1
|
García-Vílchez R, Guallar D. Interplay of transposable elements and ageing: epigenetic regulation and potential epitranscriptomic influence. Curr Opin Genet Dev 2025; 92:102331. [PMID: 40101544 DOI: 10.1016/j.gde.2025.102331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/16/2025] [Accepted: 02/20/2025] [Indexed: 03/20/2025]
Abstract
Transposable elements (TEs) are mobile elements, which have been crucial for mammalian genome evolution and function. Their activity, which influences genomic stability, gene expression and chromatin state, is tightly regulated by complex mechanisms. This review examines recent findings on TE regulation and the dynamics and connection during the ageing process. Here, we explore the interplay between chromatin state, DNA, RNA, and histone modifications in controlling TE activity, with a special emphasis in elucidating the emerging role of epitranscriptomic modifications in TE regulation. Additionally, we analyse the connection between TE activation and ageing, with the perspective for future research that could reveal novel targets for alleviating physiological and pathological ageing and age-related diseases.
Collapse
Affiliation(s)
- Raquel García-Vílchez
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Barcelona Avenue s/n, Santiago de Compostela, A Coruña 15782, Spain. https://twitter.com/@raquelgarcv
| | - Diana Guallar
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Barcelona Avenue s/n, Santiago de Compostela, A Coruña 15782, Spain.
| |
Collapse
|
2
|
Artz O, White J, Rousseau B, Argiles G, Foote M, Johannet P, Patel M, Abdelfattah S, Patel S, Wilde C, Mieles D, Diaz L. The role of recurrent somatic mutations that alter conserved m 6A motifs in human cancer. NAR Cancer 2025; 7:zcaf014. [PMID: 40271220 PMCID: PMC12015683 DOI: 10.1093/narcan/zcaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/19/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025] Open
Abstract
N6-methyladenosine (m6A) is the most abundant internal RNA modification in eukaryotes and plays a key role in cellular growth and development. Global changes in cellular methylated RNA and m6A-mediated transcript regulation significantly impact oncogenesis. Here, we investigate how recurrent synonymous and non-synonymous somatic mutations abolishing individual canonical methylated m6A motifs affect transcript levels and survival of patients with cancer. Moreover, we explore the effect of these mutations on creating de novo m6A motifs. To this end, we compared publicly available data on m6A sites with mutations reported in The Cancer Genome Atlas (TCGA). We find that mutations disrupting or creating m6A motifs display a low recurrence and have a negligible impact on RNA abundance. Patients with the highest number of disrupted m6A sites or newly generated m6A motifs did not generally exhibit alterations in mortality risk or outcomes. Hence, our data suggest that mutational alterations in the m6A motif landscape are unlikely to be a primary mechanism for regulating gene function across most cancer types. This may be attributed to the fact that mutations typically affect individual m6A sites, which is likely insufficient to significantly impact gene expression.
Collapse
Affiliation(s)
- Oliver Artz
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - James R White
- Resphera Biosciences, Baltimore, MD 21231, United States
| | - Benoit Rousseau
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - Guillem Argiles
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - Michael B Foote
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - Paul Johannet
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - Miteshkumar Patel
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - Somer Abdelfattah
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - Shrey Patel
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - Callahan Wilde
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - David Mieles
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| | - Luis A Diaz
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering, New York City, NY 10065, United States
| |
Collapse
|
3
|
Sun H, Lu B, Zhang Z, Xiao Y, Zhou Z, Xi L, Li Z, Jiang Z, Zhang J, Wang M, Liu C, Ma Y, Peng J, Wang XJ, Yi C. Mild and ultrafast GLORI enables absolute quantification of m 6A methylome from low-input samples. Nat Methods 2025:10.1038/s41592-025-02680-9. [PMID: 40325216 DOI: 10.1038/s41592-025-02680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 03/24/2025] [Indexed: 05/07/2025]
Abstract
Methods for absolute quantification of N6-methyladenosine (m6A) have emerged as powerful tools in epitranscriptomics. We previously reported GLORI, a chemical-assisted approach to achieve unbiased and precise m6A measurement. However, its lengthy reaction time and severe RNA degradation have limited its applicability, particularly for low-input samples. Here, we present two updated GLORI approaches that are ultrafast, mild and enable absolute m6A quantification from one to two orders of magnitude less than the RNA starting material: GLORI 2.0 is compatible with RNA from ~10,000 cells and enhances sensitivity for both transcriptome-wide and locus-specific m6A detection; GLORI 3.0 further utilizes a reverse transcription-silent carrier RNA to achieve m6A quantification from as low as 500-1,000 cells. Using limited RNA from mouse dorsal hippocampus, we reveal a high modification level in synapse-related gene sets. We envision that the updated GLORI methods will greatly expand the applicability of absolute quantification of m6A in biology.
Collapse
Affiliation(s)
- Hanxiao Sun
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Bo Lu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Zeyu Zhang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Ye Xiao
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhe Zhou
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Lin Xi
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Zhichao Li
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Zhe Jiang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Jiayi Zhang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Meng Wang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Cong Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Yichen Ma
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Jinying Peng
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Xiu-Jie Wang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, China.
| | - Chengqi Yi
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- Department of Chemical Biology and Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China.
| |
Collapse
|
4
|
VanInsberghe M, van Oudenaarden A. Sequencing technologies to measure translation in single cells. Nat Rev Mol Cell Biol 2025; 26:337-346. [PMID: 39833532 DOI: 10.1038/s41580-024-00822-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/22/2025]
Abstract
Translation is one of the most energy-intensive processes in a cell and, accordingly, is tightly regulated. Genome-wide methods to measure translation and the translatome and to study the complex regulation of protein synthesis have enabled unprecedented characterization of this crucial step of gene expression. However, technological limitations have hampered our understanding of translation control in multicellular tissues, rare cell types and dynamic cellular processes. Recent optimizations, adaptations and new techniques have enabled these measurements to be made at single-cell resolution. In this Progress, we discuss single-cell sequencing technologies to measure translation, including ribosome profiling, ribosome affinity purification and spatial translatome methods.
Collapse
Affiliation(s)
- Michael VanInsberghe
- Oncode Institute, Utrecht, the Netherlands.
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, the Netherlands.
- University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Alexander van Oudenaarden
- Oncode Institute, Utrecht, the Netherlands
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, the Netherlands
- University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
5
|
Ali H, Wadas J, Bendoumou M, Chen HC, Maiuri P, Dutilleul A, Selberg S, Nestola L, Lalik K, Avettand-Fenoël V, Necsoi C, Marcello A, Kankuri E, Karelson M, De Wit S, Pyrc K, Pasternak AO, Van Lint C, Kula-Pacurar A. Inhibition of ALKBH5 demethylase of m 6A pathway potentiates HIV-1 reactivation from latency. Virol J 2025; 22:124. [PMID: 40296171 PMCID: PMC12039216 DOI: 10.1186/s12985-025-02744-4] [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: 11/04/2024] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Current latency-reversing agents (LRAs) employed in the "shock-and-kill" strategy primarily focus on relieving epigenetic and transcriptional blocks to reactivate the latent HIV-1. However, their clinical efficacy is limited, partly due to their inability to fully reverse latency and the lack of LRAs specifically targeting post-transcriptional mechanisms. N6-methyladenosine (m6A) modification in HIV-1 RNA is emerging as an important post-transcriptional regulator of HIV-1 gene expression, yet its role in latency and reactivation remains largely unrecognized. Here, we explored the potential of small chemical compounds targeting the m6A pathway, specifically investigating the inhibition of ALKBH5 and its effect on latent HIV-1 reactivation mediated by the LRA romidepsin. METHODS We used four in vitro cellular models of latency, primary model of CD4+ T cells HIV-1 infection and ex vivo cultures of CD8+-depleted PMBCs from ART-treated HIV+ patients. We measured latent viral reactivation by evaluating the expression of reporter protein GFP by flow cytometry, viral production by CA-p24 ELISA, and viral transcripts by RT-qPCR. CRISPR/Cas9 method was used to deplete ALKBH5. MeRIP and immuno-RNA FISH were used to address the m6A methylation levels on HIV-1 RNA upon ALKBH5 inhibition. RESULTS We showed that ALKBH5 inhibitor 3 (ALKi-3) potentiated romidepsin-mediated viral reactivation in in vitro models of latency, primary model of CD4+ T cells infected with HIV-1 as well as in ex vivo cultures of CD8+-depleted PBMCs from ART-treated HIV+ patients. CRISPR/Cas9-mediated depletion of ALKBH5 mimicked the effects of ALKi-3. ALKi-3 increased levels of m6A-methylated HIV-1 RNA as shown by meRIP and immuno-RNA FISH. CONCLUSION Our study provides a proof-of-concept for the modulation of the m6A pathway in enhancing HIV-1 reactivation. This approach represents a promising adjunct to existing reactivation protocols and provides a concept of "dual-kick", aiming to target transcriptional and post-transcriptional steps in HIV-1 reactivation from latency.
Collapse
Affiliation(s)
- Haider Ali
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Jakub Wadas
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Maryam Bendoumou
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Heng-Chang Chen
- Quantitative Virology Research Group, Population Diagnostics Center, Lukasiewicz Research Network- PORT Polish Center for Technology Development, Wroclaw, Poland
- The Laboratory of Quantitative Virology, Centre for Advanced Materials and Technologies, Warsaw University of Technology, 19 Poleczki St, Warsaw, 02-822, Poland
| | - Paolo Maiuri
- Dept of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Antoine Dutilleul
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Simona Selberg
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Lorena Nestola
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Kamil Lalik
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Veronique Avettand-Fenoël
- Université Paris Cité, INSERM U1016, CNRS UMR8104, Institut Cochin, Paris, France
- CHU d'Orléans, Orléans, France
| | - Coca Necsoi
- Service des Maladies Infectieuses, CHU St-Pierre, Université Libre de Bruxelles (ULB), Brussels, 1000, Belgium
| | - Alessandro Marcello
- Laboratory of Molecular Virology, The International Centre of Genetic Engineering and Biotechnology, Trieste, Italy
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Mati Karelson
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Stéphane De Wit
- Service des Maladies Infectieuses, CHU St-Pierre, Université Libre de Bruxelles (ULB), Brussels, 1000, Belgium
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Alexander O Pasternak
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Carine Van Lint
- Service of Molecular Virology, Department of Molecular Biology (DBM), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Anna Kula-Pacurar
- Laboratory of Molecular Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
6
|
Artika IM, Arianti R, Demény MÁ, Kristóf E. RNA modifications and their role in gene expression. Front Mol Biosci 2025; 12:1537861. [PMID: 40351534 PMCID: PMC12061695 DOI: 10.3389/fmolb.2025.1537861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 04/02/2025] [Indexed: 05/14/2025] Open
Abstract
Post-transcriptional RNA modifications have recently emerged as critical regulators of gene expression programs. Understanding normal tissue development and disease susceptibility requires knowledge of the various cellular mechanisms which control gene expression in multicellular organisms. Research into how different RNA modifications such as in N6-methyladenosine (m6A), inosine (I), 5-methylcytosine (m5C), pseudouridine (Ψ), 5-hydroxymethylcytosine (hm5C), N1-methyladenosine (m1A), N6,2'-O-dimethyladenosine (m6Am), 2'-O-methylation (Nm), N7-methylguanosine (m7G) etc. affect the expression of genes could be valuable. This review highlights the current understanding of RNA modification, methods used to study RNA modification, types of RNA modification, and molecular mechanisms underlying RNA modification. The role of RNA modification in modulating gene expression in both physiological and diseased states is discussed. The potential applications of RNA modification in therapeutic development are elucidated.
