled screen unveils design principles of PspCas13b for potent and off-target-free RNA silencing. Nat Struct Mol Biol 2024; 31:1702-1716. [PMID: 38951623 PMCID: PMC11564092 DOI: 10.1038/s41594-024-01336-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/15/2024] [Indexed: 07/03/2024]
Abstract
The development of precise RNA-editing tools is essential for the advancement of RNA therapeutics. CRISPR (clustered regularly interspaced short palindromic repeats) PspCas13b is a programmable RNA nuclease predicted to offer superior specificity because of its 30-nucleotide spacer sequence. However, its design principles and its on-target, off-target and collateral activities remain poorly characterized. Here, we present single-base tiled screening and computational analyses that identify key design principles for potent and highly selective RNA recognition and cleavage in human cells. We show that the de novo design of spacers containing guanosine bases at precise positions can greatly enhance the catalytic activity of inefficient CRISPR RNAs (crRNAs). These validated design principles (integrated into an online tool, https://cas13target.azurewebsites.net/ ) can predict highly effective crRNAs with ~90% accuracy. Furthermore, the comprehensive spacer-target mutagenesis revealed that PspCas13b can tolerate only up to four mismatches and requires ~26-nucleotide base pairing with the target to activate its nuclease domains, highlighting its superior specificity compared to other RNA or DNA interference tools. On the basis of this targeting resolution, we predict an extremely low probability of PspCas13b having off-target effects on other cellular transcripts. Proteomic analysis validated this prediction and showed that, unlike other Cas13 orthologs, PspCas13b exhibits potent on-target activity and lacks collateral effects.
Collapse
Affiliation(s)
- Wenxin Hu
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Amit Kumar
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Diagnostic Genomics, Monash Health Pathology, Monash Medical Centre, Clayton, Victoria, Australia
| | - Syed Faraz Ahmed
- Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Shijiao Qi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - David K G Ma
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Honglin Chen
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Gurjeet J Singh
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Joshua M L Casan
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ilia Voskoboinik
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Matthew R McKay
- Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Joseph A Trapani
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul G Ekert
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Mohamed Fareh
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
32
|
Pupak A, Rodríguez-Navarro I, Sathasivam K, Singh A, Essmann A, Del Toro D, Ginés S, Mouro Pinto R, Bates GP, Vang Ørom UA, Martí E, Brito V. m 6A modification of mutant huntingtin RNA promotes the biogenesis of pathogenic huntingtin transcripts. EMBO Rep 2024; 25:5026-5052. [PMID: 39394467 PMCID: PMC11549361 DOI: 10.1038/s44319-024-00283-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 09/20/2024] [Accepted: 09/27/2024] [Indexed: 10/13/2024] Open
Abstract
In Huntington's disease (HD), aberrant processing of huntingtin (HTT) mRNA produces HTT1a transcripts that encode the pathogenic HTT exon 1 protein. The mechanisms behind HTT1a production are not fully understood. Considering the role of m6A in RNA processing and splicing, we investigated its involvement in HTT1a generation. Here, we show that m6A methylation is increased before the cryptic poly(A) sites (IpA1 and IpA2) within the huntingtin RNA in the striatum of Hdh+/Q111 mice and human HD samples. We further assessed m6A's role in mutant Htt mRNA processing by pharmacological inhibition and knockdown of METTL3, as well as targeted demethylation of Htt intron 1 using a dCas13-ALKBH5 system in HD mouse cells. Our data reveal that Htt1a transcript levels are regulated by both METTL3 and the methylation status of Htt intron 1. They also show that m6A methylation in intron 1 depends on expanded CAG repeats. Our findings highlight a potential role for m6A in aberrant splicing of Htt mRNA.
Collapse
Affiliation(s)
- Anika Pupak
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Irene Rodríguez-Navarro
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Kirupa Sathasivam
- Department of Neurodegenerative Disease, Huntington's Disease Centre and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, UCL, London, WC1N 3BG, UK
| | - Ankita Singh
- Department for Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Amelie Essmann
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Daniel Del Toro
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Silvia Ginés
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ricardo Mouro Pinto
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gillian P Bates
- Department of Neurodegenerative Disease, Huntington's Disease Centre and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, UCL, London, WC1N 3BG, UK
| | | | - Eulàlia Martí
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Verónica Brito
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurosciències, Universitat de Barcelona, Barcelona, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
33
|
Núñez-Álvarez Y, Espie-Caullet T, Buhagiar G, Rubio-Zulaika A, Alonso-Marañón J, Luna-Pérez E, Blazquez L, Luco R. A CRISPR-dCas13 RNA-editing tool to study alternative splicing. Nucleic Acids Res 2024; 52:11926-11939. [PMID: 39162234 PMCID: PMC11514487 DOI: 10.1093/nar/gkae682] [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: 04/30/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alternative splicing allows multiple transcripts to be generated from the same gene to diversify the protein repertoire and gain new functions despite a limited coding genome. It can impact a wide spectrum of biological processes, including disease. However, its significance has long been underestimated due to limitations in dissecting the precise role of each splicing isoform in a physiological context. Furthermore, identifying key regulatory elements to correct deleterious splicing isoforms has proven equally challenging, increasing the difficulty of tackling the role of alternative splicing in cell biology. In this work, we take advantage of dCasRx, a catalytically inactive RNA targeting CRISPR-dCas13 ortholog, to efficiently switch alternative splicing patterns of endogenous transcripts without affecting overall gene expression levels cost-effectively. Additionally, we demonstrate a new application for the dCasRx splice-editing system to identify key regulatory RNA elements of specific splicing events. With this approach, we are expanding the RNA toolkit to better understand the regulatory mechanisms underlying alternative splicing and its physiological impact in various biological processes, including pathological conditions.
