Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.131
Filter
1.
Nat Commun ; 15(1): 6615, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103360

ABSTRACT

RNA base editing relies on the introduction of adenosine-to-inosine changes into target RNAs in a highly programmable manner in order to repair disease-causing mutations. Here, we propose that RNA base editing could be broadly applied to perturb protein function by removal of regulatory phosphorylation and acetylation sites. We demonstrate the feasibility on more than 70 sites in various signaling proteins and identify key determinants for high editing efficiency and potent down-stream effects. For the JAK/STAT pathway, we demonstrate both, negative and positive regulation. To achieve high editing efficiency over a broad codon scope, we applied an improved version of the SNAP-ADAR tool. The transient nature of RNA base editing enables the comparably fast (hours to days), dose-dependent (thus partial) and reversible manipulation of regulatory sites, which is a key advantage over DNA (base) editing approaches. In summary, PTM interference might become a valuable field of application of RNA base editing.


Subject(s)
Protein Processing, Post-Translational , RNA Editing , Humans , Phosphorylation , HEK293 Cells , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , RNA/metabolism , RNA/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Acetylation
2.
Cells ; 13(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39120288

ABSTRACT

Vascular smooth muscle cells (VSMCs) play a critical role in maintaining vascular integrity. VSMC dysfunction leads to numerous vascular diseases. Adenosine deaminases acting on RNA 1 (ADAR1), an RNA editing enzyme, has shown both RNA editing and non-editing functions. Global deletion of ADAR1 causes embryonic lethality, but the phenotype of homozygous ADAR1 deletion specifically in SMCs (ADAR1sm-/-) remains to be determined. By crossing ADAR1fl/fl mice with Myh11-CreERT2 mice followed by Tamoxifen induction, we found that ADAR1sm-/- leads to lethality in adult mice 14 days after the induction. Gross examination revealed extensive hemorrhage and detrimental vascular damage in different organs. Histological analyses revealed destruction of artery structural integrity with detachment of elastin laminae from VSMCs in ADAR1sm-/- aortas. Furthermore, ADAR1sm-/- resulted in severe VSMC apoptosis and mitochondrial dysfunction. RNA sequencing analyses of ADAR1sm-/- aorta segments demonstrated profound transcriptional alteration of genes impacting vascular health including a decrease in fibrillin-1 expression. More importantly, ADAR1sm-/- disrupts the elastin and fibrillin-1 interaction, a molecular event essential for artery structure. Our results indicate that ADAR1 plays a critical role in maintaining SMC survival and vascular stability and resilience.


Subject(s)
Adenosine Deaminase , Homeostasis , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Animals , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Aorta/metabolism , Aorta/pathology , Apoptosis/genetics , Fibrillin-1/genetics , Fibrillin-1/metabolism , Elastin/metabolism , Mice, Knockout , Mice, Inbred C57BL , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
3.
PLoS One ; 19(8): e0307450, 2024.
Article in English | MEDLINE | ID: mdl-39178184

ABSTRACT

Adenosine to inosine (A-to-I) RNA editing by ADAR1 has been implicated in maintaining self-tolerance, preventing autoimmunity, and mediating antiviral immunity. Foreign viral double-stranded RNA triggers rapid interferon response and activates ADAR1 in the host immune system. Emerging data points to a role of ADAR1 A-to-I editing in the inflammatory response associated with severe COVID-19 disease. We identify A-to-I editing events within human whole transcriptome data from SARS-CoV-2 infected individuals, non-infected individuals, and individuals with other viral illnesses from nasopharyngeal swabs. High levels of RNA editing in host cells are associated with low SARS-CoV-2 viral load (p = 9.27 E-06), suggesting an inhibitory effect of ADAR1 on viral infection. Additionally, we find differentially expressed genes associated with RNA-modifications and interferon response. Single cell RNA-sequencing analysis of SARS-CoV-2 infected nasopharyngeal swabs reveals that cytotoxic CD8 T cells upregulate ADAR1 in COVID-19 positive samples (p = 0.0269). We further reveal ADAR1 expression increases with CD4 and CD8 T cell activation, and knockdown of ADAR1 leads to apoptosis and aberrant IL-2 secretion. Together, our data suggests A-to-I RNA editing is required to maintain healthy homeostasis of activated T cells to combat SARS-CoV-2 infection.