Collapse
Affiliation(s)
- I. Made Artika
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Bogor, Indonesia
| | - Rini Arianti
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Universitas Muhammadiyah Bangka Belitung, Pangkalpinang, Indonesia
| | - Máté Á. Demény
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Endre Kristóf
- Laboratory of Cell Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
7
|
Wei S, Tao HY, Duan Z, Wang Y. Environmental Exposure, Epitranscriptomic Perturbations, and Human Diseases. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:6387-6399. [PMID: 40126397 PMCID: PMC11978485 DOI: 10.1021/acs.est.5c00907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Epitranscriptomics is a rapidly evolving field, and it examines how chemical modifications on RNA regulate gene expression. Increasing lines of evidence support that exposure to various environmental agents can change substantially chemical modifications on RNA, thereby perturbing gene expression and contributing to disease development in humans. However, the molecular mechanisms through which environmental exposure impairs RNA modification-associated proteins ("reader", "writer", and "eraser" or RWE proteins) and alters the landscape of RNA modifications remain poorly understood. Here, we provide our perspectives on the current knowledge about how environmental exposure alters the epitranscriptome, where we focus on dynamic changes in RNA modifications and their regulatory proteins elicited by exposure to environmental agents. We discuss how these epitranscriptomic alterations may contribute to the development of human diseases, especially neurodegeneration and cancer. We also discuss the potential and technical challenges of harnessing RNA modifications as biomarkers for monitoring environmental exposure. Finally, we emphasize the need to integrate multiomics approaches to decipher the complex interplay between environmental exposure and the epitranscriptome and offer a forward-looking viewpoint on future research priorities that may inform public health interventions and environmental regulations.
Collapse
Affiliation(s)
- Songbo Wei
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Huan-Yu Tao
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Zheng Duan
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, California 92521-0403, United States
| |
Collapse
|
8
|
Yang L, Ma M, Gao Y, Liu J. Decoding N 6-methyladenosine's dynamic role in stem cell fate and early embryo development: insights into RNA-chromatin interactions. Curr Opin Genet Dev 2025; 91:102311. [PMID: 39908649 DOI: 10.1016/j.gde.2025.102311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
N6-methyladenosine (m6A), a reversible and dynamic RNA modification, plays pivotal roles in regulating stem cell pluripotency and early embryogenesis. Disruptions in m6A homeostasis lead to profound developmental defects, impairing processes such as stem cell self-renewal, lineage specification, oocyte maturation, zygotic genome activation, and maternal RNA degradation after fertilization. Beyond its well-recognized roles in mRNA transport, stability, and translation, recent studies have highlighted m6A's critical role in transcriptional regulation through intricate RNA-chromatin interactions, notably involving chromatin-associated regulatory RNAs (carRNAs) and retrotransposon RNAs. This review delves into the dynamic regulatory landscape of m6A, highlighting its critical interplay with chromatin modifications, and explores its broader implications in stem cell biology and early embryonic development.
Collapse
Affiliation(s)
- Lei Yang
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Mingli Ma
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yawei Gao
- State Key Laboratory of Cardiology and Medical Innovation Center, Department of Reproductive Medicine Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Sycamore Research Institute of Life Sciences, Shanghai 201203, China.
| | - Jun Liu
- State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China.
| |
Collapse
|
9
|
Cai Z, Song P, Yu K, Jia G. Advanced reactivity-based sequencing methods for mRNA epitranscriptome profiling. RSC Chem Biol 2025; 6:150-169. [PMID: 39759443 PMCID: PMC11694185 DOI: 10.1039/d4cb00215f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Currently, over 170 chemical modifications identified in RNA introduce an additional regulatory attribute to gene expression, known as the epitranscriptome. The development of detection methods to pinpoint the location and quantify these dynamic and reversible modifications has significantly expanded our understanding of their roles. This review goes deep into the latest progress in enzyme- and chemical-assisted sequencing methods, highlighting the opportunities presented by these reactivity-based techniques for detailed characterization of RNA modifications. Our survey provides a deeper understanding of the function and biological roles of RNA modification.
Collapse
Affiliation(s)
- Zhihe Cai
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
| | - Peizhe Song
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
| | - Kemiao Yu
- Peking-Tsinghua Center for Life Sciences, Peking University Beijing 100871 China
| | - Guifang Jia
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
- Peking-Tsinghua Center for Life Sciences, Peking University Beijing 100871 China
- Beijing Advanced Center of RNA Biology, Peking University Beijing 100871 China
| |
Collapse
|
10
|
Li P, Lin Y, Ma H, Zhang J, Zhang Q, Yan R, Fan Y. Epigenetic regulation in female reproduction: the impact of m6A on maternal-fetal health. Cell Death Discov 2025; 11:43. [PMID: 39904996 PMCID: PMC11794895 DOI: 10.1038/s41420-025-02324-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/09/2025] [Accepted: 01/24/2025] [Indexed: 02/06/2025] Open
Abstract
With the development of public health, female diseases have become the focus of current concern. The unique reproductive anatomy of women leads to the development of gynecological diseases gradually become an important part of the socio-economic burden. Epigenetics plays an irreplaceable role in gynecologic diseases. As an important mRNA modification, m6A is involved in the maturation of ovum cells and maternal-fetal microenvironment. At present, researchers have found that m6A is involved in the regulation of gestational diabetes and other reproductive system diseases, but the specific mechanism is not clear. In this manuscript, we summarize the components of m6A, the biological function of m6A, the progression of m6A in the maternal-fetal microenvironment and a variety of gynecological diseases as well as the progression of targeted m6A treatment-related diseases, providing a new perspective for clinical treatment-related diseases.
Collapse
Affiliation(s)
- Peipei Li
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hongyun Ma
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Jiao Zhang
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Qiaorui Zhang
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Ruihua Yan
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Yang Fan
- Department of Obstetrics and Gynecology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China.
| |
Collapse
|
11
|
Xu M, Liu X, Wang M, Luo T, Gao Y, Liu J, Shi J. BASAL: a universal mapping algorithm for nucleotide base-conversion sequencing. Nucleic Acids Res 2025; 53:gkae1201. [PMID: 39658059 PMCID: PMC11754667 DOI: 10.1093/nar/gkae1201] [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: 06/06/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Utilizing base-conversion (BC) techniques, single-base resolution profiling of RNA and DNA modifications has significantly advanced. BC strategies range from one-way conversions (e.g. cytosine-to-thymine for 5-methylcytosine, adenine-to-guanine for N6-methyladenosine), to multi-way conversions (e.g. adenine to cytosine/guanine/thymine for N1-methyladenosine) and deletion-induced conversions (e.g. pseudouridine-to-deletion). Existing sequence aligners struggle with these diverse conversions, often leading to misaligning or inefficiency. We introduce BASAL (BAse-conversion Sequencing ALigner), which leverages bit-masking technology to accurately calculate mismatch penalties and supports all BC strategies. BASAL outperforms state-of-the-art tools in both mapping accuracy and efficiency. Through simulated and real data testing, along with experimental validation, we demonstrate that BASAL excels at identifying reliable modification sites. Moreover, BASAL enhances single-cell m6A analysis, revealing cell subpopulations and a cell evolutionary direction that align with biological functions, which other aligners fall short. BASAL's versatility establishes it as a universal aligner for RNA and DNA modification sequencing, facilitating groundbreaking discoveries in epigenomics and epitranscriptomics.
Collapse
Affiliation(s)
- Moping Xu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Xiaoyang Liu
- State Key Laboratory of Protein and Plant Gene Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | - Miao Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Tingting Luo
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Yawei Gao
- Department of Reproductive Medicine Center, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Jun Liu
- State Key Laboratory of Protein and Plant Gene Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, 5 Yiheyuan Road, Beijing 100871, China
| | - Jiejun Shi
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, 1239 Siping Road, Shanghai 200092, China
| |
Collapse
|
12
|
Catacalos-Goad C, Chakrabarti M, Salem DH, Camporeale C, Somalraju S, Tegowski M, Singh R, Reid RW, Janies DA, Meyer KD, Janga SC, Hunt AG, Chakrabarti K. Nucleotide-resolution Mapping of RNA N6-Methyladenosine (m6A) modifications and comprehensive analysis of global polyadenylation events in mRNA 3' end processing in malaria pathogen Plasmodium falciparum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631827. [PMID: 39829786 PMCID: PMC11741415 DOI: 10.1101/2025.01.07.631827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Plasmodium falciparum is an obligate human parasite of the phylum Apicomplexa and is the causative agent of the most lethal form of human malaria. Although N6-methyladenosine modification is thought to be one of the major post-transcriptional regulatory mechanisms for stage-specific gene expression in apicomplexan parasites, the precise base position of m6A in mRNAs or noncoding RNAs in these parasites remains unknown. Here, we report global nucleotide-resolution mapping of m6A residues in P. falciparum using DART-seq technology, which quantitatively displayed a stage-specific, dynamic distribution pattern with enrichment near mRNA 3' ends. In this process we identified 894, 788, and 1,762 m6A-modified genes in Ring, Trophozoite and Schizont stages respectively, with an average of 5-7 m6A sites per-transcript at the individual gene level. Notably, several genes involved in malaria pathophysiology, such as KAHRP, ETRAMPs, SERA and stress response genes, such as members of Heat Shock Protein (HSP) family are highly enriched in m6A and therefore could be regulated by this RNA modification. Since we observed preferential methylation at the 3' ends of P. falciparum transcripts and because malaria polyadenylation specificity factor PfCPSF30 harbors an m6A reader 'YTH' domain, we reasoned that m6A might play an important role in 3'-end processing of malaria mRNAs. To investigate this, we used two complementary high-throughput RNA 3'-end mapping approaches, which provided an initial framework to explore potential roles of m6A in the regulation of alternative polyadenylation (APA) during malaria development in human hosts.
