1
|
Alkhammash A. Pharmacology of epitranscriptomic modifications: Decoding the therapeutic potential of RNA modifications in drug resistance. Eur J Pharmacol 2025; 994:177397. [PMID: 39978710 DOI: 10.1016/j.ejphar.2025.177397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
RNA modifications, collectively known as epitranscriptomic modifications, have emerged as critical regulators of gene expression, cellular adaptation, and therapeutic resistance. This review explores the pharmacological potential of targeting RNA modifications, including N6-methyladenosine (m6A) and 5-methylcytosine (m5C), as strategies to overcome drug resistance in cancer. We examine key regulatory enzymes, writers, erasers, and readers-and their roles in modulating RNA stability, translation, and splicing. Advances in combination therapies, integrating RNA modification modulators with conventional chemotherapies and immune checkpoint inhibitors, have shown promising outcomes in reversing multidrug resistance (MDR). Emerging RNA-targeting technologies, such as CRISPR/Cas13 systems and advanced RNA sequencing platforms, further enable precision manipulation of RNA molecules, opening new therapeutic frontiers. However, several challenges persist, including issues related to pharmacokinetics, acquired resistance, and the complexity of epitranscriptomic networks. This review underscores the need for innovative delivery systems, such as lipid nanoparticles and tissue-specific targeting strategies, and highlights the dynamic nature of RNA modifications in response to environmental and therapeutic stress. Ongoing research into non-coding RNA modifications and the interplay between epitranscriptomics and epigenetics offers exciting possibilities for developing novel RNA-targeting therapies. The continued evolution of RNA-based technologies will be crucial in advancing precision medicine, addressing drug resistance, and improving clinical outcomes across multiple diseases.
Collapse
Affiliation(s)
- Abdullah Alkhammash
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia.
| |
Collapse
|
2
|
Wilcox XE, Zhang H, Mah JL, Cazet JF, Mozumder S, Venkatesh S, Juliano CE, Beal PA, Fisher AJ. Phylogenetic and structural analysis of Hydra ADAR. Arch Biochem Biophys 2025; 767:110353. [PMID: 39986343 DOI: 10.1016/j.abb.2025.110353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Adenosine deaminases acting on RNAs (ADARs) perform adenosine-to-inosine (A-to-I) RNA editing for essential biological functions. While studies of editing sites in diverse animals have revealed unique biological roles of ADAR editing including temperature adaptation and reproductive maturation, rigorous biochemical and structural studies of these ADARs are lacking. Here, we present a phylogenetic sequence analysis and AlphaFold computational structure prediction to reveal that medusozoan ADAR2s contain five dsRNA binding domains (dsRBDs) with several RNA binding residues in the dsRBDs and deaminase domain conserved. Additionally, we identified evolutionary divergence between the medusozoan (e.g. Hydra) and anthozoan cnidarian subphyla. The anthozoan ADAR deaminase domains more closely resemble human ADARs with longer 5' RNA binding loops, glutamate base-flipping residues, and a conserved TWDG dimerization motif. Conversely, medusozoan ADAR deaminase domains have short 5' binding loops, glutamine flipping residues, and non-conserved helix dimerization motif. We also report the direct detection of A-to-I RNA editing by an ADAR ortholog from the freshwater cnidarian Hydra vulgaris (hyADAR). We solved the crystal structure of the monomeric deaminase domain of hyADAR (hyADARd) to 2.0 Å resolution, showing conserved active site architecture and the presence of a buried inositol hexakisphosphate known to be required for ADAR activity. In addition, these data demonstrate that medusozoans have evolved novel ADAR structural features, however the physiological consequence of this remains unknown. In addition, these results provide a framework for biochemically and structurally characterizing ADARs from evolutionarily distant organisms to understand the diverse roles of ADAR editing amongst metazoans.
Collapse
Affiliation(s)
- Xander E Wilcox
- Department of Chemistry, University of California, Davis, CA, USA
| | - Howard Zhang
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Jasmine L Mah
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Jack F Cazet
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Sukanya Mozumder
- Department of Chemistry, University of California, Davis, CA, USA
| | - Srinidhi Venkatesh
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Celina E Juliano
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Peter A Beal
- Department of Chemistry, University of California, Davis, CA, USA
| | - Andrew J Fisher
- Department of Chemistry, University of California, Davis, CA, USA; Department of Molecular and Cellular Biology, University of California, Davis, CA, USA.
| |
Collapse
|
3
|
Ashley CN, Broni E, Pena-Martinez M, Wood CM, Kwofie SK, Miller WA. Computer-Aided Discovery of Natural Compounds Targeting the ADAR2 dsRBD2-RNA Interface and Computational Modeling of Full-Length ADAR2 Protein Structure. Int J Mol Sci 2025; 26:4075. [PMID: 40362314 PMCID: PMC12072074 DOI: 10.3390/ijms26094075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Mesothelioma is a rare and aggressive cancer linked to asbestos exposure and characterized by rapid metastasis and poor prognosis. Inhibition of adenosine deaminase acting on dsRNA 2 (ADAR2) RNA binding but not ADAR2 editing has shown antitumor effects in mesothelioma. Natural compounds from the Traditional Chinese Medicine (TCM) database were docked to the RNA-binding interface of ADAR2's second dsRNA binding domain (dsRBD2), and their drug-likeness and predicted safety were assessed. Eight ligands (ZINC000085597263, ZINC000085633079, ZINC000014649947, ZINC000034512861, ZINC000070454124, ZINC000085594944, ZINC000085633008, and ZINC000095909822) showed high binding affinity to dsRBD2 from molecular mechanics Poisson-Boltzmann surface area (MM/PBSA) calculations. Protein-ligand interactions were analyzed to identify key residues contributing to these binding affinities. Molecular dynamics (MD) simulations of dsRBD-ligand-RNA complexes revealed that four compounds (ZINC000085597263, ZINC000085633079, ZINC000014649947, and ZINC000034512861) had negative binding affinities to dsRBD2 in the presence of the RNA substrate GluR-2. Key residues, including Val164, Met165, Lys209, and Lys212, were crucial for ligand binding, even with RNA present, suggesting these compounds could inhibit dsRBD2's RNA-binding function. The predicted biological activities of these compounds indicate potential anticancer properties, particularly for the treatment of mesothelioma. These compounds are structurally similar to known anti-mesothelioma agents or anticancer drugs, highlighting their therapeutic potential. Current mesothelioma treatments are limited. Optimization of these compounds, alone or in combination with current therapeutics, has potential for mesothelioma treatment. Additionally, five high-quality full-length ADAR2 models were developed. These models provide insights into ADAR2 function, mutation impacts, and potential areas for protein engineering to enhance stability, RNA-binding specificity, or protein interactions, particularly concerning dimerization or complex formation with other proteins and RNAs.
Collapse
Affiliation(s)
- Carolyn N. Ashley
- Department of Medicine, Loyola University Medical Center, Loyola University Chicago, Maywood, IL 60153, USA; (C.N.A.); (E.B.)
| | - Emmanuel Broni
- Department of Medicine, Loyola University Medical Center, Loyola University Chicago, Maywood, IL 60153, USA; (C.N.A.); (E.B.)
| | - Michelle Pena-Martinez
- Department of Medicine, Loyola University Medical Center, Loyola University Chicago, Maywood, IL 60153, USA; (C.N.A.); (E.B.)
| | - Chanyah M. Wood
- Department of Molecular Pharmacology & Neuroscience, Loyola University Medical Center, Loyola University Chicago, Maywood, IL 60153, USA
| | - Samuel K. Kwofie
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic & Applied Sciences, University of Ghana, Legon, Accra LG 77, Ghana;
| | - Whelton A. Miller
- Department of Medicine, Loyola University Medical Center, Loyola University Chicago, Maywood, IL 60153, USA; (C.N.A.); (E.B.)
- Department of Molecular Pharmacology & Neuroscience, Loyola University Medical Center, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
4
|
Liang H, Qi H, Wang C, Wang Y, Liu M, Chen J, Sun X, Xia T, Feng S, Chen C, Zheng D. Analysis of the complete mitogenomes of three high economic value tea plants (Tea-oil Camellia) provide insights into evolution and phylogeny relationship. FRONTIERS IN PLANT SCIENCE 2025; 16:1549185. [PMID: 40343121 PMCID: PMC12058841 DOI: 10.3389/fpls.2025.1549185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/18/2025] [Indexed: 05/11/2025]
Abstract
Introduction Tea-oil Camellia species play a crucial economic and ecological role worldwide, yet their mitochondrial genomes remain largely unexplored. Methods In this study, we assembled and analyzed the complete mitochondrial genomes of Camellia oleifera and C. meiocarpa, revealing multi-branch structures that deviate from the typical circular mitochondrial genome observed in most plants. The assembled mitogenomes span 953,690 bp (C. oleifera) and 923,117 bp (C. meiocarpa), containing 74 and 76 annotated mitochondrial genes, respectively. Results Comparative genomic analyses indicated that C. oleifera and C. meiocarpa share a closer genetic relationship, whereas C. drupifera is more distantly related. Codon usage analysis revealed that natural selection plays a dominant role in shaping codon bias in these mitochondrial genomes. Additionally, extensive gene transfer events were detected among the three species, highlighting the dynamic nature of mitochondrial genome evolution in Tea-oil Camellia. Phylogenetic reconstruction based on mitochondrial genes exhibited incongruence with chloroplast phylogenies, suggesting potential discordance due to hybridization events, incomplete lineage sorting (ILS), or horizontal gene transfer (HGT). Furthermore, we identified species-specific mitochondrial markers, which provide valuable molecular tools for distinguishing Tea-oil Camellia species. Discussion Our findings enhance the understanding of mitochondrial genome evolution and genetic diversity in Tea-oil Camellia, offering essential genomic resources for phylogenetics, species identification, and evolutionary research in woody plants.
Collapse
Affiliation(s)
- Heng Liang
- Institute of Tropical Horticulture Research, Hainan Academy of Agricultural Sciences, Haikou, China
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Haikou, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Huasha Qi
- Institute of Tropical Horticulture Research, Hainan Academy of Agricultural Sciences, Haikou, China
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Haikou, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Chunmei Wang
- Institute of Tropical Horticulture Research, Hainan Academy of Agricultural Sciences, Haikou, China
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Haikou, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Yidan Wang
- School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Moyang Liu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Jiali Chen
- Institute of Tropical Horticulture Research, Hainan Academy of Agricultural Sciences, Haikou, China
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Haikou, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Xiuxiu Sun
- Institute of Tropical Horticulture Research, Hainan Academy of Agricultural Sciences, Haikou, China
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Haikou, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Tengfei Xia
- Institute of Tropical Horticulture Research, Hainan Academy of Agricultural Sciences, Haikou, China
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Haikou, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Shiling Feng
- College of Life Science, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Cheng Chen
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Daojun Zheng
- Institute of Tropical Horticulture Research, Hainan Academy of Agricultural Sciences, Haikou, China
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Haikou, China
- Key Laboratory of Tropic Special Economic Plant Innovation and Utilization, Hainan Academy of Agricultural Sciences, Haikou, China
| |
Collapse
|
5
|
Li JB, Walkley CR. Leveraging genetics to understand ADAR1-mediated RNA editing in health and disease. Nat Rev Genet 2025:10.1038/s41576-025-00830-5. [PMID: 40229561 DOI: 10.1038/s41576-025-00830-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2025] [Indexed: 04/16/2025]
Abstract
Endogenous, long double-stranded RNA (dsRNA) can resemble viral dsRNA and be recognized by cytosolic dsRNA sensors, triggering autoimmunity. Genetic studies of rare, inherited human diseases and experiments using mouse models have established the importance of adenosine-to-inosine RNA editing by the enzyme adenosine deaminase acting on RNA 1 (ADAR1) as a critical safeguard against autoinflammatory responses to cellular dsRNA. More recently, human genetic studies have revealed that dsRNA editing and sensing mechanisms are involved in common inflammatory diseases, emphasizing the broader role of dsRNA in modulating immune responses and disease pathogenesis. These findings have highlighted the therapeutic potential of targeting dsRNA editing and sensing, as exemplified by the emergence of ADAR1 inhibition in cancer therapy.