Subject(s)
Adenosine Deaminase , COVID-19 , Homeostasis , RNA Editing , RNA-Binding Proteins , SARS-CoV-2 , Humans , COVID-19/immunology , COVID-19/virology , COVID-19/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , SARS-CoV-2/physiology , SARS-CoV-2/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Viral Load , Inosine/metabolism , Adenosine/metabolism , Lymphocyte Activation/immunology
4.
Sci Rep ; 14(1): 15395, 2024 07 04.
Article in English | MEDLINE | ID: mdl-38965255

ABSTRACT

The APOBEC/AID family is known for its mutator activity, and recent evidence also supports the potential impact of ADARs. Furthermore, the mutator impacts of APOBEC/ADAR mutations have not yet been investigated. Assessment of pancancer TCGA exomes identified enriched somatic variants among exomes with nonsynonymous APOBEC1, APOBEC3B, APOBEC3C, ADAR, and ADARB1 mutations, compared to exomes with synonymous ones. Principal component (PC) analysis reduced the number of potential players to eight in cancer exomes/genomes, and to five in cancer types. Multivariate regression analysis was used to assess the impact of the PCs on each COSMIC mutational signature among pancancer exomes/genomes and particular cancers, identifying several novel links, including SBS17b, SBS18, and ID7 mainly determined by APOBEC1 mRNA levels; SBS40, ID1, and ID2 by age; SBS3 and SBS16 by APOBEC3A/APOBEC3B mRNA levels; ID5 and DBS9 by DNA repair/replication (DRR) defects; and SBS7a-d, SBS38, ID4, ID8, ID13, and DBS1 by ultraviolet (UV) radiation/ADARB1 mRNA levels. APOBEC/ADAR mutations appeared to potentiate the impact of DRR defects on several mutational signatures, and some factors seemed to inversely affect certain signatures. These findings potentially implicate certain APOBEC/ADAR mutations/mRNA levels in distinct mutational signatures, particularly APOBEC1 mRNA levels in aging-related signatures and ADARB1 mRNA levels in UV radiation-related signatures.


Subject(s)
Adenosine Deaminase , Aging , Mutation , RNA, Messenger , RNA-Binding Proteins , Ultraviolet Rays , Humans , Ultraviolet Rays/adverse effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Aging/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , APOBEC-1 Deaminase/genetics , APOBEC-1 Deaminase/metabolism , APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Neoplasms/genetics , Exome
5.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(7): 849-852, 2024 Jul 10.
Article in Chinese | MEDLINE | ID: mdl-38946371

ABSTRACT

OBJECTIVE: To investigate the clinical and genetic features of a child with Dyschromatosis symmetrica hereditaria (DSH) and variant of the ADAR1 gene. METHODS: A child who was admitted to the Department of Dermatology of the First Affiliated Hospital of Zhengzhou University in June 2020 due to irregular pigmented maculopapular rash on the dorsum of hands was selected as the study subject. Whole exome sequencing (WES) was carried out for the child and his similarly affected father, and Sanger sequencing was used to verify the candidate variant. SWISS-MODEL was used to predict the secondary and tertiary structures of the wild-type and mutant ADAR1 proteins. RESULTS: The child, a 13-year-old boy, had symmetrical hyperpigmented and depigmented spots on the back of his hands and was clinically diagnosed with DSH. WES and Sanger sequencing results showed that he and his father had both harbored a heterozygous c.2858dup (p.T954Dfs*20) truncating variant in exon 10 of the ADAR1 gene. Based on the guidelines from the American College of Medical Genetics and Genomics, the variant was predicted as pathogenic (PVS1+PM2_Supporting+PM1+PP3). CONCLUSION: The c.2858dup (p.T954Dfs*20) variant of the ADAR1 gene probably underlay the DSH in this pedigree.