Collapse
Affiliation(s)
- Cassandra Catacalos-Goad
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA, United States of America
| | - Manohar Chakrabarti
- School of Integrative Biological and Chemical Sciences, University of Texas Rio Grande Valley, Edinburg, TX
| | - Doaa Hassan Salem
- Department of Biomedical Engineering and Informatics, Luddy School of Informatics, Computing and Engineering, Indiana University Indianapolis (IUI), Indianapolis, Indiana, United States of America
| | - Carli Camporeale
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA, United States of America
| | - Sahiti Somalraju
- Department of Biomedical Engineering and Informatics, Luddy School of Informatics, Computing and Engineering, Indiana University Indianapolis (IUI), Indianapolis, Indiana, United States of America
| | - Matthew Tegowski
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Ruchi Singh
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA, United States of America
| | - Robert W Reid
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Daniel A Janies
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Kate D Meyer
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sarath Chandra Janga
- Department of Biomedical Engineering and Informatics, Luddy School of Informatics, Computing and Engineering, Indiana University Indianapolis (IUI), Indianapolis, Indiana, United States of America
| | - Arthur G Hunt
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA, United States of America
| | - Kausik Chakrabarti
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA, United States of America
| |
Collapse
|
13
|
Wang H, Wang Y, Zhou J, Song B, Tu G, Nguyen A, Su J, Coenen F, Wei Z, Rigden DJ, Meng J. Statistical modeling of single-cell epitranscriptomics enabled trajectory and regulatory inference of RNA methylation. CELL GENOMICS 2025; 5:100702. [PMID: 39642887 PMCID: PMC11770222 DOI: 10.1016/j.xgen.2024.100702] [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: 05/15/2024] [Revised: 10/07/2024] [Accepted: 11/06/2024] [Indexed: 12/09/2024]
Abstract
As a fundamental mechanism for gene expression regulation, post-transcriptional RNA methylation plays versatile roles in various biological processes and disease mechanisms. Recent advances in single-cell technology have enabled simultaneous profiling of transcriptome-wide RNA methylation in thousands of cells, holding the promise to provide deeper insights into the dynamics, functions, and regulation of RNA methylation. However, it remains a major challenge to determine how to best analyze single-cell epitranscriptomics data. In this study, we developed SigRM, a computational framework for effectively mining single-cell epitranscriptomics datasets with a large cell number, such as those produced by the scDART-seq technique from the SMART-seq2 platform. SigRM not only outperforms state-of-the-art models in RNA methylation site detection on both simulated and real datasets but also provides rigorous quantification metrics of RNA methylation levels. This facilitates various downstream analyses, including trajectory inference and regulatory network reconstruction concerning the dynamics of RNA methylation.
Collapse
Affiliation(s)
- Haozhe Wang
- Department of Biosciences and Bioinformatics, Center for Intelligent RNA Therapeutics, Suzhou Key Laboratory of Cancer Biology and Chronic Disease, School of Science, XJTLU Entrepreneur College, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China; Department of Computer Science, University of Liverpool, L7 8TX Liverpool, UK
| | - Yue Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| | - Jingxian Zhou
- School of AI and Advanced Computing, XJTLU Entrepreneur College, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China; Department of Computer Science, University of Liverpool, L7 8TX Liverpool, UK; Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Bowen Song
- Department of Public Health, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Gang Tu
- Department of Biosciences and Bioinformatics, Center for Intelligent RNA Therapeutics, Suzhou Key Laboratory of Cancer Biology and Chronic Disease, School of Science, XJTLU Entrepreneur College, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Anh Nguyen
- Department of Computer Science, University of Liverpool, L7 8TX Liverpool, UK
| | - Jionglong Su
- School of AI and Advanced Computing, XJTLU Entrepreneur College, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Frans Coenen
- Department of Computer Science, University of Liverpool, L7 8TX Liverpool, UK
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Daniel J Rigden
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L7 8TX Liverpool, UK
| | - Jia Meng
- Department of Biosciences and Bioinformatics, Center for Intelligent RNA Therapeutics, Suzhou Key Laboratory of Cancer Biology and Chronic Disease, School of Science, XJTLU Entrepreneur College, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China; Institute of Biomedical Research, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei Provincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L7 8TX Liverpool, UK.
| |
Collapse
|
14
|
Sun F, Li H, Sun D, Fu S, Gu L, Shao X, Wang Q, Dong X, Duan B, Xing F, Wu J, Xiao M, Zhao F, Han JDJ, Liu Q, Fan X, Li C, Wang C, Shi T. Single-cell omics: experimental workflow, data analyses and applications. SCIENCE CHINA. LIFE SCIENCES 2025; 68:5-102. [PMID: 39060615 DOI: 10.1007/s11427-023-2561-0] [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: 12/07/2023] [Accepted: 04/18/2024] [Indexed: 07/28/2024]
Abstract
Cells are the fundamental units of biological systems and exhibit unique development trajectories and molecular features. Our exploration of how the genomes orchestrate the formation and maintenance of each cell, and control the cellular phenotypes of various organismsis, is both captivating and intricate. Since the inception of the first single-cell RNA technology, technologies related to single-cell sequencing have experienced rapid advancements in recent years. These technologies have expanded horizontally to include single-cell genome, epigenome, proteome, and metabolome, while vertically, they have progressed to integrate multiple omics data and incorporate additional information such as spatial scRNA-seq and CRISPR screening. Single-cell omics represent a groundbreaking advancement in the biomedical field, offering profound insights into the understanding of complex diseases, including cancers. Here, we comprehensively summarize recent advances in single-cell omics technologies, with a specific focus on the methodology section. This overview aims to guide researchers in selecting appropriate methods for single-cell sequencing and related data analysis.
Collapse
Affiliation(s)
- Fengying Sun
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China
| | - Haoyan Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dongqing Sun
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shaliu Fu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Lei Gu
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Shao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China
| | - Qinqin Wang
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bin Duan
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Feiyang Xing
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Minmin Xiao
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China.
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Chen Li
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Tieliu Shi
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, School of Statistics, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
15
|
Ron K, Kahn J, Malka‐Tunitsky N, Sas‐Chen A. High-throughput detection of RNA modifications at single base resolution. FEBS Lett 2025; 599:19-32. [PMID: 39543833 PMCID: PMC11726149 DOI: 10.1002/1873-3468.15052] [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: 06/26/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
RNA is modified by > 170 chemical modifications that affect its structure and function. Accordingly, RNA modifications have been implicated in regulation of gene expression and cellular outcomes in a variety of species spanning the phylogenetic tree. The study of RNA modifications is accelerated by generation of high-throughput methods for detecting RNA modifications at single base resolution. Here, we review recent advancement in next generation sequencing based approaches for detection of 14 distinct RNA modifications present in rRNA, tRNA and mRNA. We further outline the molecular and computational principles underlying currently available methods.
Collapse
MESH Headings
- High-Throughput Nucleotide Sequencing/methods
- RNA Processing, Post-Transcriptional
- RNA, Transfer/genetics
- RNA, Transfer/metabolism
- RNA, Transfer/chemistry
- Humans
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Messenger/chemistry
- RNA/genetics
- RNA/metabolism
- RNA/chemistry
- RNA, Ribosomal/genetics
- RNA, Ribosomal/metabolism
- RNA, Ribosomal/chemistry
- Animals
- Sequence Analysis, RNA/methods
Collapse
Affiliation(s)
- Keren Ron
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life SciencesTel Aviv UniversityIsrael
| | - Joshua Kahn
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life SciencesTel Aviv UniversityIsrael
| | - Nofar Malka‐Tunitsky
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life SciencesTel Aviv UniversityIsrael
| | - Aldema Sas‐Chen
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty of Life SciencesTel Aviv UniversityIsrael
| |
Collapse
|
16
|
Gokhale NS, Sam RK, Somfleth K, Thompson MG, Marciniak DM, Smith JR, Genoyer E, Eggenberger J, Chu LH, Park M, Dvorkin S, Oberst A, Horner SM, Ong SE, Gale M, Savan R. Cellular RNA interacts with MAVS to promote antiviral signaling. Science 2024; 386:eadl0429. [PMID: 39700280 PMCID: PMC11905950 DOI: 10.1126/science.adl0429] [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: 09/25/2023] [Revised: 08/12/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
Antiviral signaling downstream of RIG-I-like receptors (RLRs) proceeds through a multi-protein complex organized around the adaptor protein mitochondrial antiviral signaling protein (MAVS). Protein complex function can be modulated by RNA molecules that provide allosteric regulation or act as molecular guides or scaffolds. We hypothesized that RNA plays a role in organizing MAVS signaling platforms. We found that MAVS, through its central intrinsically disordered domain, directly interacted with the 3' untranslated regions of cellular messenger RNAs. Elimination of RNA by ribonuclease treatment disrupted the MAVS signalosome, including RNA-modulated MAVS interactors that regulate RLR signaling and viral restriction, and inhibited phosphorylation of transcription factors that induce interferons. This work uncovered a function for cellular RNA in promoting signaling through MAVS and highlights generalizable principles of RNA regulatory control of immune signaling complexes.
Collapse
Affiliation(s)
| | - Russell K. Sam
- Department of Immunology, University of Washington, Seattle, WA
| | - Kim Somfleth
- Department of Immunology, University of Washington, Seattle, WA
| | | | | | - Julian R. Smith
- Department of Immunology, University of Washington, Seattle, WA
| | | | | | - Lan H. Chu
- Department of Immunology, University of Washington, Seattle, WA
| | - Moonhee Park
- Department of Integrative Immunobiology, Duke University, Durham, NC
| | - Steve Dvorkin
- Department of Immunology, University of Washington, Seattle, WA
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA
| | - Stacy M. Horner
- Department of Integrative Immunobiology, Duke University, Durham, NC
- Department of Medicine, Duke University, Durham NC
| | - Shao-En Ong
- Department of Pharmacology, University of Washington, Seattle, WA
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA
| | - Ram Savan
- Department of Immunology, University of Washington, Seattle, WA
| |
Collapse
|
17
|
Mitsuhashi H, Lin R, Chawla A, Mechawar N, Nagy C, Turecki G. Altered m6A RNA methylation profiles in depression implicate the dysregulation of discrete cellular functions in males and females. iScience 2024; 27:111316. [PMID: 39650737 PMCID: PMC11625292 DOI: 10.1016/j.isci.2024.111316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/03/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
Adverse environmental stress represents a significant risk factor for major depressive disorder (MDD), often resulting in disrupted synaptic connectivity which is known to be partly regulated by epigenetic mechanisms. N6-methyladenosine (m6A), an epitranscriptomic modification, has emerged as a crucial regulator of activity-dependent gene regulation. In this study, we characterized m6A profiles in the ventromedial prefrontal cortex (vmPFC) of individuals with MDD. Using m6A sequencing, we identified a total of 30,279 high-confidence m6A peaks, exhibiting significant enrichment in genes related to neuronal and synaptic function. The m6A peaks between males and females with MDD that passed the significance threshold showed opposite m6A patterns, while the threshold-free m6A patterns were concordant. Distinct m6A profiles were found in MDD for each sex, with dysregulation associated with microtubule movement in males and neuronal projection in females. Our results suggest the potential roles of m6A as part of the dysregulated molecular network in MDD.