Collapse
Affiliation(s)
- Yaiza Núñez-Álvarez
- Institut de Génétique Humaine, Université de Montpellier, CNRS UMR9002, Montpellier, France
| | - Tristan Espie-Caullet
- Institut de Génétique Humaine, Université de Montpellier, CNRS UMR9002, Montpellier, France
- Institut Curie, Paris-Saclay Research University, CNRS UMR3348, 91401 Orsay, France
- Team supported by la Ligue contre le Cancer, France
| | - Géraldine Buhagiar
- Institut Curie, Paris-Saclay Research University, CNRS UMR3348, 91401 Orsay, France
- Team supported by la Ligue contre le Cancer, France
| | - Ane Rubio-Zulaika
- Department of Neurosciences, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain
| | - Josune Alonso-Marañón
- Department of Neurosciences, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain
| | - Elvira Luna-Pérez
- Institut Curie, Paris-Saclay Research University, CNRS UMR3348, 91401 Orsay, France
- Team supported by la Ligue contre le Cancer, France
| | - Lorea Blazquez
- Department of Neurosciences, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- CIBERNED, ISCIII (CIBER, Carlos III Institute, Spanish Ministry of Sciences and Innovation), 28031 Madrid, Spain
| | - Reini F Luco
- Institut de Génétique Humaine, Université de Montpellier, CNRS UMR9002, Montpellier, France
- Institut Curie, Paris-Saclay Research University, CNRS UMR3348, 91401 Orsay, France
- Team supported by la Ligue contre le Cancer, France
| |
Collapse
|
34
|
Wang Y, Liu KI, Liu MM, Ooi KH, Nguyen TA, Chee JE, Teo SXD, He S, Tay JWD, Teo SY, Liew KS, Ge XY, Ng ZJ, Avagyan H, Liu H, Yi Z, Chang K, Kok EPL, Chen R, Yau CE, Koh JW, Wan Y, Tan MH. A circularly permuted CasRx platform for efficient, site-specific RNA editing. Nat Biotechnol 2024:10.1038/s41587-024-02430-w. [PMID: 39385008 DOI: 10.1038/s41587-024-02430-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 09/13/2024] [Indexed: 10/11/2024]
Abstract
Inactive Cas13 orthologs have been fused to a mutant human ADAR2 deaminase domain at the C terminus to enable programmable adenosine-to-inosine (A-to-I) RNA editing in selected transcripts. Although promising, existing RNA-editing tools generally suffer from a trade-off between efficacy and specificity, and off-target editing remains an unsolved problem. Here we describe the development of an optimized RNA-editing platform by rational protein engineering, CasRx-based Programmable Editing of RNA Technology (xPERT). We demonstrate that the topological rearrangement of a CasRx K940L mutant by circular permutation results in a robust scaffold for the tethering of a deaminase domain. We benchmark our tool against the REPAIR system and show that xPERT exhibits strong on-target activity like REPAIRv1 but low off-target editing like REPAIRv2. Our xPERT platform can be used to alter RNA sequence information without risking genome damage, effect temporary cellular changes and customize protein function.
Collapse
Affiliation(s)
- Yuanming Wang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Kaiwen Ivy Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Mengying Mandy Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Kean Hean Ooi
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Tram Anh Nguyen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Jiunn En Chee
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Shun Xiang Danny Teo
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- HP-NTU Digital Manufacturing Corporate Lab, Nanyang Technological University, Singapore, Singapore
| | - Shan He
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Jie Wen Douglas Tay
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Seok Yee Teo
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Kai Shin Liew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Xiao Yu Ge
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Zhi Jian Ng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Hasmik Avagyan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Hao Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Zirong Yi
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Keziah Chang
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Eng Piew Louis Kok
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Runjia Chen
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- National Junior College, Singapore, Singapore
| | - Chun En Yau
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Hwa Chong Institution, Singapore, Singapore
| | - Jun Wei Koh
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Hwa Chong Institution, Singapore, Singapore
| | - Yue Wan
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Meng How Tan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore.