Subject(s)
Adenosine Deaminase , Pigmentation Disorders , RNA-Binding Proteins , Humans , Male , Adenosine Deaminase/genetics , Pigmentation Disorders/genetics , Pigmentation Disorders/congenital , RNA-Binding Proteins/genetics , Adolescent , Mutation , Exome Sequencing , Exons , Genetic Testing , Pedigree
6.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000531

ABSTRACT

Epitranscriptomics is a field that delves into post-transcriptional changes. Among these modifications, the conversion of adenosine to inosine, traduced as guanosine (A>I(G)), is one of the known RNA-editing mechanisms, catalyzed by ADARs. This type of RNA editing is the most common type of editing in mammals and contributes to biological diversity. Disruption in the A>I(G) RNA-editing balance has been linked to diseases, including several types of cancer. Drug resistance in patients with cancer represents a significant public health concern, contributing to increased mortality rates resulting from therapy non-responsiveness and disease progression, representing the greatest challenge for researchers in this field. The A>I(G) RNA editing is involved in several mechanisms over the immunotherapy and genotoxic drug response and drug resistance. This review investigates the relationship between ADAR1 and specific A>I(G) RNA-edited sites, focusing particularly on breast cancer, and the impact of these sites on DNA damage repair and the immune response over anti-cancer therapy. We address the underlying mechanisms, bioinformatics, and in vitro strategies for the identification and validation of A>I(G) RNA-edited sites. We gathered databases related to A>I(G) RNA editing and cancer and discussed the potential clinical and research implications of understanding A>I(G) RNA-editing patterns. Understanding the intricate role of ADAR1-mediated A>I(G) RNA editing in breast cancer holds significant promise for the development of personalized treatment approaches tailored to individual patients' A>I(G) RNA-editing profiles.


Subject(s)
Adenosine Deaminase , Breast Neoplasms , RNA Editing , RNA-Binding Proteins , Humans , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Female , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Adenosine/metabolism , Drug Resistance, Neoplasm/genetics , Inosine/metabolism , Inosine/genetics , Animals , Guanosine/metabolism , DNA Damage
7.
J Comput Aided Mol Des ; 38(1): 25, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39014124

ABSTRACT

Adenosine deaminases acting on RNA (ADARs) are pivotal RNA-editing enzymes responsible for converting adenosine to inosine within double-stranded RNA (dsRNA). Dysregulation of ADAR1 editing activity, often arising from genetic mutations, has been linked to elevated interferon levels and the onset of autoinflammatory diseases. However, understanding the molecular underpinnings of this dysregulation is impeded by the lack of an experimentally determined structure for the ADAR1 deaminase domain. In this computational study, we utilized homology modeling and the AlphaFold2 to construct structural models of the ADAR1 deaminase domain in wild-type and two pathogenic variants, R892H and Y1112F, to decipher the structural impact on the reduced deaminase activity. Our findings illuminate the critical role of structural complementarity between the ADAR1 deaminase domain and dsRNA in enzyme-substrate recognition. That is, the relative position of E1008 and K1120 must be maintained so that they can insert into the minor and major grooves of the substrate dsRNA, respectively, facilitating the flipping-out of adenosine to be accommodated within a cavity surrounding E912. Both amino acid replacements studied, R892H at the orthosteric site and Y1112F at the allosteric site, alter K1120 position and ultimately hinder substrate RNA binding.


Subject(s)
Adenosine Deaminase , Molecular Dynamics Simulation , RNA-Binding Proteins , Adenosine Deaminase/chemistry , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Humans , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mutation , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , RNA, Double-Stranded/genetics , Protein Conformation , RNA Editing
8.
Biochem Biophys Res Commun ; 726: 150213, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-38964186