Collapse
Affiliation(s)
- Haruka Mitsuhashi
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Rixing Lin
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544 USA, USA
| | - Anjali Chawla
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
18
|
Louwagie A, Vu LP. Emerging interactions between RNA methylation and chromatin architecture. Curr Opin Genet Dev 2024; 89:102270. [PMID: 39426116 DOI: 10.1016/j.gde.2024.102270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024]
Abstract
Epitranscriptomics, the study of chemical modifications of RNA molecules, is increasingly recognized as an important component of gene expression regulation. While the majority of research has focused on N6-methyladenosine (m6A) RNA methylation on mRNAs, emerging evidence has revealed that the m6A modification extends beyond mRNAs to include chromatin-associated RNAs (caRNAs). CaRNAs constitute an important class of RNAs characterized by their interaction with the genome and epigenome. These features allow caRNAs to be actively involved in shaping genome organization. In this review, we bring into focus recent findings of the dynamic interactions between caRNAs and chromatin architecture and how RNA methylation impacts caRNAs' function in this interplay. We highlight several enabling techniques, which were critical for genome-wide profiling of caRNAs and their modifications. Given the nascent stage of the field, we emphasize on the need to address critical gaps in study of these modifications in more relevant biological systems. Overall, these exciting progress have expanded the scope and reach of epitranscriptomics, unveiling new mechanisms that underpin the control of gene expression and cellular phenotypes, with potential therapeutic implications.
Collapse
Affiliation(s)
- Amber Louwagie
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada; Terry Fox Laboratory, British Columbia Cancer Research Centre, Vancouver, Canada
| | - Ly P Vu
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada; Terry Fox Laboratory, British Columbia Cancer Research Centre, Vancouver, Canada.
| |
Collapse
|
19
|
Cui X, Li H, Huang X, Xue T, Wang S, Zhu X, Jing X. N 6-Methyladenosine Modification on the Function of Female Reproductive Development and Related Diseases. Immun Inflamm Dis 2024; 12:e70089. [PMID: 39660878 PMCID: PMC11632877 DOI: 10.1002/iid3.70089] [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: 08/18/2024] [Revised: 10/17/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) modification is a widespread and reversible epigenetic alteration in eukaryotic mRNA, playing a pivotal role in various biological functions. Its significance in female reproductive development and associated diseases has recently become a focal point of research. OBJECTIVE This review aims to consolidate current knowledge of the role of m6A modification in female reproductive tissues, emphasizing its regulatory dynamics, functional significance, and implications in reproductive health and disease. METHODS A comprehensive analysis of recent studies focusing on m6A modification in ovarian development, oocyte maturation, embryo development, and the pathogenesis of reproductive diseases. RESULTS m6A modification exhibits dynamic regulation in female reproductive tissues, influencing key developmental stages and processes. It plays critical roles in ovarian development, oocyte maturation, and embryo development, underpinning essential aspects of reproductive health. m6A modification is intricately involved in the pathogenesis of several reproductive diseases, including polycystic ovary syndrome (PCOS), premature ovarian failure (POF), and endometriosis, offering insights into potential molecular mechanisms and therapeutic targets. CONCLUSION The review highlights the crucial role of m6A modification in female reproductive development and related diseases. It underscores the need for further research to explore innovative diagnostic and therapeutic strategies for reproductive disorders, leveraging the insights gained from understanding m6A modification's impact on reproductive health.
Collapse
Affiliation(s)
- Xiangrong Cui
- Reproductive Medicine CenterThe affiliated Children's Hospital of Shanxi Medical University, Children's Hospital of Shanxi, Shanxi Maternal and Child Health HospitalTaiyuanChina
| | - Huihui Li
- Reproductive Medicine CenterThe affiliated Children's Hospital of Shanxi Medical University, Children's Hospital of Shanxi, Shanxi Maternal and Child Health HospitalTaiyuanChina
| | - Xia Huang
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Tingting Xue
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Shu Wang
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Xinyu Zhu
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| | - Xuan Jing
- Department of Clinical LaboratoryShanxi Provincial People's Hospital, Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
20
|
Tegowski M, Prater AK, Holley CL, Meyer KD. Single-cell m 6A profiling in the mouse brain uncovers cell type-specific RNA methylomes and age-dependent differential methylation. Nat Neurosci 2024; 27:2512-2520. [PMID: 39317796 PMCID: PMC11614689 DOI: 10.1038/s41593-024-01768-3] [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: 07/21/2023] [Accepted: 08/19/2024] [Indexed: 09/26/2024]
Abstract
N6-methyladenosine (m6A) is an abundant mRNA modification in the brain that has important roles in neurodevelopment and brain function. However, because of technical limitations, global profiling of m6A sites within the individual cell types that make up the brain has not been possible. Here, we develop a mouse model that enables transcriptome-wide m6A detection in any tissue of interest at single-cell resolution. We use these mice to map m6A across different brain regions and within single cells of the mouse cortex and discover a high degree of shared methylation across brain regions and cell types. However, we also identify a small number of differentially methylated mRNAs in neurons that encode important regulators of neuronal signaling, and we discover that microglia have lower levels of m6A than other cell types. Finally, we perform single-cell m6A mapping in aged mice and identify many transcripts with age-dependent changes in m6A.
Collapse
Affiliation(s)
- Matthew Tegowski
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Anna K Prater
- Department of Biochemistry, 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
| | - 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
|
21
|
Xiang JS, Schafer DM, Rothamel KL, Yeo GW. Decoding protein-RNA interactions using CLIP-based methodologies. Nat Rev Genet 2024; 25:879-895. [PMID: 38982239 DOI: 10.1038/s41576-024-00749-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 07/11/2024]
Abstract
Protein-RNA interactions are central to all RNA processing events, with pivotal roles in the regulation of gene expression and cellular functions. Dysregulation of these interactions has been increasingly linked to the pathogenesis of human diseases. High-throughput approaches to identify RNA-binding proteins and their binding sites on RNA - in particular, ultraviolet crosslinking followed by immunoprecipitation (CLIP) - have helped to map the RNA interactome, yielding transcriptome-wide protein-RNA atlases that have contributed to key mechanistic insights into gene expression and gene-regulatory networks. Here, we review these recent advances, explore the effects of cellular context on RNA binding, and discuss how these insights are shaping our understanding of cellular biology. We also review the potential therapeutic applications arising from new knowledge of protein-RNA interactions.
Collapse
Affiliation(s)
- Joy S Xiang
- Division of Biomedical Sciences, UC Riverside, Riverside, CA, USA
| | - Danielle M Schafer
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute and Stem Cell Program, UC San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, UC San Diego, La Jolla, CA, USA
| | - Katherine L Rothamel
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute and Stem Cell Program, UC San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, UC San Diego, La Jolla, CA, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, USA.
- Sanford Stem Cell Institute and Stem Cell Program, UC San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, UC San Diego, La Jolla, CA, USA.
- Sanford Laboratories for Innovative Medicines, La Jolla, CA, USA.
| |
Collapse
|
22
|
Zou Y, Guo Z, Ge XY, Qiu Y. RNA Modifications in Pathogenic Viruses: Existence, Mechanism, and Impacts. Microorganisms 2024; 12:2373. [PMID: 39597761 PMCID: PMC11596894 DOI: 10.3390/microorganisms12112373] [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: 10/29/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
RNA modification is a key posttranscriptional process playing various biological roles, and one which has been reported to exist extensively in cellular RNAs. Interestingly, recent studies have shown that viral RNAs also contain a variety of RNA modifications, which are regulated dynamically by host modification machinery and play critical roles in different stages of the viral life cycle. In this review, we summarize the reports of four typical modifications reported on viral RNAs, including N6-methyladenosine (m6A), 5-methylcytosine (m5C), N4-acetylcytosine (ac4C), and N1-methyladenosine (m1A), describe the molecular mechanisms of these modification processes, and illustrate their impacts on viral replication, pathogenicity, and innate immune responses. Notably, we find that RNA modifications in different viruses share some common features and mechanisms in their generation, regulation, and function, highlighting the potential for viral RNA modifications and the related host machinery to serve as the targets or bases for the development of antiviral therapeutics and vaccines.
Collapse
Affiliation(s)
| | | | - Xing-Yi Ge
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410012, China; (Y.Z.); (Z.G.)
| | - Ye Qiu
- Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha 410012, China; (Y.Z.); (Z.G.)
| |
Collapse
|
23
|
Wang S, Zeng Y, Zhu L, Zhang M, Zhou L, Yang W, Luo W, Wang L, Liu Y, Zhu H, Xu X, Su P, Zhang X, Ahmed M, Chen W, Chen M, Chen S, Slobodyanyuk M, Xie Z, Guan J, Zhang W, Khan AA, Sakashita S, Liu N, Pham NA, Boutros PC, Ke Z, Moran MF, Cai Z, Cheng C, Yu J, Tsao MS, He HH. The N6-methyladenosine Epitranscriptomic Landscape of Lung Adenocarcinoma. Cancer Discov 2024; 14:2279-2299. [PMID: 38922581 PMCID: PMC11528209 DOI: 10.1158/2159-8290.cd-23-1212] [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: 10/13/2023] [Revised: 04/25/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
Comprehensive N6-methyladenosine (m6A) epitranscriptomic profiling of primary tumors remains largely uncharted. Here, we profiled the m6A epitranscriptome of 10 nonneoplastic lung tissues and 51 lung adenocarcinoma (LUAD) tumors, integrating the corresponding transcriptomic, proteomic, and extensive clinical annotations. We identified distinct clusters and genes that were exclusively linked to disease progression through m6A modifications. In comparison with nonneoplastic lung tissues, we identified 430 transcripts to be hypo-methylated and 222 to be hyper-methylated in tumors. Among these genes, EML4 emerged as a novel metastatic driver, displaying significant hypermethylation in tumors. m6A modification promoted the translation of EML4, leading to its widespread overexpression in primary tumors. Functionally, EML4 modulated cytoskeleton dynamics by interacting with ARPC1A, enhancing lamellipodia formation, cellular motility, local invasion, and metastasis. Clinically, high EML4 protein abundance correlated with features of metastasis. METTL3 small-molecule inhibitor markedly diminished both EML4 m6A and protein abundance and efficiently suppressed lung metastases in vivo. Significance: Our study reveals a dynamic and functional epitranscriptomic landscape in LUAD, offering a valuable resource for further research in the field. We identified EML4 hypermethylation as a key driver of tumor metastasis, highlighting a novel therapeutic strategy of targeting EML4 to prevent LUAD metastasis.