Collapse
Affiliation(s)
- Jin Billy Li
- Department of Genetics, Stanford University, Stanford, CA, USA.
| | - Carl R Walkley
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
- Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia.
| |
Collapse
|
6
|
Duan Z, Liu X. Emerging antiviral defense systems in fungi. Cell Host Microbe 2025; 33:461-463. [PMID: 40209673 DOI: 10.1016/j.chom.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 04/12/2025]
Abstract
In this issue of Cell Host & Microbe, Honda et al. identify an antiviral defense system in fungi. Mycoviruses trigger the expression of RNA-editing enzymes, which catalyze the editing of mRNAs encoding zinc finger transcription factors. This system differs from the RNA interference and is conserved in filamentous fungi.
Collapse
Affiliation(s)
- Zeyu Duan
- State Key Laboratory of Microbial Diversity and Innovative Utilization, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiao Liu
- State Key Laboratory of Microbial Diversity and Innovative Utilization, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
7
|
Luo H, Yao J, Zhang R. Harnessing RNA base editing for diverse applications in RNA biology and RNA therapeutics. ADVANCED BIOTECHNOLOGY 2025; 3:11. [PMID: 40198443 PMCID: PMC11979053 DOI: 10.1007/s44307-025-00063-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Recent advancements in molecular engineering have established RNA-based technologies as powerful tools for both fundamental research and translational applications. Among the various RNA-based technologies developed, RNA base editing has recently emerged as a groundbreaking advancement. It primarily involves the conversion of adenosine (A) to inosine (I) and cytidine (C) to uridine (U), which are mediated by ADAR and APOBEC enzymes, respectively. RNA base editing has been applied in both biological research and therapeutic contexts. It enables site-directed editing within target transcripts, offering reversible, dose-dependent effects, in contrast to the permanent or heritable changes associated with DNA base editing. Additionally, RNA editing-based profiling of RNA-binding protein (RBP) binding sites facilitates transcriptome-wide mapping of RBP-RNA interactions in specific tissues and at the single-cell level. Furthermore, RNA editing-based sensors have been utilized to express effector proteins in response to specific RNA species. As RNA base editing technologies continue to evolve, we anticipate that they will significantly drive advancements in RNA therapeutics, synthetic biology, and biological research.
Collapse
Affiliation(s)
- Hui Luo
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
- Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Jing Yao
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
- Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Rui Zhang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
- Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China.
| |
Collapse
|
8
|
Xie Q, Duan Y. An Ultimate Question for Functional A-to-I mRNA Editing: Why Not a Genomic G? J Mol Evol 2025; 93:185-192. [PMID: 39964487 DOI: 10.1007/s00239-025-10238-8] [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: 12/02/2024] [Accepted: 02/07/2025] [Indexed: 04/18/2025]
Abstract
A-to-I mRNA editing resembles A-to-G mutations. Functional mRNA editing, representing only a corner of total editing events, can be inferred from the experimental removal of editing. However, it is intuitive to ask why evolution chose RNA editing rather than directly (and simply) changing the genomic sequence to G? If G is better than A, then drift or constructive neutral evolution (CNE) theory can explain the emergence of such editing, but it is still unclear why the exemplified conserved editing is perfectly maintained without observing any subsequent A-to-G DNA mutations? Virtually every functional and conserved mRNA editing site faces this ultimate question until one justifies that being editable is better than a hardwired genomic allele. While the advantage of editability has been validated in fungi, this ultimate question has not been answered for any functional editing sites in animals. By providing several conceptual arguments and specific examples, we propose that proving the evolutionary adaptiveness of an editing site is far more difficult than revealing its function.
Collapse
Affiliation(s)
- Qiuhua Xie
- Department of Entomology and State Key Laboratory of Agricultural and Forestry Biosecurity, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Yuange Duan
- Department of Entomology and State Key Laboratory of Agricultural and Forestry Biosecurity, MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
9
|
Xiao L, Qin B, Zhang X, Yao Y, Dou X, Liu Y, Duan X. Precision Control of Cell Type-Specific Behavior via RNA Sensing and Editing. SMALL METHODS 2025; 9:e2400952. [PMID: 39632651 DOI: 10.1002/smtd.202400952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/30/2024] [Indexed: 12/07/2024]
Abstract
In the realms of bioengineering and biopharmaceuticals, there exists a critical demand for advanced genetic tools that can interact with specific cell signaling pathways to accurately identify and target various cell types. This research introduces the innovative CRISPR-ADAReader system, which enables precise manipulation of cell activity through sensing target RNA. Featuring both positive and negative feedback loops, the system allows for tailored regulation across different cell types in response to various internal signals, showcasing exceptional programmability, specificity, and sensitivity. By choosing distinct RNAs as activation signals, the CRISPR-ADAReader efficiently monitors and alters targeted cell behaviors. In a case study focusing on retinoblastoma treatment, the system distinctively initiates positive feedback and self-silencing actions by detecting MCYN and Rb transcripts, thus safeguarding normal retinal pigment epithelial cells while promoting apoptosis in cancer cells. Moreover, the CRISPR-ADAReader demonstrates significant anti-tumor effectiveness in live models, markedly reducing retinoblastoma cell proliferation through the activation of several cancer-suppression pathways, outperforming the capabilities of the ADAR-sensor. Notably, the system also shows an excellent in vivo safety profile. In conclusion, the CRISPR-ADAReader system represents a groundbreaking method for the detection and editing of RNA, offering a potent instrument for the customized and precise governance of cell behavior.
Collapse
Affiliation(s)
- Lulu Xiao
- Aier Eye Hospital, Jinan University, Guangzhou, 510071, China
- Aier Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, 410023, China
| | - Bo Qin
- Aier Eye Hospital, Jinan University, Guangzhou, 510071, China
- Shenzhen Aier Eye Hospital, Aier Eye Hospital, Jinan University, Shenzhen, China Shenzhen Aier Ophthalmic Technology Institute, Shenzhen, 518032, China
| | - Xinyue Zhang
- Aier Eye Hospital, Jinan University, Guangzhou, 510071, China
- Aier Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, 410023, China
| | - Yufeng Yao
- Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, Guangdong, 518031, China
- Department of Ophthalmology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, 518031, China
| | - Xiaoyan Dou
- Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, Guangdong, 518031, China
- Department of Ophthalmology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, 518031, China
| | - Yuchen Liu
- Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, Guangdong, 518031, China
| | - Xuanchu Duan
- Aier Eye Hospital, Jinan University, Guangzhou, 510071, China
- Aier Glaucoma Institute, Changsha Aier Eye Hospital, Changsha, Hunan, 410023, China
| |
Collapse
|
10
|
Luo S, Zhou X. Post-transcriptional regulation of behavior plasticity in social insects. CURRENT OPINION IN INSECT SCIENCE 2025; 68:101329. [PMID: 39708917 DOI: 10.1016/j.cois.2024.101329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
Social insects often show remarkable behavioral plasticity, which is closely associated with their respective castes. The underpinnings of this plasticity are complex, involving genetic differences among individuals within a colony and regulation of gene expression at multiple levels. Post-transcriptional regulation, which increases the complexity of the transcriptome, plays a crucial role in the multilayer regulatory network that influences social insect behavior. We provide an overview of the impact of three post-transcriptional regulatory processes on the reproductive division of labor and worker division of labor in social insects: alternative splicing, RNA modifications, and noncoding RNAs. We also discuss the relationship between post-transcriptional regulation and chromatin modification.
Collapse
Affiliation(s)
- Shiqi Luo
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, China.
| | - Xin Zhou
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, China.
| |
Collapse
|
11
|
Duan Y, Cao Q. Systematic revelation and meditation on the significance of long exons using representative eukaryotic genomes. BMC Genomics 2025; 26:290. [PMID: 40128699 PMCID: PMC11931755 DOI: 10.1186/s12864-025-11504-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Long exons/introns are not evenly distributed in the genome, but the biological significance of this phenomenon remains elusive. MATERIALS AND METHODS Exon properties were analyzed in seven well-annotated reference genomes, including human and other representative model organisms: mouse, fruitfly, worm, mouse-ear cress, corn, and rice. RESULTS In all species, last exons in genes tend to be the longest. Additionally, we found that (1) canonical splicing motifs are strongly underrepresented in 3'UTR; (2) Last exons tend to have low GC content; (3) Comparing with other species, first exons in D. melanogaster genes demonstrate lower GC content than internal exons. CONCLUSIONS It cannot be excluded that last exons of genes exert essential regulatory roles and is subjected to natural selection, exhibiting differential splicing tendency, and GC content compared to other parts of the gene body.
Collapse
Affiliation(s)
- Yuange Duan
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Qi Cao
- Health Science Center, International Cancer Institute, Peking University, Beijing, 100191, China.
| |
Collapse
|
12
|
Wu J, Wang Y, Chen H, Xu T, Yang W, Fang X. Solid-like condensation of MORF8 inhibits RNA editing under heat stress in Arabidopsis. Nat Commun 2025; 16:2789. [PMID: 40118828 PMCID: PMC11928522 DOI: 10.1038/s41467-025-58146-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 03/10/2025] [Indexed: 03/24/2025] Open
Abstract
Heat stress inhibits photosynthesis efficiency, thereby suppressing plant growth and crop yield. However, the mechanism underlying this inhibition is not fully understood. Here, we report that the multiple organellar RNA-editing factor 8 (MORF8) forms condensates with solid-like properties in chloroplasts upon heat stress. In vitro data show that the MORF8 condensation is intrinsically heat-dependent and primarily determined by its IDR (intrinsically disordered region). Purification and characterization of MORF8 condensates show that numerous editing factors including PPR proteins and MORFs are partitioned. We provide both genetic and biochemical evidence that MORF8 condensation inhibits chloroplast RNA editing. In agreement, we find that both heat stress and MORF8 condensation lead to reduced editing of RNAs encoding NADH dehydrogenase-like (NDH) complex and impaired NDH activity and photosynthesis efficiency. These findings uncover MORF8 as a putative chloroplastic thermosensor that mediates photosynthesis inhibition by heat and highlight the functional significance of solid material properties of biomolecular condensates.
Collapse
Affiliation(s)
- Jie Wu
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yue Wang
- Key Laboratory of Photobiology, Photosynthesis Research Center, Institute of Botany, Chinese Academy of Sciences, Beijing, China
| | - Haodong Chen
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Tongda Xu
- Haixia Institute of Science and Technology, and College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Wenqiang Yang
- Key Laboratory of Photobiology, Photosynthesis Research Center, Institute of Botany, Chinese Academy of Sciences, Beijing, China
| | - Xiaofeng Fang
- Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
13
|
Stejskal S, Rájecká V, Covelo-Molares H, Sinigaglia K, Brožinová K, Kašiarová L, Dohnálková M, Reyes-Gutierrez PE, Cahová H, Keegan LP, O'Connell MA, Vaňáčová Š. Global analysis by LC-MS/MS of N6-methyladenosine and inosine in mRNA reveal complex incidence. RNA (NEW YORK, N.Y.) 2025; 31:514-528. [PMID: 39746750 PMCID: PMC11912911 DOI: 10.1261/rna.080324.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025]
Abstract
The precise and unambiguous detection and quantification of internal RNA modifications represents a critical step for understanding their physiological functions. The methods of direct RNA sequencing are quickly developing allowing for the precise location of internal RNA marks. This detection is, however, not quantitative and still presents detection limits. One of the biggest remaining challenges in the field is still the detection and quantification of m6A, m6Am, inosine, and m1A modifications of adenosine. The second intriguing and timely question remaining to be addressed is the extent to which individual marks are coregulated or potentially can affect each other. Here, we present a methodological approach to detect and quantify several key mRNA modifications in human total RNA and in mRNA, which is difficult to purify away from contaminating tRNA. We show that the adenosine demethylase FTO primarily targets m6Am marks in noncoding RNAs in HEK293T cells. Surprisingly, we observe little effect of FTO or ALKBH5 depletion on the m6A mRNA levels. Interestingly, the upregulation of ALKBH5 is accompanied by an increase in inosine level in overall mRNA.