ABSTRACT

The F11 receptor (F11R) gene encoding junctional adhesion molecule A has been associated with gastric cancer (GC) and colorectal cancer (CRC), in which its role and regulation remain to be further elucidated. Recently F11R was also identified as a potential target of adenosine-to-inosine (A-to-I) mediated by the adenosine deaminases acting on RNA (ADARs). Herein, using RNA-Seq and experimental validation, our current study revealed an F11R RNA trinucleotide over-edited by ADAR, with its regulation of gene expression and clinical significance in four GC and three CRC cohorts. Our results found an over-edited AAA trinucleotide in an AluSg located in the F11R 3'-untranslated region (3'-UTR), which showed editing levels correlated with elevated ADAR expression across all GC and CRC cohorts in our study. Overexpression and knockdown of ADAR in GC and CRC cells, followed by RNA-Seq and Sanger sequencing, confirmed the ADAR-mediated F11R 3'-UTR trinucleotide editing, which potentially disrupted an RBM45 binding site identified by crosslinking immunoprecipitation sequencing (CLIP-seq) and regulated F11R expression in luciferase reporter assays. Moreover, the F11R trinucleotide editing showed promising predictive performance for diagnosing GC and CRC across GC and CRC cohorts. Our findings thus highlight both the potential biological and clinical significance of an ADAR-edited F11R trinucleotide in GC and CRC, providing new insights into its application as a novel diagnostic biomarker for both cancers.


Subject(s)
Adenosine Deaminase , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , RNA Editing , RNA-Binding Proteins , Stomach Neoplasms , Humans , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cohort Studies , 3' Untranslated Regions/genetics , Cell Line, Tumor , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Male , Female
9.
Mol Biol Rep ; 51(1): 864, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073463

ABSTRACT

BACKGROUND: The study investigated the effect of co-administration of curcumin and donepezil on several markers of cognitive function (such as spatial memory, astrocyte activation, cholinesterase expressions) in the brain cortex and hippocampus of scopolamine-treated rats. METHOD AND RESULTS: For seven consecutive days, a pre-treatment of curcumin (50 mg/kg) and/or donepezil (2.5 mg/kg) was administered. On the seventh day, scopolamine (1 mg/kg) was administered to elicit cognitive impairment, 30 min before memory test was conducted. This was followed by evaluating changes in spatial memory, cholinesterase, and adenosine deaminase (ADA) activities, as well as nitric oxide (NO) level were determined. Additionally, RT-qPCR for glial fibrillary acidic protein (GFAP) and cholinesterase gene expressions was performed in the brain cortex and hippocampus. Also, GFAP immunohistochemistry  of the brain tissues for neuronal injury were performed in the brain cortex and hippocampus. In comparison to the control group, rats given scopolamine had impaired memory, higher levels of acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and ADA activities, as well as elevated markers of oxidative stress. In addition to enhanced GFAP immunoreactivity, there was also overexpression of the GFAP and BChE genes in the brain tissues. The combination of curcumin and donepezil was, however, observed to better ameliorate these impairments in comparison to the donepezil-administered rat group. CONCLUSION: Hence, this evidence provides more mechanisms to support the hypothesis that the concurrent administration of curcumin and donepezil mitigates markers of cognitive dysfunction in scopolamine-treated rat model.


Subject(s)
Acetylcholinesterase , Astrocytes , Curcumin , Donepezil , Glial Fibrillary Acidic Protein , Hippocampus , Scopolamine , Spatial Memory , Animals , Donepezil/pharmacology , Curcumin/pharmacology , Curcumin/administration & dosage , Scopolamine/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Rats , Male , Spatial Memory/drug effects , Acetylcholinesterase/metabolism , Acetylcholinesterase/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Brain/drug effects , Brain/metabolism , Rats, Wistar , Oxidative Stress/drug effects , Cholinesterases/metabolism , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Butyrylcholinesterase/metabolism , Butyrylcholinesterase/genetics , Nitric Oxide/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/administration & dosage
10.
J Neurol Sci ; 463: 123142, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39053342