Collapse
Affiliation(s)
- Shiyan Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yong Zeng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Lin Zhu
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Min Zhang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhou
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weixiong Yang
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weishan Luo
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lina Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanming Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Helen Zhu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute, Toronto, Canada
| | - Xin Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Peiran Su
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Xinyue Zhang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Musaddeque Ahmed
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Wei Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People’s Hospital of Nanjing Medical University, Huai’an, China
| | - Moliang Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Sujun Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Mykhaylo Slobodyanyuk
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Ontario Institute of Cancer Research, Toronto, Canada
| | - Zhongpeng Xie
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiansheng Guan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- College of Electrical Engineering and Automation, Xiamen University of Technology, Xiamen, China
| | - Wen Zhang
- Program in Cell Biology, Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | | | - Shingo Sakashita
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Ni Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Nhu-An Pham
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, California
- Department of Urology, University of California, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, California
| | - Zunfu Ke
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Michael F. Moran
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Program in Cell Biology, Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Chao Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Ming S. Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Housheng H. He
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
24
|
Li Y, Li J, Li W, Liang S, Wei W, Chu J, Lai J, Lin Y, Chen H, Su J, Hu X, Wang G, Meng J, Jiang J, Ye L, An S. Scm6A: A Fast and Low-cost Method for Quantifying m6A Modifications at the Single-cell Level. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae039. [PMID: 39436235 PMCID: PMC12016562 DOI: 10.1093/gpbjnl/qzae039] [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: 12/23/2022] [Revised: 04/03/2024] [Accepted: 05/24/2024] [Indexed: 10/23/2024]
Abstract
It is widely accepted that N6-methyladenosine (m6A) exhibits significant intercellular specificity, which poses challenges for its detection using existing m6A quantitative methods. In this study, we introduced Single-cell m6A Analysis (Scm6A), a machine learning-based approach for single-cell m6A quantification. Scm6A leverages input features derived from the expression levels of m6A trans regulators and cis sequence features, and offers remarkable prediction efficiency and reliability. To further validate the robustness and precision of Scm6A, we first applied Scm6A to single-cell RNA sequencing (scRNA-seq) data from peripheral blood mononuclear cells (PBMCs) and calculated the m6A levels in CD4+ and CD8+ T cells. We also applied a winscore-based m6A calculation method to conduct N6-methyladenosine sequencing (m6A-seq) analysis on CD4+ and CD8+ T cells isolated through magnetic-activated cell sorting (MACS) from the same samples. Notably, the m6A levels calculated by Scm6A exhibited a significant positive correlation with those quantified through m6A-seq in different cells isolated by MACS, providing compelling evidence for Scm6A's reliability. Additionally, we performed single-cell-level m6A analysis on lung cancer tissues as well as blood samples from patients with coronavirus disease 2019 (COVID-19), and demonstrated the landscape and regulatory mechanisms of m6A in different T cell subtypes from these diseases. In summary, Scm6A is a novel, dependable, and accurate method for single-cell m6A detection and has broad applications in the realm of m6A-related research.
Collapse
Affiliation(s)
- Yueqi Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine Sciences, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Biological Molecular Medicine Research, Education Department of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Jingyi Li
- Department of Pathology, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Wenxing Li
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Shuaiyi Liang
- Department of Bioinformatics, Anjin Biotechnology Co., Ltd., Guangzhou 510000, China
| | - Wudi Wei
- Life Sciences Institute & Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Guangxi Medical University, Nanning 530021, China
| | - Jiemei Chu
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Jingzhen Lai
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Yao Lin
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Hubin Chen
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Jinming Su
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Xiaopeng Hu
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Gang Wang
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Jun Meng
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Junjun Jiang
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Li Ye
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| | - Sanqi An
- Department of Biochemistry and Molecular Biology, School of Basic Medicine Sciences, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Biological Molecular Medicine Research, Education Department of Guangxi Zhuang Autonomous Region, Nanning 530021, China
- Life Sciences Institute & Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
25
|
Ren J, Luo S, Shi H, Wang X. Spatial omics advances for in situ RNA biology. Mol Cell 2024; 84:3737-3757. [PMID: 39270643 PMCID: PMC11455602 DOI: 10.1016/j.molcel.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/07/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024]
Abstract
Spatial regulation of RNA plays a critical role in gene expression regulation and cellular function. Understanding spatially resolved RNA dynamics and translation is vital for bringing new insights into biological processes such as embryonic development, neurobiology, and disease pathology. This review explores past studies in subcellular, cellular, and tissue-level spatial RNA biology driven by diverse methodologies, ranging from cell fractionation, in situ and proximity labeling, imaging, spatially indexed next-generation sequencing (NGS) approaches, and spatially informed computational modeling. Particularly, recent advances have been made for near-genome-scale profiling of RNA and multimodal biomolecules at high spatial resolution. These methods enabled new discoveries into RNA's spatiotemporal kinetics, RNA processing, translation status, and RNA-protein interactions in cells and tissues. The evolving landscape of experimental and computational strategies reveals the complexity and heterogeneity of spatial RNA biology with subcellular resolution, heralding new avenues for RNA biology research.
Collapse
Affiliation(s)
- Jingyi Ren
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shuchen Luo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hailing Shi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Xiao Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
26
|
Xhemalçe B, Miller KM, Gromak N. Epitranscriptome in action: RNA modifications in the DNA damage response. Mol Cell 2024; 84:3610-3626. [PMID: 39366350 PMCID: PMC12044609 DOI: 10.1016/j.molcel.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/20/2024] [Accepted: 09/05/2024] [Indexed: 10/06/2024]
Abstract
Complex pathways involving the DNA damage response (DDR) contend with cell-intrinsic and -extrinsic sources of DNA damage. DDR mis-regulation results in genome instability that can contribute to aging and diseases including cancer and neurodegeneration. Recent studies have highlighted key roles for several RNA species in the DDR, including short RNAs and RNA/DNA hybrids (R-loops) at DNA break sites, all contributing to efficient DNA repair. RNAs can undergo more than 170 distinct chemical modifications. These RNA modifications have emerged as key orchestrators of the DDR. Here, we highlight the function of enzyme- and non-enzyme-induced RNA modifications in the DDR, with particular emphasis on m6A, m5C, and RNA editing. We also discuss stress-induced RNA damage, including RNA alkylation/oxidation, RNA-protein crosslinks, and UV-induced RNA damage. Uncovering molecular mechanisms that underpin the contribution of RNA modifications to DDR and genome stability will have direct application to disease and approaches for therapeutic intervention.
Collapse
Affiliation(s)
- Blerta Xhemalçe
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Kyle M Miller
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Natalia Gromak
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road OX1 3RE, UK.
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
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
|
30
|
Flamand MN, Meyer KD. Simultaneous profiling of the RNA targets of two RNA-binding proteins using TRIBE-STAMP. Methods Enzymol 2024; 705:127-157. [PMID: 39389662 DOI: 10.1016/bs.mie.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
RNA-binding proteins (RBPs) are central players in RNA homeostasis and the control of gene expression. The identification of RBP targets, interactions, and the regulatory networks they control is crucial for understanding their cellular functions. Traditional methods for identifying RBP targets across the transcriptome have been insightful but are limited by their focus on a single RBP at a time and their general inability to identify individual RNA molecules that are bound by RBPs of interest. Recently, we overcame these limitations by developing TRIBE-STAMP, a method which enables concurrent identification of the RNA targets of two RBPs of interest with single-molecule resolution. TRIBE-STAMP works by tagging desired RBPs with either the ADAR or APOBEC1 RNA editing enzymes and expressing them in cells, followed by RNA-seq. Subsequent computational identification of A-to-I and C-to-U editing events enables the simultaneous identification of the ADAR- and APOBEC1-fused RBP target RNAs, respectively. Here, we present a detailed protocol for TRIBE-STAMP, including considerations for fusion protein expression in cells and step-by-step computational analysis of sequencing data. TRIBE-STAMP is a simple and highly versatile approach for single-molecule identification of the targets of RBPs which enables unprecedented insights into the biological interplay between RBP pairs in cells.
Collapse
Affiliation(s)
- Mathieu N Flamand
- Centre de Recherche du CHU de Québec-Université Laval, Axe Neurosciences, Québec, QC, Canada; Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec, QC, Canada.
| | - Kate D Meyer
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, United States; Department of Neurobiology, Duke University School of Medicine, Durham, NC, United States; Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
31
|
Joshi P, Keyvani Chahi A, Liu L, Moreira S, Vujovic A, Hope KJ. RNA binding protein-directed control of leukemic stem cell evolution and function. Hemasphere 2024; 8:e116. [PMID: 39175825 PMCID: PMC11339706 DOI: 10.1002/hem3.116] [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: 01/23/2024] [Revised: 05/06/2024] [Accepted: 05/26/2024] [Indexed: 08/24/2024] Open
Abstract
Strict control over hematopoietic stem cell decision making is essential for healthy life-long blood production and underpins the origins of hematopoietic diseases. Acute myeloid leukemia (AML) in particular is a devastating hematopoietic malignancy that arises from the clonal evolution of disease-initiating primitive cells which acquire compounding genetic changes over time and culminate in the generation of leukemic stem cells (LSCs). Understanding the molecular underpinnings of these driver cells throughout their development will be instrumental in the interception of leukemia, the enabling of effective treatment of pre-leukemic conditions, as well as the development of strategies to target frank AML disease. To this point, a number of precancerous myeloid disorders and age-related alterations are proving as instructive models to gain insights into the initiation of LSCs. Here, we explore this myeloid dysregulation at the level of post-transcriptional control, where RNA-binding proteins (RBPs) function as core effectors. Through regulating the interplay of a myriad of RNA metabolic processes, RBPs orchestrate transcript fates to govern gene expression in health and disease. We describe the expanding appreciation of the role of RBPs and their post-transcriptional networks in sustaining healthy hematopoiesis and their dysregulation in the pathogenesis of clonal myeloid disorders and AML, with a particular emphasis on findings described in human stem cells. Lastly, we discuss key breakthroughs that highlight RBPs and post-transcriptional control as actionable targets for precision therapy of AML.
Collapse
Affiliation(s)
- Pratik Joshi
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Ava Keyvani Chahi
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Lina Liu
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Steven Moreira
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Ana Vujovic
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Kristin J. Hope
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| |
Collapse
|
32
|
He B, Chen Y, Yi C. Quantitative mapping of the mammalian epitranscriptome. Curr Opin Genet Dev 2024; 87:102212. [PMID: 38823337 DOI: 10.1016/j.gde.2024.102212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/04/2024] [Accepted: 05/17/2024] [Indexed: 06/03/2024]
Abstract
The epitranscriptome encompasses a diverse array of dynamic and reversible RNA modifications, affecting both coding and noncoding RNAs. Over 170 types of RNA chemical modifications have been identified, underscoring the need for innovative detection methods to deepen our understanding of RNA modification roles and mechanisms. In particular, the base resolution and quantitative information on RNA modifications are critical for understanding the regulation and functions of RNA modifications. Based on detection throughput and principles, existing quantitative RNA modification detection methods can be categorized into two groups, including next-generation sequencing and nanopore direct RNA sequencing. In this review, we focus on methodologies for elucidating the base resolution and stoichiometric information of RNA modifications. In addition, we further discuss the challenges and the potential prospects of the quantitative RNA modification detection methods.