Collapse
Affiliation(s)
- Stanislav Stejskal
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Veronika Rájecká
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Helena Covelo-Molares
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Ketty Sinigaglia
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Květoslava Brožinová
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Linda Kašiarová
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Michaela Dohnálková
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | | | - Hana Cahová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Liam P Keegan
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Mary A O'Connell
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| | - Štěpánka Vaňáčová
- Central European Institute of Technology (CEITEC), Masaryk University, Brno 62500, Czech Republic
| |
Collapse
|
14
|
Fonzino A, Mazzacuva PL, Handen A, Silvestris DA, Arnold A, Pecori R, Pesole G, Picardi E. REDInet: a temporal convolutional network-based classifier for A-to-I RNA editing detection harnessing million known events. Brief Bioinform 2025; 26:bbaf107. [PMID: 40112338 PMCID: PMC11924403 DOI: 10.1093/bib/bbaf107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
A-to-I ribonucleic acid (RNA) editing detection is still a challenging task. Current bioinformatics tools rely on empirical filters and whole genome sequencing or whole exome sequencing data to remove background noise, sequencing errors, and artifacts. Sometimes they make use of cumbersome and time-consuming computational procedures. Here, we present REDInet, a temporal convolutional network-based deep learning algorithm, to profile RNA editing in human RNA sequencing (RNAseq) data. It has been trained on REDIportal RNA editing sites, the largest collection of human A-to-I changes from >8000 RNAseq data of the genotype-tissue expression project. REDInet can classify editing events with high accuracy harnessing RNAseq nucleotide frequencies of 101-base windows without the need for coupled genomic data.
Collapse
Affiliation(s)
- Adriano Fonzino
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
| | - Pietro Luca Mazzacuva
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, National Research Council, Via Amendola 122/O, 70126, Bari, Italy
- Department of Engineering, University Campus Bio-Medico of Rome, Via Álvaro del Portillo 21, 00128, Rome, Italy
| | - Adam Handen
- Biological Sciences Division, University of Chicago, 5841 S Maryland Avenue, 60637, Chicago, USA
| | - Domenico Alessandro Silvestris
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
| | - Annette Arnold
- Division of Immune Diversity, German Cancer Research Center, Im Neuenheimer Feld 28069120, Heidelberg, Germany
| | - Riccardo Pecori
- Division of Immune Diversity, German Cancer Research Center, Im Neuenheimer Feld 28069120, Heidelberg, Germany
- Helmholtz Institute for Translational Oncology (HI-TRON), Obere Zahlbacherstr., 55131, Mainz, Germany
| | - Graziano Pesole
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, National Research Council, Via Amendola 122/O, 70126, Bari, Italy
| | - Ernesto Picardi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125, Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, National Research Council, Via Amendola 122/O, 70126, Bari, Italy
| |
Collapse
|
15
|
Mu J, Wu C, Xu K, Liu X, Fu Y, Zhang Z, Yu J, Xue C, Wang Z, Chen X, Chen Y, Ou G, Liu Z. Conformational reorganization and phase separation drive hyper-editing of ADR-2-ADBP-1 complex. Nucleic Acids Res 2025; 53:gkaf148. [PMID: 40037706 PMCID: PMC11879458 DOI: 10.1093/nar/gkaf148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/06/2025] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
Adenosine deaminase acting on RNA (ADAR) proteins, which mediate adenosine-to-inosine editing of double-stranded ribonucleic acid (dsRNA) substrates, play essential roles in balancing innate immunity. Using cryogenic electron microscopy, we solved the structure of the Caenorhabditis elegans ADR-2-ADBP-1 complex (stoichiometric ratio, 2:2), which is an asymmetric ADR-2 dimer with one editing site blocked by the other ADR-2. Unexpectedly, dsRNA recruitment triggered dissociation of the ADR-2 dimer, exposing more competent dsRNA editing sites. Furthermore, high dsRNA and protein concentrations caused the formation of liquid-liquid phase-separated puncta, in which significantly greater editing activity was observed, indicating that organizational transitions enable the ADR-2-ADBP-1 complex to perform dsRNA hyper-editing. Our findings suggest that the ADAR editing mechanism adapts to different conditions via conformational reorganization.
Collapse
Affiliation(s)
- Jianqiang Mu
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology Shenzhen, 518055 Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Cang Wu
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology Shenzhen, 518055 Guangdong, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Kaiming Xu
- Tsinghua‐Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing 100084, China
| | - Xingang Liu
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology Shenzhen, 518055 Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Yajuan Fu
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology Shenzhen, 518055 Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Zhen Zhang
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology Shenzhen, 518055 Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Jingwei Yu
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Chenyang Xue
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology Shenzhen, 518055 Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Zi Wang
- Tsinghua‐Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing 100084, China
| | - Xinmeng Chen
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology Shenzhen, 518055 Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Yanhong Chen
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Guangshuo Ou
- Tsinghua‐Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing 100084, China
| | - Zhongmin Liu
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology Shenzhen, 518055 Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| |
Collapse
|
16
|
Santamarina-Ojeda P, Fernández AF, Fraga MF. Epitranscriptomics in the Glioma Context: A Brief Overview. Cancers (Basel) 2025; 17:578. [PMID: 40002173 PMCID: PMC11853273 DOI: 10.3390/cancers17040578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Epitranscriptomics, the study of chemical modifications in RNA, has emerged as a crucial field in cellular regulation, adding another layer to the established landscape of DNA- and histone-based epigenetics. A wide range of RNA modifications, including N6-methyladenosine, pseudouridine, and inosine, have been identified across nearly all RNA species, influencing essential processes such as transcription, splicing, RNA stability, and translation. In the context of brain tumors, particularly gliomas, specific epitranscriptomic signatures have been reported to play a role in tumorigenesis. Despite growing evidence, the biological implications of various RNA modifications remain poorly understood. This review offers an examination of the main RNA modifications, the interplay between modified and unmodified molecules, how they could contribute to glioma-like phenotypes, and the therapeutic impact of targeting these mechanisms.
Collapse
Affiliation(s)
- Pablo Santamarina-Ojeda
- Foundation for Biomedical Research and Innovation in Asturias (FINBA), 33011 Oviedo, Spain; (P.S.-O.); (A.F.F.)
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
- Nanomaterials and Nanotechnology Research Centre (CINN-CSIC), 33940 El Entrego, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 28029 Madrid, Spain
| | - Agustín F. Fernández
- Foundation for Biomedical Research and Innovation in Asturias (FINBA), 33011 Oviedo, Spain; (P.S.-O.); (A.F.F.)
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
- Nanomaterials and Nanotechnology Research Centre (CINN-CSIC), 33940 El Entrego, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 28029 Madrid, Spain
| | - Mario F. Fraga
- Foundation for Biomedical Research and Innovation in Asturias (FINBA), 33011 Oviedo, Spain; (P.S.-O.); (A.F.F.)
- Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
- Nanomaterials and Nanotechnology Research Centre (CINN-CSIC), 33940 El Entrego, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 28029 Madrid, Spain
| |
Collapse
|
17
|
Crooke PS, Tossberg JT, Aune TM. Increased unedited Alu RNA patterns found in cortex extracellular vesicles in Alzheimer's disease resemble hippocampus vasculature Alu RNA editing patterns but not cortex Alu RNA editing patterns. J Alzheimers Dis 2025; 103:1216-1225. [PMID: 39865681 DOI: 10.1177/13872877241313054] [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: 01/28/2025]
Abstract
BACKGROUND Endogenous Alu RNAs form double-stranded RNAs recognized by double-stranded RNA sensors and activate IRF and NF-kB transcriptional paths and innate immunity. Deamination of adenosines to inosines by the ADAR family of enzymes, a process termed A-to-I editing, disrupts double-stranded RNA structure and prevents innate immune activation. Innate immune activation is observed in Alzheimer's disease, the most common form of dementia. We have previously reported loss of A-to-I editing in hippocampus vasculature, but no change in cortex or cortex vasculature, associated with Alzheimer's disease. OBJECTIVE Here, we investigated the status of Alu RNA A-to-I editing in cortex extracellular vesicles in Alzheimer's disease. METHODS We used existing RNA-seq data sets and the SPRINT software package to determine levels of Alu RNA A-to-I editing in cortex extracellular vesicles in Alzheimer's disease and control groups and compared these editing profiles to those found in both total cortex and hippocampus vasculature. RESULTS We find substantial loss of Alu A-to-I editing in cortex extracellular vesicles in Alzheimer's disease. By measuring editing patterns on a gene-by-gene basis, we determined that editing patterns in cortex extracellular vesicles resemble editing patterns in hippocampus vasculature rather than total cortex. CONCLUSIONS We conclude that hippocampus vasculature unedited Alu RNAs are packaged in extracellular vesicles, travel to the cortex, deliver their cargo and stimulate innate immunity and alter other basic biological processes contributing to Alzheimer's disease progression.
Collapse
Affiliation(s)
- Philip S Crooke
- Department of Mathematics, Vanderbilt University, Nashville, TN, USA
| | - John T Tossberg
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas M Aune
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
18
|
Sannigrahi A, De N, Bhunia D, Bhadra J. Peptide nucleic acids: Recent advancements and future opportunities in biomedical applications. Bioorg Chem 2025; 155:108146. [PMID: 39817998 DOI: 10.1016/j.bioorg.2025.108146] [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: 08/07/2024] [Revised: 12/27/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
Peptide nucleic acids (PNA), synthetic molecules comprising a peptide-like backbone and natural and unnatural nucleobases, have garnered significant attention for their potential applications in gene editing and other biomedical fields. The unique properties of PNA, particularly enhanced stability/specificity/affinity towards targeted DNA and RNA sequences, achieved significant attention recently for gene silencing, gene correction, antisense therapy, drug delivery, biosensing and other various diagnostic aspects. This review explores the structure, properties, and potential of PNA in transforming genetic engineering including potent biomedical challenges. In Addition, we explore future perspectives and potential limitations of PNA-based technologies, highlighting the need for further research and development to fully realize their therapeutic and biotechnological potential.
Collapse
Affiliation(s)
- Achinta Sannigrahi
- University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Nayan De
- Institute for System Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Debmalya Bhunia
- Cold Spring Harbor Laboratory, 1 Bungtown Rd, Cold Spring Harbor, NY 11724, USA.
| | - Jhuma Bhadra
- Department of Chemistry, Sarojini Naidu College for Women, Kolkata 700028, India.
| |
Collapse
|
19
|
Marx V. Taking control with RNA. Nat Methods 2025; 22:226-230. [PMID: 39905158 DOI: 10.1038/s41592-025-02596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
|
20
|
Miller CM, Morrison JH, Bankers L, Dran R, Kendrick JM, Briggs E, Ferguson VL, Poeschla EM. ADAR1 haploinsufficiency and sustained viral RdRp dsRNA synthesis synergize to dysregulate RNA editing and cause multi-system interferonopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634124. [PMID: 39896491 PMCID: PMC11785089 DOI: 10.1101/2025.01.21.634124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Sensing of viral double-stranded RNA by MDA5 triggers abundant but transient interferon-stimulated gene (ISGs) expression. If dsRNA synthesis is made persistent by transgenically expressing a picornaviral RNA-dependent RNA polymerase (RdRp) in mice, lifelong MDA5 activation and marked, global ISG upregulation result. This confers robust protection from viral diseases but in contrast to numerous other chronic MDA5 hyperactivation states, the mice suffer no autoimmune consequences. Here we find they further confound expectations by being resistant to a strong autoimmunity (lupus) provocation. However, knockout of one allele of Adar , which by itself is also well-tolerated, breaks the protective state and results in a severe disease that resembles interferonopathies caused by MDA5 gain-of-function mutations. In Adar +/- RdRp transgenic mice, A-to-I editing is both dysregulated and increased (numbers of genes and sites). This dsRNA-driven, MDA5-wild type model establishes that viral polymerase-sourced dsRNA can drive interferonopathy pathogenesis and illuminates the autoimmunity preventing role of ADAR1, while the ADAR1-intact viral RdRp model distinctively uncouples chronic MDA5 hyperactivity and autoinflammatory disease.