ABSTRACT

Patients with amyotrophic lateral sclerosis (ALS) do not develop oculomotor disturbances and vesicorectal dysfunction until end-stage disease owing to the survival of certain motor neurons (MNs), including oculomotor neurons and MNs within Onuf's nucleus. In sporadic ALS, adenosine deaminase acting on RNA 2 (ADAR2)-mediated editing of GluA2 mRNA at the Q/R site is compromised in lower MNs. We previously developed genetically modified mice with a conditional knockout of ADAR2 in cholinergic neurons (ADAR2flox/flox/VAChT-Cre, Fast; AR2). These mice displayed slow and progressive lower motor neuron death with TAR DNA-binding protein 43 (TDP-43) pathology, attributable to insufficient editing at the GluA2 Q/R site due to ADAR2 deficiency. MN death was more common in fast-fatigable MNs owing to differential vulnerability under conditions of ADAR2 deficiency. Although facial and hypoglossal nerves were impaired in AR2 mice, cell death did not occur within the oculomotor nerve nucleus, as observed in patients with sporadic ALS. Since the basis for avoiding cystorectal damage in ALS is unknown, we compared the features of Onuf's nucleus MNs in 12-month-old AR2 mice with those in age-matched wild-type mice. Although the number of MNs was not significantly lower in AR2 mice, the neurons exhibited a shrunken morphology and TDP-43 pathology. Onuf's nucleus MNs could survive in an ADAR2-deficient state and mainly included fast fatigue-resistant (FR) and slow (S) MNs. In summary, FR and S MNs show increased resilience to ADAR2 deficiency, potentially participating in an important neuronal death avoidance mechanism in ALS.


Subject(s)
Adenosine Deaminase , Amyotrophic Lateral Sclerosis , Mice, Knockout , Motor Neurons , RNA-Binding Proteins , Animals , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Adenosine Deaminase/genetics , Adenosine Deaminase/deficiency , Adenosine Deaminase/metabolism , Motor Neurons/pathology , Motor Neurons/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mice , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Mice, Transgenic
12.
Fish Shellfish Immunol ; 151: 109743, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964433

ABSTRACT

Adenosine Deaminases Acting on RNA (ADARs) are evolutionarily conserved enzymes known to convert adenosine to inosine in double-stranded RNAs and participate in host-virus interactions. Conducting a meta-analysis of available transcriptome data, we identified and characterised eight ADAR transcripts in Chlamys farreri, a farmed marine scallop susceptible to Acute viral necrosis virus (AVNV) infections and mortality outbreaks. Accordingly, we identified six ADAR genes in the Zhikong scallop genome, revised previous gene annotations, and traced alternative splicing variants. In detail, each ADAR gene encodes a unique combination of functional domains, always including the Adenosine deaminase domain, RNA binding domains and, in one case, two copies of a Z-DNA binding domain. After phylogenetic analysis, five C. farreri ADARs clustered in the ADAR1 clade along with sequences from diverse animal phyla. Gene expression analysis indicated CF051320 as the most expressed ADAR, especially in the eye and male gonad. The other four ADAR1 genes and one ADAR2 gene exhibited variable expression levels, with CF105370 and CF051320 significantly increasing during early scallop development. ADAR-mediated single-base editing, evaluated across adult C. farreri tissues and developmental stages, was mainly detectable in intergenic regions (83 % and 85 %, respectively). Overall, the expression patterns of the six ADAR genes together with the editing and hyper-editing values computed on scallops RNA-seq samples support the adaptive value of ADAR1-mediated editing, particularly in the pre-settling larval stages.


Subject(s)
Adenosine Deaminase , Pectinidae , Phylogeny , RNA Editing , Animals , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Pectinidae/genetics , Pectinidae/immunology , Immunity, Innate/genetics , Gene Expression Profiling , Gene Expression Regulation/immunology , Amino Acid Sequence , Transcriptome , Sequence Alignment/veterinary
13.
Vaccine ; 42(21): 126153, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39060204