- HP-NTU Digital Manufacturing Corporate Lab, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
35
|
Yu L, Alariqi M, Li B, Hussain A, Zhou H, Wang Q, Wang F, Wang G, Zhu X, Hui F, Yang X, Nie X, Zhang X, Jin S. CRISPR/dCas13(Rx) Derived RNA N 6-methyladenosine (m 6A) Dynamic Modification in Plant. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401118. [PMID: 39229923 PMCID: PMC11497087 DOI: 10.1002/advs.202401118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/15/2024] [Indexed: 09/05/2024]
Abstract
N6-methyladenosine (m6A) is the most prevalent internal modification of mRNA and plays an important role in regulating plant growth. However, there is still a lack of effective tools to precisely modify m6A sites of individual transcripts in plants. Here, programmable m6A editing tools are developed by combining CRISPR/dCas13(Rx) with the methyltransferase GhMTA (Targeted RNA Methylation Editor, TME) or the demethyltransferase GhALKBH10 (Targeted RNA Demethylation Editor, TDE). These editors enable efficient deposition or removal of m6A modifications at targeted sites of endo-transcripts GhECA1 and GhDi19 within a broad editing window ranging from 0 to 46 nt. TDE editor significantly decreases m6A levels by 24%-76%, while the TME editor increases m6A enrichment, ranging from 1.37- to 2.51-fold. Furthermore, installation and removal of m6A modifications play opposing roles in regulating GhECA1 and GhDi19 mRNA transcripts, which may be attributed to the fact that their m6A sites are located in different regions of the genes. Most importantly, targeting the GhDi19 transcript with TME editor plants results in a significant increase in root length and enhanced drought resistance. Collectively, these m6A editors can be applied to study the function of specific m6A modifications and have the potential for future applications in crop improvement.
Collapse
Affiliation(s)
- Lu Yu
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Muna Alariqi
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Baoqi Li
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Amjad Hussain
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Huifang Zhou
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Qiongqiong Wang
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Fuqiu Wang
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Guanying Wang
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Xiangqian Zhu
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Fengjiao Hui
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Xiyan Yang
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Xinhui Nie
- Key Laboratory of Oasis Eco‐agriculturalXinjiang Production and Construction Corps/Agricultural CollegeShihezi UniversityShihezi832003China
| | - Xianlong Zhang
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| | - Shuangxia Jin
- National Key Laboratory of Crop Genetic ImprovementHuazhong Agricultural UniversityWuhan430070China
| |
Collapse
|
36
|
Lau CH, Liang QL, Zhu H. Next-generation CRISPR technology for genome, epigenome and mitochondrial editing. Transgenic Res 2024; 33:323-357. [PMID: 39158822 DOI: 10.1007/s11248-024-00404-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
The application of rapidly growing CRISPR toolboxes and methods has great potential to transform biomedical research. Here, we provide a snapshot of up-to-date CRISPR toolboxes, then critically discuss the promises and hurdles associated with CRISPR-based nuclear genome editing, epigenome editing, and mitochondrial editing. The technical challenges and key solutions to realize epigenome editing in vivo, in vivo base editing and prime editing, mitochondrial editing in complex tissues and animals, and CRISPR-associated transposases and integrases in targeted genomic integration of very large DNA payloads are discussed. Lastly, we discuss the latest situation of the CRISPR/Cas9 clinical trials and provide perspectives on CRISPR-based gene therapy. Apart from technical shortcomings, ethical and societal considerations for CRISPR applications in human therapeutics and research are extensively highlighted.
Collapse
Affiliation(s)
- Cia-Hin Lau
- Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, China
| | - Qing-Le Liang
- Department of Clinical Laboratory Medicine, Chongqing University Jiangjin Hospital, Chongqing, China
| | - Haibao Zhu
- Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, China.
| |
Collapse
|
37
|
Yang W, Zhao Y, Yang Y. Dynamic RNA methylation modifications and their regulatory role in mammalian development and diseases. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2084-2104. [PMID: 38833084 DOI: 10.1007/s11427-023-2526-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/15/2023] [Indexed: 06/06/2024]
Abstract
Among over 170 different types of chemical modifications on RNA nucleobases identified so far, RNA methylation is the major type of epitranscriptomic modifications existing on almost all types of RNAs, and has been demonstrated to participate in the entire process of RNA metabolism, including transcription, pre-mRNA alternative splicing and maturation, mRNA nucleus export, mRNA degradation and stabilization, mRNA translation. Attributing to the development of high-throughput detection technologies and the identification of both dynamic regulators and recognition proteins, mechanisms of RNA methylation modification in regulating the normal development of the organism as well as various disease occurrence and developmental abnormalities upon RNA methylation dysregulation have become increasingly clear. Here, we particularly focus on three types of RNA methylations: N6-methylcytosine (m6A), 5-methylcytosine (m5C), and N7-methyladenosine (m7G). We summarize the elements related to their dynamic installment and removal, specific binding proteins, and the development of high-throughput detection technologies. Then, for a comprehensive understanding of their biological significance, we also overview the latest knowledge on the underlying mechanisms and key roles of these three mRNA methylation modifications in gametogenesis, embryonic development, immune system development, as well as disease and tumor progression.