Collapse
Affiliation(s)
- Bo He
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yuting Chen
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China
| | - Chengqi Yi
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China; Department of Chemical Biology and Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
| |
Collapse
|
33
|
Song R, J Sutton G, Li F, Liu Q, Wong JJL. Variable calling of m6A and associated features in databases: a guide for end-users. Brief Bioinform 2024; 25:bbae434. [PMID: 39258883 PMCID: PMC11388104 DOI: 10.1093/bib/bbae434] [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: 01/09/2024] [Revised: 07/01/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024] Open
Abstract
N6-methyladenosine (m$^{6}$A) is a widely-studied methylation to messenger RNAs, which has been linked to diverse cellular processes and human diseases. Numerous databases that collate m$^{6}$A profiles of distinct cell types have been created to facilitate quick and easy mining of m$^{6}$A signatures associated with cell-specific phenotypes. However, these databases contain inherent complexities that have not been explicitly reported, which may lead to inaccurate identification and interpretation of m$^{6}$A-associated biology by end-users who are unaware of them. Here, we review various m$^{6}$A-related databases, and highlight several critical matters. In particular, differences in peak-calling pipelines across databases drive substantial variability in both peak number and coordinates with only moderate reproducibility, and the inclusion of peak calls from early m$^{6}$A sequencing protocols may lead to the reporting of false positives or negatives. The awareness of these matters will help end-users avoid the inclusion of potentially unreliable data in their studies and better utilize m$^{6}$A databases to derive biologically meaningful results.
Collapse
Affiliation(s)
- Renhua Song
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, The University of Sydney, Camperdown, NSW 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Gavin J Sutton
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, The University of Sydney, Camperdown, NSW 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Fuyi Li
- College of Information Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China
- South Australian immunoGENomics Cancer Institute (SAiGENCI), The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Qian Liu
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, Maryland Pkwy, NV 89154, United States
- School of Life Sciences, College of Sciences, University of Nevada, Las Vegas, Maryland Pkwy, NV 89154, United States
| | - Justin J-L Wong
- Epigenetics and RNA Biology Laboratory, School of Medical Sciences, The University of Sydney, Camperdown, NSW 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
34
|
Feng S, Tellaetxe-Abete M, Zhang Y, Peng Y, Zhou H, Dong M, Larrea E, Xue L, Zhang L, Koziol MJ. Single-cell discovery of m 6A RNA modifications in the hippocampus. Genome Res 2024; 34:822-836. [PMID: 39009472 PMCID: PMC11293556 DOI: 10.1101/gr.278424.123] [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: 08/21/2023] [Accepted: 06/11/2024] [Indexed: 07/17/2024]
Abstract
N 6-Methyladenosine (m6A) is a prevalent and highly regulated RNA modification essential for RNA metabolism and normal brain function. It is particularly important in the hippocampus, where m6A is implicated in neurogenesis and learning. Although extensively studied, its presence in specific cell types remains poorly understood. We investigated m6A in the hippocampus at a single-cell resolution, revealing a comprehensive landscape of m6A modifications within individual cells. Through our analysis, we uncovered transcripts exhibiting a dense m6A profile, notably linked to neurological disorders such as Alzheimer's disease. Our findings suggest a pivotal role of m6A-containing transcripts, particularly in the context of CAMK2A neurons. Overall, this work provides new insights into the molecular mechanisms underlying hippocampal physiology and lays the foundation for future studies investigating the dynamic nature of m6A RNA methylation in the healthy and diseased brain.
Collapse
Affiliation(s)
- Shuangshuang Feng
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
- Chinese Institute for Brain Research, Beijing 102206, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Maitena Tellaetxe-Abete
- Intelligent Systems Group, Computer Science Faculty, University of the Basque Country, Donostia/San Sebastian 20018, Spain
| | - Yujie Zhang
- Chinese Institute for Brain Research, Beijing 102206, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Yan Peng
- Chinese Institute for Brain Research, Beijing 102206, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
- Peking University, Beijing, 100871, China
| | - Han Zhou
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Mingjie Dong
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Erika Larrea
- Chinese Institute for Brain Research, Beijing 102206, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
- Tsinghua University, Beijing 100084, China
| | - Liang Xue
- Chinese Institute for Brain Research, Beijing 102206, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| | - Li Zhang
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Magdalena J Koziol
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China;
- Chinese Institute for Brain Research, Beijing 102206, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| |
Collapse
|
35
|
Xiang Y, Zhang D, Li L, Xue YX, Zhang CY, Meng QF, Wang J, Tan XL, Li YL. Detection, distribution, and functions of RNA N 6-methyladenosine (m 6A) in plant development and environmental signal responses. FRONTIERS IN PLANT SCIENCE 2024; 15:1429011. [PMID: 39081522 PMCID: PMC11286456 DOI: 10.3389/fpls.2024.1429011] [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: 05/07/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024]
Abstract
The epitranscriptomic mark N 6-methyladenosine (m6A) is the most common type of messenger RNA (mRNA) post-transcriptional modification in eukaryotes. With the discovery of the demethylase FTO (FAT MASS AND OBESITY-ASSOCIATED PROTEIN) in Homo Sapiens, this modification has been proven to be dynamically reversible. With technological advances, research on m6A modification in plants also rapidly developed. m6A modification is widely distributed in plants, which is usually enriched near the stop codons and 3'-UTRs, and has conserved modification sequences. The related proteins of m6A modification mainly consist of three components: methyltransferases (writers), demethylases (erasers), and reading proteins (readers). m6A modification mainly regulates the growth and development of plants by modulating the RNA metabolic processes and playing an important role in their responses to environmental signals. In this review, we briefly outline the development of m6A modification detection techniques; comparatively analyze the distribution characteristics of m6A in plants; summarize the methyltransferases, demethylases, and binding proteins related to m6A; elaborate on how m6A modification functions in plant growth, development, and response to environmental signals; and provide a summary and outlook on the research of m6A in plants.
Collapse
|
36
|
Yang Y, Lu Y, Wang Y, Wen X, Qi C, Piao W, Jin H. Current progress in strategies to profile transcriptomic m 6A modifications. Front Cell Dev Biol 2024; 12:1392159. [PMID: 39055651 PMCID: PMC11269109 DOI: 10.3389/fcell.2024.1392159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Various methods have been developed so far for detecting N 6-methyladenosine (m6A). The total m6A level or the m6A status at individual positions on mRNA can be detected and quantified through some sequencing-independent biochemical methods, such as LC/MS, SCARLET, SELECT, and m6A-ELISA. However, the m6A-detection techniques relying on high-throughput sequencing have more effectively advanced the understanding about biological significance of m6A-containing mRNA and m6A pathway at a transcriptomic level over the past decade. Various SGS-based (Second Generation Sequencing-based) methods with different detection principles have been widely employed for this purpose. These principles include m6A-enrichment using antibodies, discrimination of m6A from unmodified A-base by nucleases, a fusion protein strategy relying on RNA-editing enzymes, and marking m6A with chemical/biochemical reactions. Recently, TGS-based (Third Generation Sequencing-based) methods have brought a new trend by direct m6A-detection. This review first gives a brief introduction of current knowledge about m6A biogenesis and function, and then comprehensively describes m6A-profiling strategies including their principles, procedures, and features. This will guide users to pick appropriate methods according to research goals, give insights for developing novel techniques in varying areas, and continue to expand our boundary of knowledge on m6A.
Collapse
Affiliation(s)
- Yuening Yang
- Laboratory of Genetics and Disorders, Key Laboratory of Molecular Medicine and Biotherapy, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yanming Lu
- Laboratory of Genetics and Disorders, Key Laboratory of Molecular Medicine and Biotherapy, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yan Wang
- Laboratory of Genetics and Disorders, Key Laboratory of Molecular Medicine and Biotherapy, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xianghui Wen
- Laboratory of Genetics and Disorders, Key Laboratory of Molecular Medicine and Biotherapy, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Changhai Qi
- Department of Pathology, Aerospace Center Hospital, Beijing, China
| | - Weilan Piao
- Laboratory of Genetics and Disorders, Key Laboratory of Molecular Medicine and Biotherapy, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan, China
| | - Hua Jin
- Laboratory of Genetics and Disorders, Key Laboratory of Molecular Medicine and Biotherapy, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, China
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan, China
| |
Collapse
|
37
|
XIONG J, FENG T, YUAN BF. [Advances in mapping analysis of ribonucleic acid modifications through sequencing]. Se Pu 2024; 42:632-645. [PMID: 38966972 PMCID: PMC11224946 DOI: 10.3724/sp.j.1123.2023.12025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Indexed: 07/06/2024] Open
Abstract
Over 170 chemical modifications have been discovered in various types of ribonucleic acids (RNAs), including messenger RNA (mRNA), ribosomal RNA (rRNA), transfer RNA (tRNA), and small nuclear RNA (snRNA). These RNA modifications play crucial roles in a wide range of biological processes such as gene expression regulation, RNA stability maintenance, and protein translation. RNA modifications represent a new dimension of gene expression regulation known as the "epitranscriptome". The discovery of RNA modifications and the relevant writers, erasers, and readers provides an important basis for studies on the dynamic regulation and physiological functions of RNA modifications. Owing to the development of detection technologies for RNA modifications, studies on RNA epitranscriptomes have progressed to the single-base resolution, multilayer, and full-coverage stage. Transcriptome-wide methods help discover new RNA modification sites and are of great importance for elucidating the molecular regulatory mechanisms of epitranscriptomics, exploring the disease associations of RNA modifications, and understanding their clinical applications. The existing RNA modification sequencing technologies can be categorized according to the pretreatment approach and sequencing principle as direct high-throughput sequencing, antibody-enrichment sequencing, enzyme-assisted sequencing, chemical labeling-assisted sequencing, metabolic labeling sequencing, and nanopore sequencing technologies. These methods, as well as studies on the functions of RNA modifications, have greatly expanded our understanding of epitranscriptomics. In this review, we summarize the recent progress in RNA modification detection technologies, focusing on the basic principles, advantages, and limitations of different methods. Direct high-throughput sequencing methods do not require complex RNA pretreatment and allow for the mapping of RNA modifications using conventional RNA sequencing methods. However, only a few RNA modifications can be analyzed by high-throughput sequencing. Antibody enrichment followed by high-throughput sequencing has emerged as a crucial approach for mapping RNA modifications, significantly advancing the understanding of RNA modifications and their regulatory functions in different species. However, the resolution of antibody-enrichment sequencing is limited to approximately 100-200 bp. Although chemical crosslinking techniques can achieve single-base resolution, these methods are often complex, and the specificity of the antibodies used in these methods has raised concerns. In particular, the issue of off-target binding by the antibodies requires urgent attention. Enzyme-assisted sequencing has improved the accuracy of the localization analysis of RNA modifications and enables stoichiometric detection with single-base resolution. However, the enzymes used in this technique show poor reactivity, specificity, and sequence preference. Chemical labeling sequencing has become a widely used approach for profiling RNA modifications, particularly by altering reverse transcription (RT) signatures such as RT stops, misincorporations, and deletions. Chemical-assisted sequencing provides a sequence-independent RNA modification detection strategy that enables the localization of multiple RNA modifications. Additionally, when combined with the biotin-streptavidin affinity method, low-abundance RNA modifications can be enriched and detected. Nevertheless, the specificity of many chemical reactions remains problematic, and the development of specific reaction probes for particular modifications should continue in the future to achieve the precise localization of RNA modifications. As an indirect localization method, metabolic labeling sequencing specifically localizes the sites at which modifying enzymes act, which is of great significance in the study of RNA modification functions. However, this method is limited by the intracellular labeling of RNA and cannot be applied to biological samples such as clinical tissues and blood samples. Nanopore sequencing is a direct RNA-sequencing method that does not require RT or the polymerase chain reaction (PCR). However, challenges in analyzing the data obtained from nanopore sequencing, such as the high rate of false positives, must be resolved. Discussing sequencing analysis methods for various types of RNA modifications is instructive for the future development of novel RNA modification mapping technologies, and will aid studies on the functions of RNA modifications across the entire transcriptome.