ABSTRACT

Vaccines and host genetic factors can influence the SARS-CoV-2 evolution and emergence of new variants. Even vaccinated cases get affected as virus continues to evolve, raising concerns about vaccine efficacy and the emergence of immune escape variants. Here, we have analyzed 2295 whole-genome sequences of SARS-CoV-2 collected from vaccinated and unvaccinated cases to evaluate the impact of vaccines on virus diversity within hosts. Our comparative analysis revealed a significant higher incidence of intra-host single nucleotides variants (iSNVs) in vaccinated cases compared to unvaccinated ones (p value<0.0001). Furthermore, we have found that specific mutational processes, including APOBEC (C > T) mediated and ADAR1 (A > G) mediated mutations, were found more prevalent in vaccinated cases. Vaccinated cases exhibited higher accumulation of nonsynonymous mutation than unvaccinated cases. Fixed iSNVs were predominantly located in the ORF1ab and spike genes, several key omicron defining immune escape variants S477N, Q493R, Q498R, Y505H, L452R, and N501Y were identified in the RBD domain of spike gene in vaccinated cases. Our findings suggest that vaccine plays an important role in the evolution of the virus genome. The virus genome acquires random mutations due to error-prone replication of the virus, host modification through APOBEC and ADAR1 mediated editing mechanism, and oxidative stress. These mutations become fixed in the viral population due to the selective pressure imposed by vaccination.


Subject(s)
COVID-19 Vaccines , COVID-19 , Evolution, Molecular , Immune Evasion , Mutation , SARS-CoV-2 , Vaccination , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Immune Evasion/genetics , COVID-19 Vaccines/immunology , Genome, Viral , Whole Genome Sequencing , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Adenosine Deaminase/genetics , Adenosine Deaminase/immunology
14.
Nucleic Acids Res ; 52(14): 8628-8642, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-38994565

ABSTRACT

Precise gene regulation and programmable RNA editing are vital RNA-level regulatory mechanisms. Gene repression tools grounded in small non-coding RNAs, microRNAs, and CRISPR-dCas proteins, along with RNA editing tools anchored in Adenosine Deaminases acting on RNA (ADARs), have found extensive application in molecular biology and cellular engineering. Here, we introduced a novel approach wherein we developed an EcCas6e mediated crRNA-mRNA annealing system for gene repression in Escherichia coli and RNA editing in Saccharomyces cerevisiae. We found that EcCas6e possesses inherent RNA annealing ability attributed to a secondary positively charged cleft, enhancing crRNA-mRNA hybridization and stability. Based on this, we demonstrated that EcCas6e, along with its cognate crRNA repeat containing a complementary region to the ribosome binding site of a target mRNA, effectively represses gene expression up to 25-fold. Furthermore, we demonstrated that multiple crRNAs can be easily assembled and can simultaneously target up to 13 genes. Lastly, the EcCas6e-crRNA system was developed as an RNA editing tool by fusing it with the ADAR2 deaminase domain. The EcCas6e-crRNA mediated gene repression and RNA editing tools hold broad applications for research and biotechnology.


Subject(s)
Escherichia coli , RNA Editing , RNA, Antisense , RNA, Messenger , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , RNA, Antisense/genetics , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , CRISPR-Cas Systems , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/genetics
15.
Genome Biol ; 25(1): 173, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956576

ABSTRACT

BACKGROUND: RNA-seq has brought forth significant discoveries regarding aberrations in RNA processing, implicating these RNA variants in a variety of diseases. Aberrant splicing and single nucleotide variants (SNVs) in RNA have been demonstrated to alter transcript stability, localization, and function. In particular, the upregulation of ADAR, an enzyme that mediates adenosine-to-inosine editing, has been previously linked to an increase in the invasiveness of lung adenocarcinoma cells and associated with splicing regulation. Despite the functional importance of studying splicing and SNVs, the use of short-read RNA-seq has limited the community's ability to interrogate both forms of RNA variation simultaneously. RESULTS: We employ long-read sequencing technology to obtain full-length transcript sequences, elucidating cis-effects of variants on splicing changes at a single molecule level. We develop a computational workflow that augments FLAIR, a tool that calls isoform models expressed in long-read data, to integrate RNA variant calls with the associated isoforms that bear them. We generate nanopore data with high sequence accuracy from H1975 lung adenocarcinoma cells with and without knockdown of ADAR. We apply our workflow to identify key inosine isoform associations to help clarify the prominence of ADAR in tumorigenesis. CONCLUSIONS: Ultimately, we find that a long-read approach provides valuable insight toward characterizing the relationship between RNA variants and splicing patterns.