Collapse
Affiliation(s)
- Wenlan Yang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
| | - Yongliang Zhao
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
| | - Yungui Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- China National Center for Bioinformation, Beijing, 100101, China.
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 101408, China.
| |
Collapse
|
38
|
Luo Z, Yu L, Xu Z, Liu K, Gu L. Comprehensive Review and Assessment of Computational Methods for Prediction of N6-Methyladenosine Sites. BIOLOGY 2024; 13:777. [PMID: 39452086 PMCID: PMC11504118 DOI: 10.3390/biology13100777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
N6-methyladenosine (m6A) plays a crucial regulatory role in the control of cellular functions and gene expression. Recent advances in sequencing techniques for transcriptome-wide m6A mapping have accelerated the accumulation of m6A site information at a single-nucleotide level, providing more high-confidence training data to develop computational approaches for m6A site prediction. However, it is still a major challenge to precisely predict m6A sites using in silico approaches. To advance the computational support for m6A site identification, here, we curated 13 up-to-date benchmark datasets from nine different species (i.e., H. sapiens, M. musculus, Rat, S. cerevisiae, Zebrafish, A. thaliana, Pig, Rhesus, and Chimpanzee). This will assist the research community in conducting an unbiased evaluation of alternative approaches and support future research on m6A modification. We revisited 52 computational approaches published since 2015 for m6A site identification, including 30 traditional machine learning-based, 14 deep learning-based, and 8 ensemble learning-based methods. We comprehensively reviewed these computational approaches in terms of their training datasets, calculated features, computational methodologies, performance evaluation strategy, and webserver/software usability. Using these benchmark datasets, we benchmarked nine predictors with available online websites or stand-alone software and assessed their prediction performance. We found that deep learning and traditional machine learning approaches generally outperformed scoring function-based approaches. In summary, the curated benchmark dataset repository and the systematic assessment in this study serve to inform the design and implementation of state-of-the-art computational approaches for m6A identification and facilitate more rigorous comparisons of new methods in the future.
Collapse
Affiliation(s)
- Zhengtao Luo
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei 230036, China;
- Anhui Provincial Key Laboratory of Smart Agriculture Technology and Equipment, Anhui Agricultural University, Hefei 230036, China
| | - Liyi Yu
- Computer Department, Jingdezhen Ceramic University, Jingdezhen 333403, China; (L.Y.); (Z.X.)
| | - Zhaochun Xu
- Computer Department, Jingdezhen Ceramic University, Jingdezhen 333403, China; (L.Y.); (Z.X.)
- School for Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin 150076, China
| | - Kening Liu
- Computer Department, Jingdezhen Ceramic University, Jingdezhen 333403, China; (L.Y.); (Z.X.)
| | - Lichuan Gu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei 230036, China;
- Anhui Provincial Key Laboratory of Smart Agriculture Technology and Equipment, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
39
|
Zhu G, Zhou X, Wen M, Qiao J, Li G, Yao Y. CRISPR-Cas13: Pioneering RNA Editing for Nucleic Acid Therapeutics. BIODESIGN RESEARCH 2024; 6:0041. [PMID: 39228750 PMCID: PMC11371277 DOI: 10.34133/bdr.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/15/2024] [Indexed: 09/05/2024] Open
Abstract
The CRISPR-Cas13 system has emerged as a revolutionary tool for RNA editing, offering new opportunities for the development of nucleic acid therapeutics. Unlike DNA-targeting CRISPR-Cas9, Cas13 targets and cleaves RNA, enabling gene silencing and preventing genomic instability. Its applications include suppressing disease-causing genes, correcting splicing errors, and modulating immune responses. Despite these advances, challenges persist, such as the need to refine specificity, mitigate off-target impacts, and ensure effective delivery. This review provides an overview of the CRISPR-Cas13 mechanism, elucidating its role in RNA-targeted therapies and its transformative potential for disease treatment. Furthermore, it addresses the ongoing challenges that the scientific community is striving to overcome.