Collapse
|
38
|
Rabelo-Fernández RJ, Yuen M, Batista PJ. The metabolic baton: conducting the dance of N6-methyladenosine writing and erasing. Curr Opin Genet Dev 2024; 86:102206. [PMID: 38788488 PMCID: PMC11212039 DOI: 10.1016/j.gde.2024.102206] [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: 02/13/2024] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024]
Abstract
The modification N6-methyladenosine (m6A) plays an important role in determining the functional output of gene expression programs. Throughout the transcriptome, the levels of m6A are tightly regulated by the opposing activities of methyltransferases and demethylases, as well as the interaction of modified transcripts with m6A-dependent RNA-binding proteins that modulate transcript stability, often referred to as writers, erasers, and readers. The enzymatic activities of both writers and erasers are tightly linked to the cellular metabolic environment, as these enzymatic reactions rely on metabolism intermediaries as cofactors. In this review, we highlight the examples of intersection between metabolism and m6A-dependent gene regulation and discuss the different contexts where this interaction plays important roles.
Collapse
Affiliation(s)
- Robert J Rabelo-Fernández
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Madeline Yuen
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pedro J Batista
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
39
|
Vignolini T, Couble JEC, Doré GRG, Baumgarten S. Transcript tinkering: RNA modifications in protozoan parasites. Curr Opin Microbiol 2024; 79:102477. [PMID: 38663181 DOI: 10.1016/j.mib.2024.102477] [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: 05/05/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 06/11/2024]
Abstract
Apicomplexan and trypanosomatid parasites have evolved a wide range of post-transcriptional processes that allow them to replicate, differentiate, and transmit within and among multiple different tissue, host, and vector environments. In this review, we highlight the recent advances that point toward the regulatory potential of RNA modifications in mediating these processes on the coding and noncoding transcriptome throughout the life cycle of protozoan parasites. We discuss the recent technical advancements enabling the study of the 'epitranscriptome' and how parasites evolved RNA modification-mediated mechanisms adapted to their unique lifestyles.
Collapse
Affiliation(s)
- Tiziano Vignolini
- Institut Pasteur, Université Paris Cité, G5 Parasite RNA Biology, Department of Parasites and Insect Vectors, F-75015 Paris, France
| | - Justine E C Couble
- Institut Pasteur, Université Paris Cité, G5 Parasite RNA Biology, Department of Parasites and Insect Vectors, F-75015 Paris, France
| | - Grégory R G Doré
- Institut Pasteur, Université Paris Cité, G5 Parasite RNA Biology, Department of Parasites and Insect Vectors, F-75015 Paris, France
| | - Sebastian Baumgarten
- Institut Pasteur, Université Paris Cité, G5 Parasite RNA Biology, Department of Parasites and Insect Vectors, F-75015 Paris, France.
| |
Collapse
|
40
|
Shin KWD, Hamanaka RB. Tamping Down the Fire: Taming Pyroptosis through RNA Methylation in Acute Lung Injury. Am J Respir Cell Mol Biol 2024; 70:331-333. [PMID: 38353587 PMCID: PMC11109585 DOI: 10.1165/rcmb.2024-0001ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Affiliation(s)
- Kun Woo D Shin
- Section of Pulmonary and Critical Care Medicine The University of Chicago Chicago, Illinois
| | - Robert B Hamanaka
- Section of Pulmonary and Critical Care Medicine The University of Chicago Chicago, Illinois
| |
Collapse
|
41
|
Tegowski M, Meyer KD. Studying m 6A in the brain: a perspective on current methods, challenges, and future directions. Front Mol Neurosci 2024; 17:1393973. [PMID: 38711483 PMCID: PMC11070500 DOI: 10.3389/fnmol.2024.1393973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/12/2024] [Indexed: 05/08/2024] Open
Abstract
A major mechanism of post-transcriptional RNA regulation in cells is the addition of chemical modifications to RNA nucleosides, which contributes to nearly every aspect of the RNA life cycle. N6-methyladenosine (m6A) is a highly prevalent modification in cellular mRNAs and non-coding RNAs, and it plays important roles in the control of gene expression and cellular function. Within the brain, proper regulation of m6A is critical for neurodevelopment, learning and memory, and the response to injury, and m6A dysregulation has been implicated in a variety of neurological disorders. Thus, understanding m6A and how it is regulated in the brain is important for uncovering its roles in brain function and potentially identifying novel therapeutic pathways for human disease. Much of our knowledge of m6A has been driven by technical advances in the ability to map and quantify m6A sites. Here, we review current technologies for characterizing m6A and highlight emerging methods. We discuss the advantages and limitations of current tools as well as major challenges going forward, and we provide our perspective on how continued developments in this area can propel our understanding of m6A in the brain and its role in brain disease.
Collapse
Affiliation(s)
- Matthew Tegowski
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, United States
| | - Kate D. Meyer
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, United States
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
42
|
Horner SM, Thompson MG. Challenges to mapping and defining m 6A function in viral RNA. RNA (NEW YORK, N.Y.) 2024; 30:482-490. [PMID: 38531643 PMCID: PMC11019751 DOI: 10.1261/rna.079959.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/09/2024] [Indexed: 03/28/2024]
Abstract
Viral RNA molecules contain multiple layers of regulatory information. This includes features beyond the primary sequence, such as RNA structures and RNA modifications, including N6-methyladenosine (m6A). Many recent studies have identified the presence and location of m6A in viral RNA and have found diverse regulatory roles for this modification during viral infection. However, to date, viral m6A mapping strategies have limitations that prevent a complete understanding of the function of m6A on individual viral RNA molecules. While m6A sites have been profiled on bulk RNA from many viruses, the resulting m6A maps of viral RNAs described to date present a composite picture of m6A across viral RNA molecules in the infected cell. Thus, for most viruses, it is unknown if unique viral m6A profiles exist throughout infection, nor if they regulate specific viral life cycle stages. Here, we describe several challenges to defining the function of m6A in viral RNA molecules and provide a framework for future studies to help in the understanding of how m6A regulates viral infection.
Collapse
Affiliation(s)
- Stacy M Horner
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Matthew G Thompson
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
43
|
Witzenberger M, Schwartz S. Directing RNA-modifying machineries towards endogenous RNAs: opportunities and challenges. Trends Genet 2024; 40:313-325. [PMID: 38350740 DOI: 10.1016/j.tig.2024.01.001] [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: 11/16/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 02/15/2024]
Abstract
Over 170 chemical modifications can be naturally installed on RNA, all of which are catalyzed by dedicated machineries. These modifications can alter RNA sequence structure, stability, and translation as well as serving as quality control marks that record aspects of RNA processing. The diverse roles played by RNAs within cells has motivated endeavors to exogenously introduce RNA modifications at target sites for diverse purposes ranging from recording RNA:protein interactions to therapeutic applications. Here, we discuss these applications and the approaches that have been employed to engineer RNA-modifying machineries, and highlight persisting challenges and perspectives.
Collapse
Affiliation(s)
- Monika Witzenberger
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7630031, Israel.
| | - Schraga Schwartz
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7630031, Israel.
| |
Collapse
|
44
|
Li Y, Wang Y, Vera-Rodriguez M, Lindeman LC, Skuggen LE, Rasmussen EMK, Jermstad I, Khan S, Fosslie M, Skuland T, Indahl M, Khodeer S, Klemsdal EK, Jin KX, Dalen KT, Fedorcsak P, Greggains GD, Lerdrup M, Klungland A, Au KF, Dahl JA. Single-cell m 6A mapping in vivo using picoMeRIP-seq. Nat Biotechnol 2024; 42:591-596. [PMID: 37349523 PMCID: PMC10739642 DOI: 10.1038/s41587-023-01831-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 05/17/2023] [Indexed: 06/24/2023]
Abstract
Current N6-methyladenosine (m6A) mapping methods need large amounts of RNA or are limited to cultured cells. Through optimized sample recovery and signal-to-noise ratio, we developed picogram-scale m6A RNA immunoprecipitation and sequencing (picoMeRIP-seq) for studying m6A in vivo in single cells and scarce cell types using standard laboratory equipment. We benchmark m6A mapping on titrations of poly(A) RNA and embryonic stem cells and in single zebrafish zygotes, mouse oocytes and embryos.
Collapse
Affiliation(s)
- Yanjiao Li
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Yunhao Wang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Maria Vera-Rodriguez
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Reproductive Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | | | | | - Erik M K Rasmussen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Aas, Norway
| | - Ingunn Jermstad
- Norwegian Transgenic Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Shaista Khan
- Norwegian Transgenic Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Madeleine Fosslie
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Trine Skuland
- Department of Reproductive Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Division of Gynaecology and Obstetrics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marie Indahl
- Department of Reproductive Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Division of Gynaecology and Obstetrics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sherif Khodeer
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- KU Leuven-University of Leuven, Department of Development and Regeneration, Leuven Institute for Single-Cell Omics (LISCO), Leuven Stem Cell Institute, Leuven, Belgium
| | | | - Kang-Xuan Jin
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Knut Tomas Dalen
- Norwegian Transgenic Centre, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Peter Fedorcsak
- Department of Reproductive Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Division of Gynaecology and Obstetrics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gareth D Greggains
- Department of Reproductive Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Mads Lerdrup
- Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Klungland
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
- Department of Microbiology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Kin Fai Au
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
- Biomedical Informatics Shared Resources, The Ohio State University, Columbus, OH, USA.
| | - John Arne Dahl
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| |
Collapse
|
45
|
Liu WW, Zheng SQ, Li T, Fei YF, Wang C, Zhang S, Wang F, Jiang GM, Wang H. RNA modifications in cellular metabolism: implications for metabolism-targeted therapy and immunotherapy. Signal Transduct Target Ther 2024; 9:70. [PMID: 38531882 DOI: 10.1038/s41392-024-01777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/28/2024] Open
Abstract
Cellular metabolism is an intricate network satisfying bioenergetic and biosynthesis requirements of cells. Relevant studies have been constantly making inroads in our understanding of pathophysiology, and inspiring development of therapeutics. As a crucial component of epigenetics at post-transcription level, RNA modification significantly determines RNA fates, further affecting various biological processes and cellular phenotypes. To be noted, immunometabolism defines the metabolic alterations occur on immune cells in different stages and immunological contexts. In this review, we characterize the distribution features, modifying mechanisms and biological functions of 8 RNA modifications, including N6-methyladenosine (m6A), N6,2'-O-dimethyladenosine (m6Am), N1-methyladenosine (m1A), 5-methylcytosine (m5C), N4-acetylcytosine (ac4C), N7-methylguanosine (m7G), Pseudouridine (Ψ), adenosine-to-inosine (A-to-I) editing, which are relatively the most studied types. Then regulatory roles of these RNA modification on metabolism in diverse health and disease contexts are comprehensively described, categorized as glucose, lipid, amino acid, and mitochondrial metabolism. And we highlight the regulation of RNA modifications on immunometabolism, further influencing immune responses. Above all, we provide a thorough discussion about clinical implications of RNA modification in metabolism-targeted therapy and immunotherapy, progression of RNA modification-targeted agents, and its potential in RNA-targeted therapeutics. Eventually, we give legitimate perspectives for future researches in this field from methodological requirements, mechanistic insights, to therapeutic applications.