Collapse
|
21
|
Akira A, Levanon E, Ben Aroya S. Leveraging Saccharomyces cerevisiae for ADAR research: From high-yield purification to high-throughput screening and therapeutic applications. Methods Enzymol 2025; 710:1-18. [PMID: 39870441 DOI: 10.1016/bs.mie.2024.11.026] [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: 01/29/2025]
Abstract
Saccharomyces cerevisiae, a model eukaryotic organism with a rich history in research and industry, has become a pivotal tool for studying Adenosine Deaminase Acting on RNA (ADAR) enzymes despite lacking these enzymes endogenously. This chapter reviews the diverse methodologies harnessed using yeast to elucidate ADAR structure and function, emphasizing its role in advancing our understanding of RNA editing. Initially, Saccharomyces cerevisiae was instrumental in the high-yield purification of ADARs, addressing challenges associated with enzyme stability and activity in other systems. The chapter highlights the successful application of yeast in high-throughput screening platforms that identify key structural motifs and substrate preferences of ADARs, showcasing its utility in revealing complex enzyme mechanics. Furthermore, we discuss the development of yeast-based systems to optimize guide RNA sequences for site-directed RNA editing (SDRE), demonstrating how these systems can be employed to refine therapeutic strategies targeting genetic mutations. Additionally, exogenous expression of ADARs from various species in yeast has shed light on enzyme potency and substrate recognition across different temperatures, offering insights into evolutionary adaptations. Overall, Saccharomyces cerevisiae has proven to be an invaluable asset in ADAR research, facilitating significant advances in our understanding of RNA editing mechanisms and therapeutic applications.
Collapse
Affiliation(s)
- Adi Akira
- Life Science, Bar Ilan University, Ramat Gan, Israel
| | - Erez Levanon
- Life Science, Bar Ilan University, Ramat Gan, Israel
| | | |
Collapse
|
22
|
Yang X, Woldemichael K, Guo X, Zhao S, Qian Y, Huang ZJ. CellREADR: An ADAR-based RNA sensor-actuator device. Methods Enzymol 2025; 710:207-227. [PMID: 39870446 DOI: 10.1016/bs.mie.2024.11.027] [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: 01/29/2025]
Abstract
RNAs are central mediators of genetic information flow and gene regulation that underlie diverse cell types and cell states across species. Thus, methods that can sense and respond to RNA profiles in living cells will have broad applications in biology and medicine. CellREADR - Cell access through RNA sensing by Endogenous ADAR (adenosine deaminase acting on RNA), is a programmable RNA sensor-actuator technology that couples the detection of a cell-defining RNA to the translation of an effector protein to monitor and manipulate the cell. The CellREADR RNA device consists of a 5' sensor region complementary to a cellular RNA and a 3' protein payload coding region. Payload translation is gated by the removal of a STOP codon in the sensor region upon base pairing with the cognate cellular RNA through an ADAR-mediated A-to-I editing mechanism ubiquitous to metazoan cells. CellREADR thus represents a new generation of programmable RNA device for monitoring and manipulating animal cells in ways that are simple, versatile, and generalizable across tissues and species. Here, we describe a detailed procedure for implementing CellREADR experiments in cell culture systems and in animals. The procedure includes sensor and payload design, cloning, validation and characterization in mammalian cell cultures. The in vivo protocol focuses on AAV-based delivery of CellREADR through expression vectors using brain tissue as an example. We describe current best practices and various experimental controls.
Collapse
Affiliation(s)
- Xiaolu Yang
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, United States
| | - Kehali Woldemichael
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, United States
| | - Xiao Guo
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Shengli Zhao
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, United States
| | - Yongjun Qian
- College of Future technology, Peking-Tsinghua Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, P.R. China
| | - Z Josh Huang
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, United States; Department of Biomedical Engineering, Duke University, Durham, NC, United States.
| |
Collapse
|
23
|
Qin PP, Chen PR, Tan L, Chu X, Ye BC, Yin BC. Programming ADAR-recruiting hairpin RNA sensor to detect endogenous molecules. Nucleic Acids Res 2025; 53:gkae1146. [PMID: 39673485 PMCID: PMC11724285 DOI: 10.1093/nar/gkae1146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 10/25/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024] Open
Abstract
RNA editing leveraging ADARs (adenosine deaminases acting on RNA) shows promising potential for in vivo biosensing beyond gene therapy. However, current ADAR sensors sense only a single target of RNA transcripts, thus limiting their use in different biosensing scenarios. Here, we report a hairpin RNA sensor that exploits new mechanisms to generate intramolecular duplex substrates for efficient ADAR recruitment and editing and apply it to detection of various intracellular molecules, including messenger RNA, small molecules and proteins. We utilize the base pairing interactions between neighbouring bases for enhanced stability, as well as the reverse effects to sense RNA transcripts and single-nucleotide variants with high sensitivity and specificity, irrespective of sequence requirement for complementarity to an UAG stop codon. In addition, we integrate RNA aptamers into the hairpin RNA sensor to realize the detection of the primary energy-supplying molecule, ATP, and a transcription factor, nuclear factor-kappa B (NF-κB), in live cells via a simple conformational change for programming the activation of hairpin RNA. This sensor not only broadens the detection of applicable molecules, but also offers potential for diverse cell manipulation.
Collapse
Affiliation(s)
- Pei-Pei Qin
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, No.18 Chao Wang Road, Gongshu District, Hangzhou 310014, China
| | - Pin-Ru Chen
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, No.18 Chao Wang Road, Gongshu District, Hangzhou 310014, China
| | - Liu Tan
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, No.18 Chao Wang Road, Gongshu District, Hangzhou 310014, China
| | - Xiaohe Chu
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, No.18 Chao Wang Road, Gongshu District, Hangzhou 310014, China
| | - Bang-Ce Ye
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, No.18 Chao Wang Road, Gongshu District, Hangzhou 310014, China
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, No.130 Meilong Road, Xuhui District, Shanghai 200237, China
| | - Bin-Cheng Yin
- Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, No.18 Chao Wang Road, Gongshu District, Hangzhou 310014, China
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, No.130 Meilong Road, Xuhui District, Shanghai 200237, China
- School of Chemistry and Chemical Engineering, Shihezi University, No.221 North Fourth Road, Uighur autonomous region, Shihezi 832000, Xinjiang, China
| |
Collapse
|
24
|
D’Addabbo P, Cohen-Fultheim R, Twersky I, Fonzino A, Silvestris DA, Prakash A, Mazzacuva PL, Vizcaino JA, Green A, Sweeney B, Yates A, Lussi Y, Luo J, Martin MJ, Eisenberg E, Levanon EY, Pesole G, Picardi E. REDIportal: toward an integrated view of the A-to-I editing. Nucleic Acids Res 2025; 53:D233-D242. [PMID: 39588754 PMCID: PMC11701558 DOI: 10.1093/nar/gkae1083] [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: 09/17/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
A-to-I RNA editing is the most common non-transient epitranscriptome modification. It plays several roles in human physiology and has been linked to several disorders. Large-scale deep transcriptome sequencing has fostered the characterization of A-to-I editing at the single nucleotide level and the development of dedicated computational resources. REDIportal is a unique and specialized database collecting ∼16 million of putative A-to-I editing sites designed to face the current challenges of epitranscriptomics. Its running version has been enriched with sites from the TCGA project (using data from 31 studies). REDIportal provides an accurate, sustainable and accessible tool enriched with interconnections with widespread ELIXIR core resources such as Ensembl, RNAcentral, UniProt and PRIDE. Additionally, REDIportal now includes information regarding RNA editing in putative double-stranded RNAs, relevant for the immune-related roles of editing, as well as an extended catalog of recoding events. Finally, we report a reliability score per site calculated using a deep learning model trained using a huge collection of positive and negative instances. REDIportal is available at http://srv00.recas.ba.infn.it/atlas/.
Collapse
Affiliation(s)
- Pietro D’Addabbo
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, via Orabona 4, 70125, Bari, Italy
| | - Roni Cohen-Fultheim
- Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, 52900, Israel
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 52900, Israel
| | - Itamar Twersky
- Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, 52900, Israel
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 52900, Israel
| | - Adriano Fonzino
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, via Orabona 4, 70125, Bari, Italy
| | - Domenico Alessandro Silvestris
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, via Orabona 4, 70125, Bari, Italy
| | - Ananth Prakash
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Pietro Luca Mazzacuva
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, National Research Council, via Amendola 122/O, 70126, Bari, Italy
| | - Juan Antonio Vizcaino
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Andrew Green
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Blake Sweeney
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Andy Yates
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Yvonne Lussi
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Jie Luo
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Maria-Jesus Martin
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| | - Eli Eisenberg
- School of Physics and Astronomy, Tel Aviv University, Tel Aviv, 699781, Israel
| | - Erez Y Levanon
- Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, 52900, Israel
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 52900, Israel
| | - Graziano Pesole
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, via Orabona 4, 70125, Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, National Research Council, via Amendola 122/O, 70126, Bari, Italy
| | - Ernesto Picardi
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, via Orabona 4, 70125, Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, National Research Council, via Amendola 122/O, 70126, Bari, Italy
| |
Collapse
|
25
|
Lamb E, Pant D, Yang B, Hundley HA. A probe-based capture enrichment method for detection of A-to-I editing in low abundance transcripts. Methods Enzymol 2025; 710:55-75. [PMID: 39870451 DOI: 10.1016/bs.mie.2024.11.033] [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: 01/29/2025]
Abstract
Exactly two decades ago, the ability to use high-throughput RNA sequencing technology to identify sites of editing by ADARs was employed for the first time. Since that time, RNA sequencing has become a standard tool for researchers studying RNA biology and led to the discovery of RNA editing sites present in a multitude of organisms, across tissue types, and in disease. However, transcriptome-wide sequencing is not without limitations. Most notably, RNA sequencing depth of a given transcript is correlated with expression, and sequencing depth impacts the ability to robustly detect RNA editing events. This chapter focuses on a method for enrichment of low-abundance transcripts that can facilitate more efficient sequencing and detection of RNA editing events. An important note is that while we describe aspects of the protocol important for capturing intron-containing transcripts, this probe-based enrichment method could be easily modified to assess editing within any low-abundance transcript. We also provide some perspectives on the current limitations as well as important future directions for expanding this technology to gain more insights into how RNA editing can impact transcript diversity.
Collapse
Affiliation(s)
- Emma Lamb
- Genome, Cell and Developmental Biology Graduate Program, Indiana University, Bloomington, Indiana, United States
| | - Dyuti Pant
- Department of Biology, Indiana University, Bloomington, Indiana, United States
| | - Boyoon Yang
- Biochemistry Graduate Program, Indiana University, Bloomington, Indiana, United States
| | - Heather A Hundley
- Department of Biology, Indiana University, Bloomington, Indiana, United States.
| |
Collapse
|
26
|
Fishman A, Lamm AT. Obstacles in quantifying A-to-I RNA editing by Sanger sequencing. Methods Enzymol 2025; 710:285-302. [PMID: 39870450 DOI: 10.1016/bs.mie.2024.11.032] [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: 01/29/2025]
Abstract
Adenosine-to-Inosine (A-to-I) RNA editing is the most prevalent type of RNA editing, in which adenosine within a completely or largely double-stranded RNA (dsRNA) is converted to inosine by deamination. RNA editing was shown to be involved in many neurological diseases and cancer; therefore, detection of A-to-I RNA editing and quantitation of editing levels are necessary for both basic and clinical biomedical research. While high-throughput sequencing (HTS) is widely used for global detection of editing events, Sanger sequencing is the method of choice for precise characterization of editing site clusters (hyper-editing) and for comparing levels of editing at a particular site under different environmental conditions, developmental stages, genetic backgrounds, or disease states. To detect A-to-I editing events and quantify them using Sanger sequencing, RNA samples are reverse transcribed, cDNA is amplified using gene-specific primers, and then sequenced. The chromatogram outputs are then compared to the genomic DNA sequence. As editing occurs in the context of dsRNA, the reverse transcription step is performed at a temperature as high as 65 °C, using thermostable reverse transcriptase to open double-stranded structures. However, this measure alone is insufficient for transcripts possessing long stems comprised of hundreds of nucleotide pairs. Consequently, the editing levels detected by Sanger sequencing are significantly lower than those obtained by HTS, and the amplification yield is low. We suggest that the reverse transcription is biased towards unedited transcripts, and the severity of the bias is dependent on the transcript's secondary structure. Here, we show how this bias can be significantly reduced to allow reliable detection of editing levels and sufficient product yield.