Collapse
Affiliation(s)
- Guanglin Zhu
- School of Chemical Engineering and Technology,
Tianjin University, Tianjin 300072, China
| | - Xinzhi Zhou
- ZJU-Hangzhou Global Scientific and Technological Innovation Center,
Zhejiang University, Hangzhou, Zhejiang 311200, China
- College of Chemical and Biological Engineering,
Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Mingzhang Wen
- School of Chemical Engineering and Technology,
Tianjin University, Tianjin 300072, China
- Zhejiang Institute of Tianjin University, Shaoxing 312300, China
- Frontiers Science Center for Synthetic Biology (Ministry of Education),
Tianjin University, Tianjin 300072, P. R. China
| | - Jianjun Qiao
- School of Chemical Engineering and Technology,
Tianjin University, Tianjin 300072, China
- Zhejiang Institute of Tianjin University, Shaoxing 312300, China
- Frontiers Science Center for Synthetic Biology (Ministry of Education),
Tianjin University, Tianjin 300072, P. R. China
| | - Guo Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Center,
Zhejiang University, Hangzhou, Zhejiang 311200, China
- College of Chemical and Biological Engineering,
Zhejiang University, Hangzhou, Zhejiang 310027, China
- Xianghu Laboratory, Hangzhou 311231, China
| | - Yuan Yao
- ZJU-Hangzhou Global Scientific and Technological Innovation Center,
Zhejiang University, Hangzhou, Zhejiang 311200, China
- College of Chemical and Biological Engineering,
Zhejiang University, Hangzhou, Zhejiang 310027, China
- Zhejiang Institute of Tianjin University, Shaoxing 312300, China
| |
Collapse
|
40
|
Xu GE, Zhao X, Li G, Gokulnath P, Wang L, Xiao J. The landscape of epigenetic regulation and therapeutic application of N 6-methyladenosine modifications in non-coding RNAs. Genes Dis 2024; 11:101045. [PMID: 38988321 PMCID: PMC11233902 DOI: 10.1016/j.gendis.2023.06.015] [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: 11/17/2022] [Revised: 05/18/2023] [Accepted: 06/04/2023] [Indexed: 07/12/2024] Open
Abstract
RNA N6-methyladenosine (m6A) methylation is the most abundant and conserved RNA modification in eukaryotes. It participates in the regulation of RNA metabolism and various pathophysiological processes. Non-coding RNAs (ncRNAs) are defined as small or long transcripts which do not encode proteins and display numerous biological regulatory functions. Similar to mRNAs, m6A deposition is observed in ncRNAs. Studying RNA m6A modifications on ncRNAs is of great importance specifically to deepen our understanding of their biological roles and clinical implications. In this review, we summarized the recent research findings regarding the mutual regulation between RNA m6A modification and ncRNAs (with a specific focus on microRNAs, long non-coding RNAs, and circular RNAs) and their functions. We also discussed the challenges of m6A-containing ncRNAs and RNA m6A as therapeutic targets in human diseases and their future perspective in translational roles.
Collapse
Affiliation(s)
- Gui-E Xu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Sciences, Shanghai University, Nantong, Jiangsu 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xuan Zhao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Sciences, Shanghai University, Nantong, Jiangsu 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Priyanka Gokulnath
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Lijun Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Sciences, Shanghai University, Nantong, Jiangsu 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Sciences, Shanghai University, Nantong, Jiangsu 226011, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
41
|
Marayati BF, Thompson MG, Holley CL, Horner SM, Meyer KD. Programmable protein expression using a genetically encoded m 6A sensor. Nat Biotechnol 2024; 42:1417-1428. [PMID: 38168988 PMCID: PMC11217150 DOI: 10.1038/s41587-023-01978-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/01/2023] [Indexed: 01/05/2024]
Abstract
The N6-methyladenosine (m6A) modification is found in thousands of cellular mRNAs and is a critical regulator of gene expression and cellular physiology. m6A dysregulation contributes to several human diseases, and the m6A methyltransferase machinery has emerged as a promising therapeutic target. However, current methods for studying m6A require RNA isolation and do not provide a real-time readout of mRNA methylation in living cells. Here we present a genetically encoded m6A sensor (GEMS) technology, which couples a fluorescent signal with cellular mRNA methylation. GEMS detects changes in m6A caused by pharmacological inhibition of the m6A methyltransferase, giving it potential utility for drug discovery efforts. Additionally, GEMS can be programmed to achieve m6A-dependent delivery of custom protein payloads in cells. Thus, GEMS is a versatile platform for m6A sensing that provides both a simple readout for m6A methylation and a system for m6A-coupled protein expression.
Collapse
Affiliation(s)
- Bahjat F Marayati
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Matthew G Thompson
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Christopher L Holley
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Stacy M Horner
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Kate D Meyer
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA.