Collapse
Affiliation(s)
- Wei-Wei Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Clinical Medicine, Shandong University, Jinan, China
| | - Si-Qing Zheng
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Tian Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Yun-Fei Fei
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Chen Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Shuang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Fei Wang
- Neurosurgical Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Guan-Min Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Hao Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China.
| |
Collapse
|
46
|
Ozato Y, Hara T, Meng S, Sato H, Tatekawa S, Uemura M, Yabumoto T, Uchida S, Ogawa K, Doki Y, Eguchi H, Ishii H. RNA methylation in inflammatory bowel disease. Cancer Sci 2024; 115:723-733. [PMID: 38263895 PMCID: PMC10920996 DOI: 10.1111/cas.16048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/29/2023] [Accepted: 12/06/2023] [Indexed: 01/25/2024] Open
Abstract
RNA modifications, including the renowned m6A, have recently garnered significant attention. This chemical alteration, present in mRNA, exerts a profound influence on protein expression levels by affecting splicing, nuclear export, stability, translation, and other critical processes. Although the role of RNA methylation in the pathogenesis and progression of IBD and colorectal cancer has been reported, many aspects remain unresolved. In this comprehensive review, we present recent studies on RNA methylation in IBD and colorectal cancer, with a particular focus on m6A and its regulators. We highlight the pivotal role of m6A in the pathogenesis of IBD and colorectal cancer and explore the potential applications of m6A modifications in the diagnosis and treatment of these diseases.
Collapse
Grants
- 18KK0251 19K22658 20H00541 21K19526 Ministry of Education, Culture, Sports, Science and Technology
- 22H03146 22K19559 23K19505 16H06279 (PAGS) Ministry of Education, Culture, Sports, Science and Technology
- grant nos. 17cm0106414h0002 JP21lm0203007 Ministry of Education, Culture, Sports, Science and Technology
- 2021-48 Mitsubishi Foundation
- Ministry of Education, Culture, Sports, Science and Technology
- Mitsubishi Foundation
Collapse
Affiliation(s)
- Yuki Ozato
- Department of Medical Data ScienceCenter of Medical Innovation and Translational Research, Osaka University Graduate School of MedicineSuitaJapan
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | - Tomoaki Hara
- Department of Medical Data ScienceCenter of Medical Innovation and Translational Research, Osaka University Graduate School of MedicineSuitaJapan
| | - Sikun Meng
- Department of Medical Data ScienceCenter of Medical Innovation and Translational Research, Osaka University Graduate School of MedicineSuitaJapan
| | - Hiromichi Sato
- Department of Medical Data ScienceCenter of Medical Innovation and Translational Research, Osaka University Graduate School of MedicineSuitaJapan
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | - Shotaro Tatekawa
- Department of Radiation OncologyOsaka University Graduate School of MedicineSuitaJapan
| | - Mamoru Uemura
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | | | - Shizuka Uchida
- Department of Clinical Medicine, Center for RNA MedicineAalborg UniversityCopenhagen SVDenmark
| | - Kazuhiko Ogawa
- Department of Radiation OncologyOsaka University Graduate School of MedicineSuitaJapan
| | - Yuichiro Doki
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | - Hidetoshi Eguchi
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineSuitaJapan
| | - Hideshi Ishii
- Department of Medical Data ScienceCenter of Medical Innovation and Translational Research, Osaka University Graduate School of MedicineSuitaJapan
| |
Collapse
|
47
|
Goh WSS, Kuang Y. Heterogeneity of chemical modifications on RNA. Biophys Rev 2024; 16:79-87. [PMID: 38495447 PMCID: PMC10937866 DOI: 10.1007/s12551-023-01128-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/27/2023] [Indexed: 03/19/2024] Open
Abstract
The chemical modifications of RNAs broadly impact almost all cellular events and influence various diseases. The rapid advance of sequencing and other technologies opened the door to global methods for profiling all RNA modifications, namely the "epitranscriptome." The mapping of epitranscriptomes in different cells and tissues unveiled that RNA modifications exhibit extensive heterogeneity, in type, amount, and in location. In this mini review, we first introduce the current understanding of modifications on major types of RNAs and the methods that enabled their discovery. We next discuss the tissue and cell heterogeneity of RNA modifications and briefly address the limitations of current technologies. With much still remaining unknown, the development of the epitranscriptomic field lies in the further developments of novel technologies.
Collapse
Affiliation(s)
- W. S. Sho Goh
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
48
|
Maestri S, Furlan M, Mulroney L, Coscujuela Tarrero L, Ugolini C, Dalla Pozza F, Leonardi T, Birney E, Nicassio F, Pelizzola M. Benchmarking of computational methods for m6A profiling with Nanopore direct RNA sequencing. Brief Bioinform 2024; 25:bbae001. [PMID: 38279646 PMCID: PMC10818168 DOI: 10.1093/bib/bbae001] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/27/2023] [Accepted: 12/28/2023] [Indexed: 01/28/2024] Open
Abstract
N6-methyladenosine (m6A) is the most abundant internal eukaryotic mRNA modification, and is involved in the regulation of various biological processes. Direct Nanopore sequencing of native RNA (dRNA-seq) emerged as a leading approach for its identification. Several software were published for m6A detection and there is a strong need for independent studies benchmarking their performance on data from different species, and against various reference datasets. Moreover, a computational workflow is needed to streamline the execution of tools whose installation and execution remains complicated. We developed NanOlympicsMod, a Nextflow pipeline exploiting containerized technology for comparing 14 tools for m6A detection on dRNA-seq data. NanOlympicsMod was tested on dRNA-seq data generated from in vitro (un)modified synthetic oligos. The m6A hits returned by each tool were compared to the m6A position known by design of the oligos. In addition, NanOlympicsMod was used on dRNA-seq datasets from wild-type and m6A-depleted yeast, mouse and human, and each tool's hits were compared to reference m6A sets generated by leading orthogonal methods. The performance of the tools markedly differed across datasets, and methods adopting different approaches showed different preferences in terms of precision and recall. Changing the stringency cut-offs allowed for tuning the precision-recall trade-off towards user preferences. Finally, we determined that precision and recall of tools are markedly influenced by sequencing depth, and that additional sequencing would likely reveal additional m6A sites. Thanks to the possibility of including novel tools, NanOlympicsMod will streamline the benchmarking of m6A detection tools on dRNA-seq data, improving future RNA modification characterization.
Collapse
Affiliation(s)
- Simone Maestri
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Mattia Furlan
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Logan Mulroney
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridgeshire, U.K
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory (EMBL), Rome, Italy
| | - Lucia Coscujuela Tarrero
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Camilla Ugolini
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Fabio Dalla Pozza
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Tommaso Leonardi
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Ewan Birney
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridgeshire, U.K
| | - Francesco Nicassio
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Mattia Pelizzola
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
49
|
Liang Z, Ye H, Ma J, Wei Z, Wang Y, Zhang Y, Huang D, Song B, Meng J, Rigden DJ, Chen K. m6A-Atlas v2.0: updated resources for unraveling the N6-methyladenosine (m6A) epitranscriptome among multiple species. Nucleic Acids Res 2024; 52:D194-D202. [PMID: 37587690 PMCID: PMC10768109 DOI: 10.1093/nar/gkad691] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023] Open
Abstract
N 6-Methyladenosine (m6A) is one of the most abundant internal chemical modifications on eukaryote mRNA and is involved in numerous essential molecular functions and biological processes. To facilitate the study of this important post-transcriptional modification, we present here m6A-Atlas v2.0, an updated version of m6A-Atlas. It was expanded to include a total of 797 091 reliable m6A sites from 13 high-resolution technologies and two single-cell m6A profiles. Additionally, three methods (exomePeaks2, MACS2 and TRESS) were used to identify >16 million m6A enrichment peaks from 2712 MeRIP-seq experiments covering 651 conditions in 42 species. Quality control results of MeRIP-seq samples were also provided to help users to select reliable peaks. We also estimated the condition-specific quantitative m6A profiles (i.e. differential methylation) under 172 experimental conditions for 19 species. Further, to provide insights into potential functional circuitry, the m6A epitranscriptomics were annotated with various genomic features, interactions with RNA-binding proteins and microRNA, potentially linked splicing events and single nucleotide polymorphisms. The collected m6A sites and their functional annotations can be freely queried and downloaded via a user-friendly graphical interface at: http://rnamd.org/m6a.
Collapse
Affiliation(s)
- Zhanmin Liang
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350004, China
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
| | - Haokai Ye
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, Liverpool, UK
| | - Jiongming Ma
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350004, China
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, Liverpool, UK
| | - Zhen Wei
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L69 7ZB, UK
| | - Yue Wang
- Department of Mathematical Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
- Department of Computer Science, University of Liverpool, Liverpool L69 7ZB, UK
| | - Yuxin Zhang
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350004, China
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, Liverpool, UK
| | - Daiyun Huang
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
- Department of Computer Science, University of Liverpool, Liverpool L69 7ZB, UK
| | - Bowen Song
- Department of Public Health, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jia Meng
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
- AI University Research Centre, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, Liverpool, UK
| | - Daniel J Rigden
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, Liverpool, UK
| | - Kunqi Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350004, China
| |
Collapse
|
50
|
Cerneckis J, Ming GL, Song H, He C, Shi Y. The rise of epitranscriptomics: recent developments and future directions. Trends Pharmacol Sci 2024; 45:24-38. [PMID: 38103979 PMCID: PMC10843569 DOI: 10.1016/j.tips.2023.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 12/19/2023]
Abstract
The epitranscriptomics field has undergone tremendous growth since the discovery that the RNA N6-methyladenosine (m6A) modification is reversible and is distributed throughout the transcriptome. Efforts to map RNA modifications transcriptome-wide and reshape the epitranscriptome in disease settings have facilitated mechanistic understanding and drug discovery in the field. In this review we discuss recent advancements in RNA modification detection methods and consider how these developments can be applied to gain novel insights into the epitranscriptome. We also highlight drug discovery efforts aimed at developing epitranscriptomic therapeutics for cancer and other diseases. Finally, we consider engineering of the epitranscriptome as an emerging direction to investigate RNA modifications and their causal effects on RNA processing at high specificity.
Collapse
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Department of Cell and Developmental Biology, Department of Psychiatry, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Department of Cell and Developmental Biology, the Epigenetics Institute, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, Howard Hughes Medical Institute, the University of Chicago, Chicago, IL 60637, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|