Collapse
Affiliation(s)
- Alla Fishman
- Faculty of Biology, Technion - Israel Institute of Technology, Technion City, Haifa, Israel.
| | - Ayelet T Lamm
- Faculty of Biology, Technion - Israel Institute of Technology, Technion City, Haifa, Israel.
| |
Collapse
|
27
|
Schultz S, Gomard-Henshaw K, Muller M. RNA Modifications and Their Role in Regulating KSHV Replication and Pathogenic Mechanisms. J Med Virol 2025; 97:e70140. [PMID: 39740054 DOI: 10.1002/jmv.70140] [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: 10/31/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Kaposi's sarcoma-associated herpesvirus is an oncogenic gammaherpesvirus that plays a major role in several human malignancies, including Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. The complexity of KSHV biology is reflected in the sophisticated regulation of its biphasic life cycle, consisting of a quiescent latent phase and virion-producing lytic replication. KSHV expresses coding and noncoding RNAs, including microRNAs and long noncoding RNAs, which play crucial roles in modulating viral gene expression, immune evasion, and intercellular communication. Recent studies have highlighted the importance of RNA modifications, also known as the epitranscriptome, in regulating KSHV-encoded RNAs, adding a novel layer of posttranscriptional control previously unknown. These RNA modifications, such as N6-methyladenosine, A-to-I editing, and N4-acetylcytidine, are involved in fine-tuning KSHV gene expression during both latency and lytic replication. Understanding the role of RNA modifications in KSHV infection is essential for revealing new regulatory mechanisms and identifying therapeutic opportunities. Targeting these RNA modifications could serve as a strategy to disrupt key viral processes, offering promising insights into KSHV pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- S Schultz
- Microbiology Department, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - K Gomard-Henshaw
- Microbiology Department, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - M Muller
- Microbiology Department, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
28
|
Garcia-Segura P, Llop-Peiró A, Novau-Ferré N, Mestres-Truyol J, Saldivar-Espinoza B, Pujadas G, Garcia-Vallvé S. SARS-CoV-2 main protease (M-pro) mutational profiling: An insight into mutation coldspots. Comput Biol Med 2025; 184:109344. [PMID: 39531923 DOI: 10.1016/j.compbiomed.2024.109344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/20/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
SARS-CoV-2 and the COVID-19 pandemic have marked a milestone in the history of scientific research worldwide. To ensure that treatments are successful in the mid-long term, it is crucial to characterize SARS-CoV-2 mutations, as they might lead to viral resistance. Data from >5,700,000 SARS-CoV-2 genomes available at GISAID was used to report SARS-CoV-2 mutations. Given the pivotal role of its main protease (M-pro) in virus replication, a detailed analysis of SARS-CoV-2 M-pro mutations was conducted, with particular attention to mutation-resistant residues or mutation coldspots, defined as those residues that have mutated in five or fewer genomes. 32 mutation coldspots were identified, most of which mediate interprotomer interactions or funneling interaction networks from the substrate-binding site towards the dimerization surface and vice versa. Besides, mutation coldspots were virtually conserved in all main proteases from other CoVs. Our results provide valuable information about key residues to M-pro structure that could be useful in rational target-directed drug design and establish a solid groundwork based on mutation analyses for the inhibition of M-pro dimerization, with a potential applicability to future coronavirus outbreaks.
Collapse
Affiliation(s)
- Pol Garcia-Segura
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Ariadna Llop-Peiró
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Nil Novau-Ferré
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Júlia Mestres-Truyol
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Bryan Saldivar-Espinoza
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Gerard Pujadas
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain
| | - Santiago Garcia-Vallvé
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| |
Collapse
|
29
|
Rehwinkel J, Mehdipour P. ADAR1: from basic mechanisms to inhibitors. Trends Cell Biol 2025; 35:59-73. [PMID: 39030076 PMCID: PMC11718369 DOI: 10.1016/j.tcb.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/21/2024]
Abstract
Adenosine deaminase acting on RNA 1 (ADAR1) converts adenosine to inosine in double-stranded RNA (dsRNA) molecules, a process known as A-to-I editing. ADAR1 deficiency in humans and mice results in profound inflammatory diseases characterised by the spontaneous induction of innate immunity. In cells lacking ADAR1, unedited RNAs activate RNA sensors. These include melanoma differentiation-associated gene 5 (MDA5) that induces the expression of cytokines, particularly type I interferons (IFNs), protein kinase R (PKR), oligoadenylate synthase (OAS), and Z-DNA/RNA binding protein 1 (ZBP1). Immunogenic RNAs 'defused' by ADAR1 may include transcripts from repetitive elements and other long duplex RNAs. Here, we review these recent fundamental discoveries and discuss implications for human diseases. Some tumours depend on ADAR1 to escape immune surveillance, opening the possibility of unleashing anticancer therapies with ADAR1 inhibitors.
Collapse
Affiliation(s)
- Jan Rehwinkel
- Medical Research Council Translational Immune Discovery Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK.
| | - Parinaz Mehdipour
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK.
| |
Collapse
|
30
|
Yabuki A, Fujii C, Yazaki E, Tame A, Mizuno K, Obayashi Y, Takao Y. Massive RNA Editing in Ascetosporean Mitochondria. Microbes Environ 2025; 40:ME24070. [PMID: 40090735 PMCID: PMC11946409 DOI: 10.1264/jsme2.me24070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/24/2024] [Indexed: 03/18/2025] Open
Abstract
Ascetosporeans are parasitic protists of invertebrates. A deep sequencing ana-lysis of species within the orders Mikrocytida, Paramyxida, and Haplosporida using metagenomic approaches revealed that their mitochondria were functionally reduced and their organellar genomes were lacking. Ascetosporeans belonging to the order Paradinida have not been sequenced, and the nature of their mitochondria remains unclear. We herein established two cultures of Paradinida and conducted DNA and RNA sequencing ana-lyses. The results obtained indicate that mitochondrial function in paradinids was not reduced and their organellar genomes were retained. In contrast, their mitochondrial genomes were involved in massive A-to-I and C-to-U substitution types of RNA editing. All edits in protein-coding genes were nonsynonymous substitutions, and likely had a restorative function against negative mutations. Furthermore, we detected possible sequences of DYW type of pentatricopeptide repeat (PPR-DYW) protein and a homologue of adenosine deaminase acting on RNA (ADAR-like), which are key enzymes for C-to-U and A-to-I substitutions, respectively. An immunofluorescence ana-lysis showed that ADAR-like of paradinids may specifically localize within mitochondria. These results expand our knowledge of the diversity and complexity of organellar RNA editing phenomena.
Collapse
Affiliation(s)
- Akinori Yabuki
- Japan Agency for Marine-Earth Science and Technology, Yokosuka, Kanagawa 237–0061, Japan
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980–8572 Japan
- Advanced Institute for Marine Ecosystem Change (WPI-AIMEC), Yokohama, Kanagawa 236–0001, Japan
| | - Chihaya Fujii
- Japan Agency for Marine-Earth Science and Technology, Yokosuka, Kanagawa 237–0061, Japan
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 980–8572 Japan
| | - Euki Yazaki
- Research Center for Advanced Analysis, National Agriculture and Food Research Organization, Tsukuba, 305–8518 Japan
| | - Akihiro Tame
- Marine Works Japan Ltd., Yokosuka, Kanagawa 237–0063, Japan
| | - Keiko Mizuno
- Japan Agency for Marine-Earth Science and Technology, Yokosuka, Kanagawa 237–0061, Japan
| | - Yumiko Obayashi
- Center for Marine Environmental Studies, Ehime University, Matsuyama, Ehime 790–8577, Japan
| | - Yoshitake Takao
- Department of Marine Science and Technology, Faculty of Marine Science and Technology, Fukui Prefecture University, Obama, Fukui 917–0003, Japan
| |
Collapse
|
31
|
Cheng L, Liu Z, Shen C, Xiong Y, Shin SY, Hwang Y, Yang S, Chen Z, Zhang X. A Wonderful Journey: The Diverse Roles of Adenosine Deaminase Action on RNA 1 (ADAR1) in Central Nervous System Diseases. CNS Neurosci Ther 2025; 31:e70208. [PMID: 39753993 PMCID: PMC11702419 DOI: 10.1111/cns.70208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/03/2024] [Accepted: 12/20/2024] [Indexed: 01/14/2025] Open
Abstract
BACKGROUND Adenosine deaminase action on RNA 1 (ADAR1) can convert the adenosine in double-stranded RNA (dsRNA) molecules into inosine in a process known as A-to-I RNA editing. ADAR1 regulates gene expression output by interacting with RNA and other proteins; plays important roles in development, including growth; and is linked to innate immunity, tumors, and central nervous system (CNS) diseases. RESULTS In recent years, the role of ADAR1 in tumors has been widely discussed, but its role in CNS diseases has not been reviewed. It is worth noting that recent studies have shown ADAR1 has great potential in the treatment of neurodegenerative diseases, but the mechanisms are still unclear. Therefore, it is necessary to elaborate on the role of ADAR1 in CNS diseases. CONCLUSIONS Here, we focus on the effects and mechanisms of ADAR1 on CNS diseases such as Aicardi-AicardiGoutières syndrome, Alzheimer's disease, Parkinson's disease, glioblastoma, epilepsy, amyotrophic lateral sclerosis, and autism. We also evaluate the impact of ADAR1-based treatment strategies on these diseases, with a particular focus on the development and treatment strategies of new technologies such as microRNAs, nanotechnology, gene editing, and stem cell therapy. We hope to provide new directions and insights for the future development of ADAR1 gene editing technology in brain science and the treatment of CNS diseases.
Collapse
Affiliation(s)
- Lin Cheng
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Ziying Liu
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Chunxiao Shen
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Yinyi Xiong
- Department of RehabilitationAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Sang Yol Shin
- Department of Emergency Medical TechnologyWonkwang University College of MedicineIksanJeonbuk‐doRepublic of Korea
| | - Yong Hwang
- Department of Emergency MedicineWonkwang University College of MedicineIksanJeonbuk‐doRepublic of Korea
| | - Seung‐Bum Yang
- Department of ParamedicineWonkwang Health Science UniversityIksanJeonbuk‐doRepublic of Korea
| | - Zhiying Chen
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Xiaorong Zhang
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
- Department of PathologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| |
Collapse
|
32
|
Checa-Robles FJ, Salvetat N, Cayzac C, Menhem M, Favier M, Vetter D, Ouna I, Nani JV, Hayashi MAF, Brietzke E, Weissmann D. RNA Editing Signatures Powered by Artificial Intelligence: A New Frontier in Differentiating Schizophrenia, Bipolar, and Schizoaffective Disorders. Int J Mol Sci 2024; 25:12981. [PMID: 39684694 DOI: 10.3390/ijms252312981] [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: 10/22/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Mental health disorders are devastating illnesses, often misdiagnosed due to overlapping clinical symptoms. Among these conditions, bipolar disorder, schizophrenia, and schizoaffective disorder are particularly difficult to distinguish, as they share alternating positive and negative mood symptoms. Accurate and timely diagnosis of these diseases is crucial to ensure effective treatment and to tailor therapeutic management to each individual patient. In this context, it is essential to move beyond standard clinical assessment and employ innovative approaches to identify new biomarkers that can be reliably quantified. We previously identified a panel of RNA editing biomarkers capable of differentiating healthy controls from depressed patients and, among depressed patients, those with major depressive disorder and those with bipolar disorder. In this study, we integrated Adenosine-to-Inosine RNA editing blood biomarkers with clinical data through machine learning algorithms to establish specific signatures for bipolar disorder and schizophrenia spectrum disorders. This groundbreaking study paves the way for the application of RNA editing in other psychiatric disorders, such as schizophrenia and schizoaffective disorder. It represents a first proof-of-concept and provides compelling evidence for the establishment of an RNA editing signature for the diagnosis of these psychiatric conditions.