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
42
|
Chen D, Gu X, Nurzat Y, Xu L, Li X, Wu L, Jiao H, Gao P, Zhu X, Yan D, Li S, Xue C. Writers, readers, and erasers RNA modifications and drug resistance in cancer. Mol Cancer 2024; 23:178. [PMID: 39215288 PMCID: PMC11363509 DOI: 10.1186/s12943-024-02089-6] [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: 07/03/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Drug resistance in cancer cells significantly diminishes treatment efficacy, leading to recurrence and metastasis. A critical factor contributing to this resistance is the epigenetic alteration of gene expression via RNA modifications, such as N6-methyladenosine (m6A), N1-methyladenosine (m1A), 5-methylcytosine (m5C), 7-methylguanosine (m7G), pseudouridine (Ψ), and adenosine-to-inosine (A-to-I) editing. These modifications are pivotal in regulating RNA splicing, translation, transport, degradation, and stability. Governed by "writers," "readers," and "erasers," RNA modifications impact numerous biological processes and cancer progression, including cell proliferation, stemness, autophagy, invasion, and apoptosis. Aberrant RNA modifications can lead to drug resistance and adverse outcomes in various cancers. Thus, targeting RNA modification regulators offers a promising strategy for overcoming drug resistance and enhancing treatment efficacy. This review consolidates recent research on the role of prevalent RNA modifications in cancer drug resistance, with a focus on m6A, m1A, m5C, m7G, Ψ, and A-to-I editing. Additionally, it examines the regulatory mechanisms of RNA modifications linked to drug resistance in cancer and underscores the existing limitations in this field.
Collapse
Affiliation(s)
- Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yeltai Nurzat
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lixia Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xueyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Lixin Wu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Henan Jiao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Peng Gao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xuqiang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Shaohua Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
43
|
Liu C, Dou X, Zhao Y, Zhang L, Zhang L, Dai Q, Liu J, Wu T, Xiao Y, He C. IGF2BP3 promotes mRNA degradation through internal m 7G modification. Nat Commun 2024; 15:7421. [PMID: 39198433 PMCID: PMC11358264 DOI: 10.1038/s41467-024-51634-w] [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: 08/24/2023] [Accepted: 08/11/2024] [Indexed: 09/01/2024] Open
Abstract
Recent studies have suggested that mRNA internal m7G and its writer protein METTL1 are closely related to cell metabolism and cancer regulation. Here, we identify that IGF2BP family proteins IGF2BP1-3 can preferentially bind internal mRNA m7G. Such interactions, especially IGF2BP3 with m7G, could promote the degradation of m7G target transcripts in cancer cells. IGF2BP3 is more responsive to changes of m7G modification, while IGF2BP1 prefers m6A to stabilize the bound transcripts. We also demonstrate that p53 transcript, TP53, is m7G-modified at its 3'UTR in cancer cells. In glioblastoma, the methylation level and the half lifetime of the modified transcript could be modulated by tuning IGF2BP3, or by site-specific targeting of m7G through a dCas13b-guided system, resulting in modulation of cancer progression and chemosensitivity.
Collapse
Affiliation(s)
- Chang Liu
- 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
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Xiaoyang Dou
- 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
| | - 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
| | - Linda Zhang
- 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
| | - Lisheng Zhang
- 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
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Qing Dai
- 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
| | - Jun Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, 100871, China
| | - Tong Wu
- 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
| | - Yu Xiao
- 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
|
44
|
Roth GV, Gengaro IR, Qi LS. Precision epigenetic editing: Technological advances, enduring challenges, and therapeutic applications. Cell Chem Biol 2024; 31:S2451-9456(24)00309-X. [PMID: 39137782 PMCID: PMC11799355 DOI: 10.1016/j.chembiol.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/31/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024]
Abstract
The epigenome is a complex framework through which gene expression is precisely and flexibly modulated to incorporate heritable memory and responses to environmental stimuli. It governs diverse cellular processes, including cell fate, disease, and aging. The need to understand this system and precisely control gene expression outputs for therapeutic purposes has precipitated the development of a diverse set of epigenetic editing tools. Here, we review the existing toolbox for targeted epigenetic editing, technical considerations of the current technologies, and opportunities for future development. We describe applications of therapeutic epigenetic editing and their potential for treating disease, with a discussion of ongoing delivery challenges that impede certain clinical interventions, particularly in the brain. With simultaneous advancements in available engineering tools and appropriate delivery technologies, we predict that epigenetic editing will increasingly cement itself as a powerful approach for safely treating a wide range of disorders in all tissues of the body.
Collapse
Affiliation(s)
- Goldie V Roth
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Isabella R Gengaro
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA; Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Lei S Qi
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
45
|
Cheng Y, Zhou Y, Wang M. Targeted gene regulation through epigenome editing in plants. CURRENT OPINION IN PLANT BIOLOGY 2024; 80:102552. [PMID: 38776571 DOI: 10.1016/j.pbi.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
The precise targeted gene regulation in plants is essential for improving plant traits and gaining a comprehensive understanding of gene functions. The regulation of gene expression in eukaryotes can be achieved through transcriptional and epigenetic mechanisms. Over the last decade, advancements in gene-targeting technologies, along with an expanded understanding of epigenetic gene regulation mechanisms, have significantly contributed to the development of programmable gene regulation tools. In this review, we will discuss the recent progress in targeted plant gene regulation through epigenome editing, emphasizing the role of effector proteins in modulating target gene expression via diverse mechanisms, including DNA methylation, histone modifications, and chromatin remodeling. Additionally, we will also briefly review targeted gene regulation by transcriptional regulation and mRNA modifications in plants.
Collapse
Affiliation(s)
- Yuejing Cheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Zhou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ming Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
46
|
Mehravar M, Wong JJL. Interplay between N 6-adenosine RNA methylation and mRNA splicing. Curr Opin Genet Dev 2024; 87:102211. [PMID: 38838495 DOI: 10.1016/j.gde.2024.102211] [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: 02/24/2024] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/07/2024]
Abstract
N6-methyladenosine (m6A) is the most abundant modification to mRNAs. Loss-of-function studies of main m6A regulators have indicated the role of m6A in pre-mRNA splicing. Recent studies have reported the role of splicing in preventing m6A deposition. Understanding the interplay between m6A and mRNA splicing holds the potential to clarify the significance of these fundamental molecular mechanisms in cell development and function, thereby shedding light on their involvement in the pathogenesis of myriad diseases.
Collapse
Affiliation(s)
- Majid Mehravar
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
| | - Justin J-L Wong
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia.
| |
Collapse
|
47
|
Li X, Li Y, Zhang W, Jiang F, Lin L, Wang Y, Wu L, Zeng H, Zheng J. The IGF2BP3/Notch/Jag1 pathway: A key regulator of hepatic stellate cell ferroptosis in liver fibrosis. Clin Transl Med 2024; 14:e1793. [PMID: 39113232 PMCID: PMC11306284 DOI: 10.1002/ctm2.1793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/11/2024] Open
Abstract
INTRODUCTION Liver fibrosis is primarily driven by the activation of hepatic stellate cells (HSCs), which involves various epigenetic modifications. OBJECTIVES N6-methyladenosine (m6A), the most prevalent RNA modification in eukaryotic cells, influences numerous physiological and pathological processes. Nevertheless, the role of insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), a reader gene mediating m6A modifications, in liver fibrosis remains unclear. METHODS AND RESULTS This study demonstrated that IGF2BP3 knockout reduces liver fibrosis by promoting HSC ferroptosis (FPT) and inactivating HSCs. Multi-omics analysis revealed that HSC-specific IGF2BP3 knockout decreased m6A content in Jagged1 (Jag1), a key component of the Notch signalling pathway. Furthermore, IGF2BP3 deficiency significantly reduced the expression of hairy and enhancer of split-1 (Hes1), a transcription factor in the Notch/Jag1 signalling pathway, with mRNA levels declining to 35%-62% and protein levels to 28%-35%. Additionally, it suppressed glutathione peroxidase 4 (GPX4) (decreased to approximately 31%-38%), a negative regulator of FPT, thereby facilitating HSC FPT progression and reducing profibrotic gene expression. CONCLUSION These findings uncover a novel IGF2BP3/Notch/Jag1 signalling pathway involving HSC FPT, suggesting promising targets for ameliorating liver fibrosis. KEY POINTS/HIGHLIGHTS IGF2BP3 deficiency inactivates Jag1 signalling. IGF2BP3 deficiency-mediated m6A modifications promote HSC ferroptosis. IGF2BP3 inhibition facilitates ferroptosis in HSCs via the Hes1/GPX4 axis. IGF2BP3 deficiency inactivates Jag1/Notch1/3/Hes1 signalling pathway inactivation, leading to the decrease in GPX4, which contributes to HSC ferroptosis.
Collapse
Affiliation(s)
- Xinmiao Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yifei Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Weizhi Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Feng Jiang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Lifan Lin
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yining Wang
- School of Mental HealthWenzhou Medical UniversityWenzhouChina
| | - Lingling Wu
- Renji CollegeWenzhou Medical UniversityWenzhouChina
| | - Han Zeng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jianjian Zheng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and TranslationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
48
|
Sighel D, Destefanis E, Quattrone A. Therapeutic strategies to target the epitranscriptomic machinery. Curr Opin Genet Dev 2024; 87:102230. [PMID: 39024774 DOI: 10.1016/j.gde.2024.102230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
Altered RNA modification patterns and dysregulated expression of epitranscriptomic machinery proteins (EMPs) have been causatively correlated with several diseases. Modulation of EMP gene expression has shown promise in reversing disease-associated phenotypes, making EMPs attractive therapeutic targets. Various therapeutic strategies, including small-molecule modulators, proteolysis-targeting chimeras, and molecular tools for site-specific engineering of RNA modifications, have been introduced to modulate EMPs and RNA modifications themselves and are currently being investigated to enrich the physician's armamentarium. At the forefront of research are small-molecule inhibitors of the key players involved in the N6-methyladenosine RNA modification, with an inhibitor of methyltransferase 3 in clinical trials. Preclinical studies have also demonstrated proof-of-concept for the other approaches, raising expectations for this exciting new frontier of therapy.