Collapse
Affiliation(s)
- Francisco J Checa-Robles
- ALCEDIAG, Parc Euromédecine, 34184 Montpellier Cedex 4, France
- Sys2Diag, UMR 9005 CNRS/ALCEN, Parc Euromédecine, 34184 Montpellier Cedex 4, France
| | - Nicolas Salvetat
- ALCEDIAG, Parc Euromédecine, 34184 Montpellier Cedex 4, France
- Sys2Diag, UMR 9005 CNRS/ALCEN, Parc Euromédecine, 34184 Montpellier Cedex 4, France
| | - Christopher Cayzac
- ALCEDIAG, Parc Euromédecine, 34184 Montpellier Cedex 4, France
- Sys2Diag, UMR 9005 CNRS/ALCEN, Parc Euromédecine, 34184 Montpellier Cedex 4, France
| | - Mary Menhem
- ALCEDIAG, Parc Euromédecine, 34184 Montpellier Cedex 4, France
- Sys2Diag, UMR 9005 CNRS/ALCEN, Parc Euromédecine, 34184 Montpellier Cedex 4, France
| | - Mathieu Favier
- ALCEDIAG, Parc Euromédecine, 34184 Montpellier Cedex 4, France
- Sys2Diag, UMR 9005 CNRS/ALCEN, Parc Euromédecine, 34184 Montpellier Cedex 4, France
| | - Diana Vetter
- ALCEDIAG, Parc Euromédecine, 34184 Montpellier Cedex 4, France
- Sys2Diag, UMR 9005 CNRS/ALCEN, Parc Euromédecine, 34184 Montpellier Cedex 4, France
| | - Ilhème Ouna
- Sys2Diag, UMR 9005 CNRS/ALCEN, Parc Euromédecine, 34184 Montpellier Cedex 4, France
| | - João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo CEP 04044-20, Brazil
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto CEP 14040-900, Brazil
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo CEP 04044-20, Brazil
- National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto CEP 14040-900, Brazil
| | - Elisa Brietzke
- Department of Psychiatry, School of Medicine, Queen's University, Kingston, ON K7L 7X3, Canada
| | - Dinah Weissmann
- ALCEDIAG, Parc Euromédecine, 34184 Montpellier Cedex 4, France
- Sys2Diag, UMR 9005 CNRS/ALCEN, Parc Euromédecine, 34184 Montpellier Cedex 4, France
| |
Collapse
|
33
|
He D, Niu C, Bai R, Chen N, Cui J. ADAR1 Promotes Invasion and Migration and Inhibits Ferroptosis via the FAK/AKT Pathway in Colorectal Cancer. Mol Carcinog 2024; 63:2401-2413. [PMID: 39239920 DOI: 10.1002/mc.23818] [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: 05/06/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/07/2024]
Abstract
The role of adenosine deaminase acting on RNA1 (ADAR1) in colorectal cancer (CRC) is poorly understood. This study investigated the roles and underlying molecular mechanisms of ADAR1 and its isoforms, explored the correlations between ADAR1 expression and the immune microenvironment and anticancer drug sensitivity, and examined the potential synergy of using ADAR1 expression and clinical parameters to determine the prognosis of CRC patients. CRC samples showed significant upregulation of ADAR1, and high ADAR1 expression was correlated with poor prognosis. Silencing ADAR1 inhibited the proliferation, invasion, and migration of CRC cells and induced ferroptosis by suppressing FAK/AKT activation, and the results of rescue assays were consistent with these mechanisms. Both ADAR1-p110 and ADAR1-p150 were demonstrated to regulate the FAK/AKT pathway, with ADAR1-p110 playing a particularly substantial role. In evaluating the prognosis of CRC patients, combining ADAR1 expression with clinical parameters produced a substantial synergistic effect. The in vivo tumorigenesis of CRC was significantly inhibited by silencing ADAR1. Furthermore, ADAR1 expression was positively correlated with tumor mutational burden (TMB) and microsatellite status (p < 0.05), indicating that ADAR1 plays a complex role in CRC immunotherapy. In conclusion, ADAR1 plays oncogenic roles in CRC both in vitro and in vivo, potentially by inhibiting ferroptosis via downregulation of the FAK/AKT pathway. Thus, ADAR1 serves as a potential prognostic biomarker and a promising target for CRC therapy.
Collapse
Affiliation(s)
- Dongsheng He
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Chao Niu
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Rilan Bai
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Naifei Chen
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiuwei Cui
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
34
|
Zheng C, Ma L, Song F, Tian L, Cai W, Li H, Duan Y. Comparative genomic analyses reveal evidence for adaptive A-to-I RNA editing in insect Adar gene. Epigenetics 2024; 19:2333665. [PMID: 38525798 DOI: 10.1080/15592294.2024.2333665] [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: 11/07/2023] [Accepted: 03/17/2024] [Indexed: 03/26/2024] Open
Abstract
Although A-to-I RNA editing leads to similar effects to A-to-G DNA mutation, nonsynonymous RNA editing (recoding) is believed to confer its adaptiveness by 'epigenetically' regulating proteomic diversity in a temporospatial manner, avoiding the pleiotropic effect of genomic mutations. Recent discoveries on the evolutionary trajectory of Ser>Gly auto-editing site in insect Adar gene demonstrated a selective advantage to having an editable codon compared to uneditable ones. However, apart from pure observations, quantitative approaches for justifying the adaptiveness of individual RNA editing sites are still lacking. We performed a comparative genomic analysis on 113 Diptera species, focusing on the Adar Ser>Gly auto-recoding site in Drosophila. We only found one species having a derived Gly at the corresponding site, and this occurrence was significantly lower than genome-wide random expectation. This suggests that the Adar Ser>Gly site is unlikely to be genomically replaced with G during evolution, and thus indicating the advantage of editable status over hardwired genomic alleles. Similar trends were observed for the conserved Ile>Met recoding in gene Syt1. In the light of evolution, we established a comparative genomic approach for quantitatively justifying the adaptiveness of individual editing sites. Priority should be given to such adaptive editing sites in future functional studies.
Collapse
Affiliation(s)
- Caiqing Zheng
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Ling Ma
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Fan Song
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Li Tian
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Wanzhi Cai
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Hu Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yuange Duan
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
35
|
Zhao T, Ma L, Xu S, Cai W, Li H, Duan Y. Narrowing down the candidates of beneficial A-to-I RNA editing by comparing the recoding sites with uneditable counterparts. Nucleus 2024; 15:2304503. [PMID: 38286757 PMCID: PMC10826634 DOI: 10.1080/19491034.2024.2304503] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/08/2024] [Indexed: 01/31/2024] Open
Abstract
Adar-mediated adenosine-to-inosine (A-to-I) RNA editing mainly occurs in nucleus and diversifies the transcriptome in a flexible manner. It has been a challenging task to identify beneficial editing sites from the sea of total editing events. The functional Ser>Gly auto-recoding site in insect Adar gene has uneditable Ser codons in ancestral nodes, indicating the selective advantage to having an editable status. Here, we extended this case study to more metazoan species, and also looked for all Drosophila recoding events with potential uneditable synonymous codons. Interestingly, in D. melanogaster, the abundant nonsynonymous editing is enriched in the codons that have uneditable counterparts, but the Adar Ser>Gly case suggests that the editable orthologous codons in other species are not necessarily edited. The use of editable versus ancestral uneditable codon is a smart way to infer the selective advantage of RNA editing, and priority might be given to these editing sites for functional studies due to the feasibility to construct an uneditable allele. Our study proposes an idea to narrow down the candidates of beneficial recoding sites. Meanwhile, we stress that the matched transcriptomes are needed to verify the conservation of editing events during evolution.
Collapse
Affiliation(s)
- Tianyou Zhao
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Ling Ma
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Shiwen Xu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Wanzhi Cai
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Hu Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yuange Duan
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
36
|
Yang X, Woldemichael K, Hover J, Zhao S, Guo X, Qian Y, Gillis J, Huang ZJ. RNA programmable cell targeting and manipulation with CellREADR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625312. [PMID: 39651297 PMCID: PMC11623600 DOI: 10.1101/2024.11.26.625312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Methods that provide specific, easy, and scalable experimental access to animal cell types and cell states will have broad applications in biology and medicine. CellREADR - Cell access through RNA sensing by Endogenous ADAR (adenosine deaminase acting on RNA), is a programmable RNA sensor-actuator technology that couples the detection of a cell-defining RNA to the translation of an effector protein to monitor and manipulate the cell. The CellREADR RNA device consists of a 5' sensor region complementary to a cellular RNA and a 3' payload coding region; payload translation is gated by the removal of a STOP codon in the sensor region upon base pairing with the cognate cellular RNA through an ADAR-mediated A- to-I editing mechanism ubiquitous to metazoan cells. CellREADR thus highlights the potential for RNA-based monitoring and manipulation of animal cells in ways that are simple, versatile, and generalizable across tissues and species. Here, we describe a detailed protocol for implementing CellREADR experiments in cell cultures and in animals. The procedure includes sensor and payload design, cloning, validation and characterization in mammalian cell cultures. The in vivo animal protocol focuses on AAV-based delivery of CellREADR using brain tissue as examples. We describe current best practices, various experimental controls and trouble-shooting steps. Beginning from sensor design, the validation of RNA sensor-actuators in cell cultures can be completed in 2-3 weeks. The construction of AAV vectors and their in vivo validation and characterization will take additional 6-8 weeks.
Collapse
|
37
|
Heraud-Farlow JE, Taylor SR, Chalk AM, Escudero A, Hu SB, Goradia A, Sun T, Li Q, Nikolic I, Li JB, Fidalgo M, Guallar D, Simpson KJ, Walkley CR. GGNBP2 regulates MDA5 sensing triggered by self double-stranded RNA following loss of ADAR1 editing. Sci Immunol 2024; 9:eadk0412. [PMID: 39576872 DOI: 10.1126/sciimmunol.adk0412] [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: 07/30/2023] [Revised: 06/05/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024]
Abstract
Adenosine-to-inosine (A-to-I) editing of double-stranded RNA (dsRNA) by ADAR1 is an essential modifier of the immunogenicity of cellular dsRNA. The role of MDA5 in sensing unedited cellular dsRNA and the downstream activation of type I interferon (IFN) signaling are well established. However, we have an incomplete understanding of pathways that modify the response to unedited dsRNA. We performed a genome-wide CRISPR screen and showed that GGNBP2, CNOT10, and CNOT11 interact and regulate sensing of unedited cellular dsRNA. We found that GGNBP2 acts between dsRNA transcription and its cytoplasmic sensing by MDA5. GGNBP2 loss prevented induction of type I IFN and autoinflammation after the loss of ADAR1 editing activity by modifying the subcellular distribution of endogenous A-to-I editing substrates and reducing cytoplasmic dsRNA load. These findings reveal previously undescribed pathways to modify diseases associated with ADAR mutations and may be determinants of response or resistance to small-molecule ADAR1 inhibitors.
Collapse
Affiliation(s)
- Jacki E Heraud-Farlow
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, St. Vincent's Hospital, Melbourne Medical School, University of Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Scott R Taylor
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Alistair M Chalk
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Adriana Escudero
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC) - Health Research Institute (IDIS), Santiago de Compostela 15782, Spain
| | - Shi-Bin Hu
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Ankita Goradia
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Tao Sun
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Qin Li
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Iva Nikolic
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology and Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jin Billy Li
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Miguel Fidalgo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC) - Health Research Institute (IDIS), Santiago de Compostela 15782, Spain
| | - Diana Guallar
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela (USC) - Health Research Institute (IDIS), Santiago de Compostela 15782, Spain
| | - Kaylene J Simpson
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology and Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Carl R Walkley
- St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, St. Vincent's Hospital, Melbourne Medical School, University of Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
38
|
Wang Q, Huang Yang M, Yu S, Chen Y, Wang K, Zhang Y, Zhao R, Li J. An improved transcriptome annotation reveals asymmetric expression and distinct regulation patterns in allotetraploid common carp. Commun Biol 2024; 7:1542. [PMID: 39567764 PMCID: PMC11579021 DOI: 10.1038/s42003-024-07177-3] [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: 11/23/2023] [Accepted: 10/30/2024] [Indexed: 11/22/2024] Open
Abstract
In allotetraploid common carp, protein-coding homoeologs presented divergent expression levels between the two subgenomes. However, whether subgenome dominance occurs in other transcriptional and post-transcriptional events remains unknown. Using Illumina RNA sequencing and PacBio full-length sequencing, we refined the common carp transcriptome annotation and explored differences in four transcriptional and post-transcriptional events between the two subgenomes. The results revealed that the B subgenome presented more alternative splicing events, as did lncRNAs and circRNAs. However, the expression levels, tissue specificity, sequence features, and functions of lncRNAs and circRNAs did not significantly differ between the two subgenomes, suggesting a common regulatory mechanism shared by the two subgenomes. Furthermore, both the number and base substitution frequency of RNA editing events were greater in the B subgenome. Functional analyses of these transcriptional events also revealed subgenome bias. Genes that undergo alternative splicing in the A subgenome participate in more biological processes, and lncRNA targets show a preference between subgenomes. CircRNA host genes in the B subgenome were associated with more biological functions, and RNA editing preferentially occurred in noncoding regions or led to nonsynonymous mutations in the B subgenome. Taken together, the refined transcriptome annotation revealed complicated and imbalanced expression strategies in allotetraploid common carp.