Collapse
Affiliation(s)
- Denise Sighel
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy. https://twitter.com/@DSighel
| | - Eliana Destefanis
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy. https://twitter.com/@Destefanis_E
| | - Alessandro Quattrone
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy.
| |
Collapse
|
49
|
Sun M, Ren J, Qu X. In situ bioorthogonal-modulation of m 6A RNA methylation in macrophages for efficient eradication of intracellular bacteria. Chem Sci 2024; 15:11657-11666. [PMID: 39055012 PMCID: PMC11268468 DOI: 10.1039/d4sc03629h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
N6-Methyladenosine (m6A) methylation plays a critical role in controlling the RNA fate. Emerging evidence has demonstrated that aberrant m6A methylation in immune cells such as macrophages could alter cell homeostasis and function, which can be a promising target for disease treatment. Despite tremendous progress in regulating the level of m6A methylation, the current methods suffer from the time-consuming operation and annoying off-target effect, which hampers the in situ manipulation of m6A methylation. Here, a bioorthogonal in situ modulation strategy of m6A methylation was proposed. Well-designed covalent organic framework (COF) dots (CIDM) could deprotect the agonist prodrug of m6A methyltransferase, resulting in a considerable hypermethylation of m6A modification. Simultaneously, the bioorthogonal catalyst CIDM showed oxidase (OXD)-mimic activity that further promoted the level of m6A methylation. Ultimately, the potential therapeutic effect of bioorthogonal controllable regulation of m6A methylation was demonstrated through intracellular bacteria eradication. The remarkable antimicrobial outcomes indicate that upregulating m6A methylation in macrophages could reprogram them into the M1 phenotype with high bactericidal activity. We believe that our bioorthogonal chemistry-controlled epigenetics regulatory strategy will provide a unique insight into the development of controllable m6A methylation.
Collapse
Affiliation(s)
- Mengyu Sun
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230029 China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230029 China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 China
- University of Science and Technology of China Hefei Anhui 230029 China
| |
Collapse
|
50
|
Shi C, Zou W, Liu X, Zhang H, Li X, Fu G, Fei Q, Qian Q, Shang L. Programmable RNA N 6-methyladenosine editing with CRISPR/dCas13a in plants. PLANT BIOTECHNOLOGY JOURNAL 2024; 22:1867-1880. [PMID: 38363049 PMCID: PMC11182597 DOI: 10.1111/pbi.14307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/07/2023] [Accepted: 01/26/2024] [Indexed: 02/17/2024]
Abstract
N6-methyladenonsine (m6A) is the most prevalent internal modification of messenger RNA (mRNA) and plays critical roles in mRNA processing and metabolism. However, perturbation of individual m6A modification to reveal its function and the phenotypic effects is still lacking in plants. Here, we describe the construction and characterization of programmable m6A editing tools by fusing the m6A writers, the core catalytic domain of the MTA and MTB complex, and the AlkB homologue 5 (ALKBH5) eraser, to catalytically dead Cas13a (dCas13a) to edit individual m6A sites on mRNAs. We demonstrated that our m6A editors could efficiently and specifically deposit and remove m6A modifications on specific RNA transcripts in both Nicotiana benthamiana and Arabidopsis thaliana. Moreover, we found that targeting SHORT-ROOT (SHR) transcripts with a methylation editor could significantly increase its m6A levels with limited off-target effects and promote its degradation. This leads to a boost in plant growth with enlarged leaves and roots, increased plant height, plant biomass, and total grain weight in Arabidopsis. Collectively, these findings suggest that our programmable m6A editing tools can be applied to study the functions of individual m6A modifications in plants, and may also have potential applications for future crop improvement.
Collapse
Affiliation(s)
- Chuanlin Shi
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural SciencesShenzhenChina
| | - Wenli Zou
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural SciencesShenzhenChina
| | - Xiangpei Liu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural SciencesShenzhenChina
| | - Hong Zhang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural SciencesShenzhenChina
| | - Xiaofang Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural SciencesShenzhenChina
- Zhengzhou Research Base, State Key Laboratory of Cotton BiologyZhengzhou UniversityZhengzhouChina
| | - Guiling Fu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural SciencesShenzhenChina
- College of AgricultureShanxi Agricultural UniversityTaiyuanShanxiChina
| | - Qili Fei
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural SciencesShenzhenChina
| | - Qian Qian
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural SciencesShenzhenChina
- State Key Laboratory of Rice BiologyChina National Rice Research InstituteHangzhouZhejiangChina
- Yazhouwan National LaboratorySanya CityHainan ProvinceChina
| | - Lianguang Shang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural AffairsAgricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural SciencesShenzhenChina
- Yazhouwan National LaboratorySanya CityHainan ProvinceChina
| |
Collapse
|