Collapse
Affiliation(s)
- Qi Wang
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing, China
| | - Meidi Huang Yang
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing, China
| | - Shuangting Yu
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing, China
| | - Yingjie Chen
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing, China
| | - Kaikuo Wang
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing, China
| | - Yan Zhang
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing, China
| | - Ran Zhao
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing, China
| | - Jiongtang Li
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing, China.
| |
Collapse
|
39
|
Dailamy A, Lyu W, Nourreddine S, Tong M, Rainaldi J, McDonald D, Panwala R, Muotri A, Breen MS, Zhang K, Mali P. Charting and probing the activity of ADARs in human development and cell-fate specification. Nat Commun 2024; 15:9818. [PMID: 39537590 PMCID: PMC11561244 DOI: 10.1038/s41467-024-53973-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Adenosine deaminases acting on RNA (ADARs) impact diverse cellular processes and pathological conditions, but their functions in early cell-fate specification remain less understood. To gain insights here, we began by charting time-course RNA editing profiles in human organs from fetal to adult stages. Next, we utilized hPSC differentiation to experimentally probe ADARs, harnessing brain organoids as neural specific, and teratomas as pan-tissue developmental models. We show that time-series teratomas faithfully recapitulate fetal developmental trends, and motivated by this, conducted pan-tissue, single-cell CRISPR-KO screens of ADARs in teratomas. Knocking out ADAR leads to a global decrease in RNA editing across all germ-layers. Intriguingly, knocking out ADAR leads to an enrichment of adipogenic cells, revealing a role for ADAR in human adipogenesis. Collectively, we present a multi-pronged framework charting time-resolved RNA editing profiles and coupled ADAR perturbations in developmental models, thereby shedding light on the role of ADARs in cell-fate specification.
Collapse
Affiliation(s)
- Amir Dailamy
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Weiqi Lyu
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Sami Nourreddine
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Michael Tong
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Joseph Rainaldi
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, San Diego, CA, USA
| | - Daniella McDonald
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, San Diego, CA, USA
| | - Rebecca Panwala
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| | - Alysson Muotri
- Department of Pediatrics and Cellular & Molecular Medicine, University of California San Diego, San Diego, CA, USA
| | | | - Kun Zhang
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Prashant Mali
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
40
|
Wei ZY, Wang LP, Gao D, Zhu L, Wu JF, Shi J, Li YN, Tang XD, Feng YM, Pan XB, Jin YY, Liu YS, Chen JH. Bulk and single-cell RNA-seq analyses reveal canonical RNA editing associated with microglia homeostasis and its role in sepsis-associated encephalopathy. Neuroscience 2024; 560:167-180. [PMID: 39293730 DOI: 10.1016/j.neuroscience.2024.09.027] [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: 05/28/2024] [Revised: 08/25/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Previous studies have demonstrated the roles of both microglia homeostasis and RNA editing in sepsis-associated encephalopathy (SAE), yet their relationship remains to be elucidated. In this study, we analyzed bulk and single-cell RNA-seq (scRNA) datasets containing 107 brain tissue and microglia samples from mice with microglial depletion and repopulation to explore canonical RNA editing associated with microglia homeostasis and evaluate its role in SAE. Analysis of mouse brain RNA-Seq revealed hallmarks of microglial repopulation, including peak expressions of Apobec1 and Apobec3 at Day 5 of repopulation and dramatically altered B2m RNA editing. Significant time-dependent changes in brain RNA editing during microglial depletion and repopulation were primarily observed in synapse-related genes, such as Tbc1d24 and Slc1a2. ScRNA-Seq revealed heterogeneous RNA editing among microglia subpopulations and their distinct changes associated with microglia homeostasis. Moreover, repopulated microglia from lipopolysaccharide (LPS)-induced sepsis mice exhibited intensified up-regulation of Apobec1 and Apobec3, with distinct RNA editing responses to LPS, mainly involved in immune-related pathways. The hippocampus from sepsis mice induced by peritoneal contamination and infection showed upregulated Apobec1 and Apobec3 expression, and altered RNA editing in immune-related genes, such as B2m and Mier1, and nervous-related lncRNA Meg3 and Snhg11, both of which were repressed by microglial depletion. Furthermore, the expression of complement-related genes, such as C4b and Cd47, was substantially correlated with RNA editing activity in microglia homeostasis and SAE. Our study demonstrates canonical RNA editing associated with microglia homeostasis and provides new insights into its potential role in SAE.
Collapse
Affiliation(s)
- Zhi-Yuan Wei
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214022, China
| | - Li-Ping Wang
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Di Gao
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Lin Zhu
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jun-Fan Wu
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jia Shi
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yu-Ning Li
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiao-Dan Tang
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yan-Meng Feng
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xu-Bin Pan
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yun-Yun Jin
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yan-Shan Liu
- Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, Jiangsu 214023, China.
| | - Jian-Huan Chen
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China; Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214022, China.
| |
Collapse
|
41
|
Jiang S, Peng J, Saneela S, Shi R, Wang D, Tang Q, Shi X, Meng Y. Bipartite nuclear localization sequence is indispensable for nuclear import and stability of self-dimerization of ADARa in Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 174:104190. [PMID: 39389319 DOI: 10.1016/j.ibmb.2024.104190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
The conservative post-transcriptional modification in mammals and Drosophila is adenosine-to-inosine (A-to-I) deamination in double-stranded RNA, catalyzed by RNA-editing enzymes known as adenosine deaminases acting on RNA (ADARs). The traditional nuclear import pathway for ADARs involves the recognition of a putative classical nuclear localization sequence (NLS) by importin α4 and α5. In our previous research, ADAR in silkworm, Bombyx mori (BmADARa) was confirmed predominantly located in the nucleus. However, the location of the NLS in BmADARa and its impact on nuclear import and self-dimerization remained unclear. Utilizing NLS prediction software, we predicted the presence of a bipartite NLS within the amino-terminal, 85 amino acids of BmADARa (N85). This prediction was validated through point mutation, which demonstrated that the bipartite NLS could directly mediate nuclear import of BmADARa. Co-immunoprecipitation analysis revealed that BmADARa is mainly dependent on BmKaryopherin α3 (homologous to mammalian importin α4) for nuclear import, although both BmKaryopherin α3 and BmImportin α5 could recognize bipartite NLS. The N-terminal truncated mutants and the bipartite NLS mutants of BmADARa suggest that the bipartite NLS is the major nuclear import site and a crucial structure for self-dimerization of BmADARa. In conclusion, the N-terminal bipartite NLS of BmADARa is recognized by BmKaryopherin α3 and BmImportin α5, facilitating its nuclear import. This promotes BmADARa self-dimerization and maintains the stability of dimerization, thereby enhancing its editing efficiency on target substrates. The results of this research demonstrate the role of bipartite NLS in BmADARa editing and laying a foundation for further research on the regulation of BmADARa in the growth and development in B. mori.
Collapse
Affiliation(s)
- Song Jiang
- School of Life Sciences, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Hefei, 230036, China; Anhui International Joint Research and Development Center of Sericulture Resources Utilization, Hefei, 230036, China
| | - Junzhe Peng
- School of Life Sciences, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Hefei, 230036, China
| | - Syeda Saneela
- School of Life Sciences, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Anhui International Joint Research and Development Center of Sericulture Resources Utilization, Hefei, 230036, China
| | - Ruoyun Shi
- School of Life Sciences, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China
| | - Daoming Wang
- School of Life Sciences, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Anhui International Joint Research and Development Center of Sericulture Resources Utilization, Hefei, 230036, China
| | - Qingheng Tang
- School of Life Sciences, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Hefei, 230036, China
| | - Xiaming Shi
- School of Life Sciences, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Hefei, 230036, China; Anhui International Joint Research and Development Center of Sericulture Resources Utilization, Hefei, 230036, China
| | - Yan Meng
- School of Life Sciences, Anhui Agricultural University, 130 West Changjiang Road, Hefei, 230036, China; Anhui Province Key Laboratory of Resource Insect Biology and Innovative Utilization, Hefei, 230036, China; Anhui International Joint Research and Development Center of Sericulture Resources Utilization, Hefei, 230036, China.
| |
Collapse
|
42
|
Liang Z, Walkley CR, Heraud-Farlow JE. A-to-I RNA editing and hematopoiesis. Exp Hematol 2024; 139:104621. [PMID: 39187172 DOI: 10.1016/j.exphem.2024.104621] [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: 06/07/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Adenosine-to-inosine (A-to-I) RNA editing plays essential roles in modulating normal development and homeostasis. This process is catalyzed by adenosine deaminase acting on RNA (ADAR) family proteins. The most well-understood biological processes modulated by A-to-I editing are innate immunity and neurological development, attributed to ADAR1 and ADAR2, respectively. A-to-I editing by ADAR1 is also critical in regulating hematopoiesis. This review will focus on the role of A-to-I RNA editing and ADAR enzymes, particularly ADAR1, during normal hematopoiesis in humans and mice. Furthermore, we will discuss Adar1 mouse models that have been developed to understand the contribution of ADAR1 to hematopoiesis and its role in innate immune pathways.
Collapse
Affiliation(s)
- Zhen Liang
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Medicine, Eastern Hill Precinct, Melbourne Medical School, University of Melbourne, Fitzroy, Victoria, Australia
| | - Carl R Walkley
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Medicine, Eastern Hill Precinct, Melbourne Medical School, University of Melbourne, Fitzroy, Victoria, Australia.
| | - Jacki E Heraud-Farlow
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Medicine, Eastern Hill Precinct, Melbourne Medical School, University of Melbourne, Fitzroy, Victoria, Australia.
| |
Collapse
|
43
|
Weldy CS, Li Q, Monteiro JP, Guo H, Galls D, Gu W, Cheng PP, Ramste M, Li D, Palmisano BT, Sharma D, Worssam MD, Zhao Q, Bhate A, Kundu RK, Nguyen T, Li JB, Quertermous T. Smooth muscle expression of RNA editing enzyme ADAR1 controls activation of RNA sensor MDA5 in atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602569. [PMID: 39026721 PMCID: PMC11257488 DOI: 10.1101/2024.07.08.602569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Mapping the genomic architecture of complex disease has been predicated on the understanding that genetic variants influence disease risk through modifying gene expression. However, recent discoveries have revealed that a significant burden of disease heritability in common autoinflammatory disorders and coronary artery disease is mediated through genetic variation modifying post-transcriptional modification of RNA through adenosine-to-inosine (A-to-I) RNA editing. This common RNA modification is catalyzed by ADAR enzymes, where ADAR1 edits specific immunogenic double stranded RNA (dsRNA) to prevent activation of the double strand RNA (dsRNA) sensor MDA5 ( IFIH1 ) and stimulation of an interferon stimulated gene (ISG) response. Multiple lines of human genetic data indicate impaired RNA editing and increased dsRNA sensing by MDA5 to be an important mechanism of coronary artery disease (CAD) risk. Here, we provide a crucial link between observations in human genetics and mechanistic cell biology leading to progression of CAD. Through analysis of human atherosclerotic plaque, we implicate the vascular smooth muscle cell (SMC) to have a unique requirement for RNA editing, and that ISG induction occurs in SMC phenotypic modulation, implicating MDA5 activation. Through culture of human coronary artery SMCs, generation of a conditional SMC specific Adar1 deletion mouse model on a pro-atherosclerosis background with additional constitutive deletion of MDA5 ( Ifih1 ), and with incorporation of single cell RNA sequencing cellular profiling, we further show that Adar1 controls SMC phenotypic state by regulating Mda5 activation, is required to maintain vascular integrity, and controls progression of atherosclerosis and vascular calcification. Through this work, we describe a fundamental mechanism of CAD, where cell type and context specific RNA editing and sensing of dsRNA mediates disease progression, bridging our understanding of human genetics and disease causality. One Sentence Summary Smooth muscle expression of RNA editing enzyme ADAR1 regulates activation of double strand RNA sensor MDA5 in novel mechanism of atherosclerosis.
Collapse
|
44
|
Young AA, Bohlin HE, Pierce JR, Cottrell KA. Suppression of double-stranded RNA sensing in cancer: molecular mechanisms and therapeutic potential. Biochem Soc Trans 2024; 52:2035-2045. [PMID: 39221819 PMCID: PMC11555700 DOI: 10.1042/bst20230727] [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: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapy has emerged as a therapeutic option for many cancers. For some tumors, immune checkpoint inhibitors show great efficacy in promoting anti-tumor immunity. However, not all tumors respond to immunotherapies. These tumors often exhibit reduced inflammation and are resistant to checkpoint inhibitors. Therapies that turn these 'cold' tumors 'hot' could improve the efficacy and applicability of checkpoint inhibitors, and in some cases may be sufficient on their own to promote anti-tumor immunity. One strategy to accomplish this goal is to activate innate immunity pathways within the tumor. Here we describe how this can be accomplished by activating double-stranded RNA (dsRNA) sensors. These sensors evolved to detect and respond to dsRNAs arising from viral infection but can also be activated by endogenous dsRNAs. A set of proteins, referred to as suppressors of dsRNA sensing, are responsible for preventing sensing 'self' dsRNA and activating innate immunity pathways. The mechanism of action of these suppressors falls into three categories: (1) Suppressors that affect mature RNAs through editing, degradation, restructuring, or binding. (2) Suppressors that affect RNA processing. (3) Suppressors that affect RNA expression. In this review we highlight suppressors that function through each mechanism, provide examples of the effects of disrupting those suppressors in cancer cell lines and tumors, and discuss the therapeutic potential of targeting these proteins and pathways.
Collapse
Affiliation(s)
- Addison A. Young
- Department of Biochemistry, Purdue University, West Lafayette, IN, U.S.A
| | - Holly E. Bohlin
- Department of Biochemistry, Purdue University, West Lafayette, IN, U.S.A
| | - Jackson R. Pierce
- Department of Biochemistry, Purdue University, West Lafayette, IN, U.S.A
| | - Kyle A. Cottrell
- Department of Biochemistry, Purdue University, West Lafayette, IN, U.S.A
| |
Collapse
|
45
|
Torkler P, Sauer M, Schwartz U, Corbacioglu S, Sommer G, Heise T. LoDEI: a robust and sensitive tool to detect transcriptome-wide differential A-to-I editing in RNA-seq data. Nat Commun 2024; 15:9121. [PMID: 39443485 PMCID: PMC11500352 DOI: 10.1038/s41467-024-53298-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
RNA editing is a highly conserved process. Adenosine deaminase acting on RNA (ADAR) mediated deamination of adenosine (A-to-I editing) is associated with human disease and immune checkpoint control. Functional implications of A-to-I editing are currently of broad interest to academic and industrial research as underscored by the fast-growing number of clinical studies applying base editors as therapeutic tools. Analyzing the dynamics of A-to-I editing, in a biological or therapeutic context, requires the sensitive detection of differential A-to-I editing, a currently unmet need. We introduce the local differential editing index (LoDEI) to detect differential A-to-I editing in RNA-seq datasets using a sliding-window approach coupled with an empirical q value calculation that detects more A-to-I editing sites at the same false-discovery rate compared to existing methods. LoDEI is validated on known and novel datasets revealing that the oncogene MYCN increases and that a specific small non-coding RNA reduces A-to-I editing.
Collapse
Affiliation(s)
- Phillipp Torkler
- Faculty of Computer Science, Deggendorf Institute of Technology, Dieter-Görlitz-Platz 1, Deggendorf, 94469, Bavaria, Germany
| | - Marina Sauer
- Department for Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensbug, 93053, Bavaria, Germany
| | - Uwe Schwartz
- NGS Analysis Center, University of Regensburg, Universitätsstraße 31, Regensburg, 93053, Bavaria, Germany
| | - Selim Corbacioglu
- Department for Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensbug, 93053, Bavaria, Germany
| | - Gunhild Sommer
- Department for Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensbug, 93053, Bavaria, Germany
| | - Tilman Heise
- Department for Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensbug, 93053, Bavaria, Germany.
| |
Collapse
|
46
|
Wulff T, Hahnke K, Lécrivain AL, Schmidt K, Ahmed-Begrich R, Finstermeier K, Charpentier E. Dynamics of diversified A-to-I editing in Streptococcus pyogenes is governed by changes in mRNA stability. Nucleic Acids Res 2024; 52:11234-11253. [PMID: 39087550 PMCID: PMC11472039 DOI: 10.1093/nar/gkae629] [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: 09/14/2023] [Revised: 07/01/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
Adenosine-to-inosine (A-to-I) RNA editing plays an important role in the post-transcriptional regulation of eukaryotic cell physiology. However, our understanding of the occurrence, function and regulation of A-to-I editing in bacteria remains limited. Bacterial mRNA editing is catalysed by the deaminase TadA, which was originally described to modify a single tRNA in Escherichia coli. Intriguingly, several bacterial species appear to perform A-to-I editing on more than one tRNA. Here, we provide evidence that in the human pathogen Streptococcus pyogenes, tRNA editing has expanded to an additional tRNA substrate. Using RNA sequencing, we identified more than 27 editing sites in the transcriptome of S. pyogenes SF370 and demonstrate that the adaptation of S. pyogenes TadA to a second tRNA substrate has also diversified the sequence context and recoding scope of mRNA editing. Based on the observation that editing is dynamically regulated in response to several infection-relevant stimuli, such as oxidative stress, we further investigated the underlying determinants of editing dynamics and identified mRNA stability as a key modulator of A-to-I editing. Overall, our findings reveal the presence and diversification of A-to-I editing in S. pyogenes and provide novel insights into the plasticity of the editome and its regulation in bacteria.
Collapse
Affiliation(s)
- Thomas F Wulff
- Max Planck Unit for the Science of Pathogens, 10117 Berlin, Germany
| | - Karin Hahnke
- Max Planck Unit for the Science of Pathogens, 10117 Berlin, Germany
| | | | - Katja Schmidt
- Max Planck Unit for the Science of Pathogens, 10117 Berlin, Germany
| | | | | | - Emmanuelle Charpentier
- Max Planck Unit for the Science of Pathogens, 10117 Berlin, Germany
- Institute for Biology, Humboldt University Berlin, 10115 Berlin, Germany
| |
Collapse
|
47
|
Zhang P, Zhang W, Li J, Liu H, Yu Y, Yang X, Jiang W. Host-dependent C-to-U RNA editing in SARS-CoV-2 creates novel viral genes with optimized expressibility. Front Cell Infect Microbiol 2024; 14:1476605. [PMID: 39445213 PMCID: PMC11496155 DOI: 10.3389/fcimb.2024.1476605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024] Open
Abstract
Rampant C-to-U RNA editing drives the mutation and evolution of SARS-CoV-2. While much attention has been paid to missense mutations, the C-to-U events leading to AUG and thus creating novel ORFs were uninvestigated. By utilizing the public time-course mutation data from the worldwide SARS-CoV-2 population, we systematically identified the "AUG-gain mutations" caused by C-to-U RNA editing. Synonymous mutations were of special focus. A total of 58 synonymous C-to-U sites are able to create out-of-frame AUG in coding sequence (CDS). These 58 synonymous sites showed significantly higher allele frequency (AF) and increasing rate (dAF/dt) than other C-to-U synonymous sites in the SARS-CoV-2 population, suggesting that these 58 AUG-gain events conferred additional benefits to the virus and are subjected to positive selection. The 58 predicted new ORFs created by AUG-gain events showed the following advantages compared to random expectation: they have longer lengths, higher codon adaptation index (CAI), higher Kozak scores, and higher tRNA adaptation index (tAI). The 58 putatively novel ORFs have high expressibility and are very likely to be functional, providing an explanation for the positive selection on the 58 AUG-gain mutations. Our study proposed a possible mechanism of the emergence of de novo genes in SARS-CoV-2. This idea should be helpful in studying the mutation and evolution of SARS-CoV-2.
Collapse
Affiliation(s)
- Pirun Zhang
- The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wenli Zhang
- Qingdao Mental Health Center, Qingdao, Shandong, China
| | - Jiahuan Li
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, Shandong, China
| | - Huiying Liu
- Qingdao Hospital of Traditional Chinese Medicine, Qingdao Haici Hospital, Qingdao, Shandong, China
| | - Yantong Yu
- Pulmonary and Critical Care Medicine Department 2, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, Shandong, China
| | - Xiaoping Yang
- Qingdao Hospital of Traditional Chinese Medicine, Qingdao Haici Hospital, Qingdao, Shandong, China
- College of First Clinical Medical, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenqing Jiang
- Qingdao Hospital of Traditional Chinese Medicine, Qingdao Haici Hospital, Qingdao, Shandong, China
- Pulmonary and Critical Care Medicine Department 2, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, Shandong, China
| |
Collapse
|
48
|
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
|
49
|
Liu J, Lu F. Beyond simple tails: poly(A) tail-mediated RNA epigenetic regulation. Trends Biochem Sci 2024; 49:846-858. [PMID: 39004583 DOI: 10.1016/j.tibs.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024]
Abstract
The poly(A) tail is an essential structural component of mRNA required for the latter's stability and translation. Recent technologies have enabled transcriptome-wide profiling of the length and composition of poly(A) tails, shedding light on their overlooked regulatory capacities. Notably, poly(A) tails contain not only adenine but also uracil, cytosine, and guanine residues. These findings strongly suggest that poly(A) tails could encode a wealth of regulatory information, similar to known reversible RNA chemical modifications. This review aims to succinctly summarize our current knowledge on the composition, dynamics, and regulatory functions of RNA poly(A) tails. Given their capacity to carry rich regulatory information beyond the genetic code, we propose the concept of 'poly(A) tail epigenetic information' as a new layer of RNA epigenetic regulation.
Collapse
Affiliation(s)
- Jingwen Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Falong Lu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
50
|
Werner A, Kanhere A, Wahlestedt C, Mattick JS. Natural antisense transcripts as versatile regulators of gene expression. Nat Rev Genet 2024; 25:730-744. [PMID: 38632496 DOI: 10.1038/s41576-024-00723-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: 03/07/2024] [Indexed: 04/19/2024]
Abstract
Long non-coding RNAs (lncRNAs) are emerging as a major class of gene products that have central roles in cell and developmental biology. Natural antisense transcripts (NATs) are an important subset of lncRNAs that are expressed from the opposite strand of protein-coding and non-coding genes and are a genome-wide phenomenon in both eukaryotes and prokaryotes. In eukaryotes, a myriad of NATs participate in regulatory pathways that affect expression of their cognate sense genes. Recent developments in the study of NATs and lncRNAs and large-scale sequencing and bioinformatics projects suggest that whether NATs regulate expression, splicing, stability or translation of the sense transcript is influenced by the pattern and degrees of overlap between the sense-antisense pair. Moreover, epigenetic gene regulatory mechanisms prevail in somatic cells whereas mechanisms dependent on the formation of double-stranded RNA intermediates are prevalent in germ cells. The modulating effects of NATs on sense transcript expression make NATs rational targets for therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | - John S Mattick
- University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|