Subject(s)
Haplotypes , Humans , Cell Line, Tumor , Polymorphism, Single Nucleotide , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Lung Neoplasms/genetics , RNA Splicing , Inosine/metabolism , Inosine/genetics , Sequence Analysis, RNA , Adenocarcinoma of Lung/genetics , RNA Editing , Software
16.
Cell Biol Toxicol ; 40(1): 57, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39060787

ABSTRACT

It is well established that sevoflurane exposure leads to widespread neuronal cell death in the developing brain. Adenosine deaminase acting on RNA-1 (ADAR1) dependent adenosine-to-inosine (A-to-I) RNA editing is dynamically regulated throughout brain development. The current investigation is designed to interrogate the contributed role of ADAR1 in developmental sevoflurane neurotoxicity. Herein, we provide evidence to show that developmental sevoflurane priming triggers neuronal pyroptosis, apoptosis and necroptosis (PANoptosis), and elicits the release of inflammatory factors including IL-1ß, IL-18, TNF-α and IFN-γ. Additionally, ADAR1-P150, but not ADAR1-P110, depresses cellular PANoptosis and inflammatory response by competing with Z-DNA/RNA binding protein 1 (ZBP1) for binding to Z-RNA in the presence of sevoflurane. Further investigation demonstrates that ADAR1-dependent A-to-I RNA editing mitigates developmental sevoflurane-induced neuronal PANoptosis. To restore RNA editing, we utilize adeno-associated virus (AAV) to deliver engineered circular ADAR-recruiting guide RNAs (cadRNAs) into cells, which is capable of recruiting endogenous adenosine deaminases to promote cellular A-to-I RNA editing. As anticipated, AAV-cadRNAs diminishes sevoflurane-induced cellular Z-RNA production and PANoptosis, which could be abolished by ADAR1-P150 shRNA transfection. Moreover, AAV-cadRNAs delivery ameliorates developmental sevoflurane-induced spatial and emotional cognitive deficits without influence on locomotor activity. Taken together, these results illustrate that ADAR1-P150 exhibits a prominent role in preventing ZBP1-dependent PANoptosis through A-to-I RNA editing in developmental sevoflurane neurotoxicity. Application of engineered cadRNAs to rectify the compromised ADAR1-dependent A-to-I RNA editing provides an inspiring direction for possible clinical preventions and therapeutics.


Subject(s)
Adenosine Deaminase , Adenosine , RNA Editing , RNA-Binding Proteins , Sevoflurane , Animals , Adenosine/metabolism , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Apoptosis/drug effects , Inosine/metabolism , Neurons/drug effects , Neurons/metabolism , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/metabolism , Pyroptosis/drug effects , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
17.
Genes (Basel) ; 15(7)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39062677

ABSTRACT

Adenosine-to-inosine (A-to-I) RNA editing is an important post-transcriptional modification mediated by the adenosine deaminases acting on RNA (ADAR) family of enzymes, expanding the transcriptome by altering selected nucleotides A to I in RNA molecules. Recently, A-to-I editing has been explored for correcting disease-causing mutations in RNA using therapeutic guide oligonucleotides to direct ADAR editing at specific sites. Humans have two active ADARs whose preferences and specificities are not well understood. To investigate their substrate specificity, we introduced hADAR1 and hADAR2, respectively, into Schizosaccharomyces pombe (S. pombe), which lacks endogenous ADARs, and evaluated their editing activities in vivo. Using transcriptome sequencing of S. pombe cultured at optimal growth temperature (30 °C), we identified 483 A-to-I high-confident editing sites for hADAR1 and 404 for hADAR2, compared with the non-editing wild-type control strain. However, these sites were mostly divergent between hADAR1 and hADAR2-expressing strains, sharing 33 common sites that are less than 9% for each strain. Their differential specificity for substrates was attributed to their differential preference for neighboring sequences of editing sites. We found that at the -3-position relative to the editing site, hADAR1 exhibits a tendency toward T, whereas hADAR2 leans toward A. Additionally, when varying the growth temperature for hADAR1- and hADAR2-expressing strains, we observed increased editing sites for them at both 20 and 35 °C, compared with them growing at 30 °C. However, we did not observe a significant shift in hADAR1 and hADAR2's preference for neighboring sequences across three temperatures. The vast changes in RNA editing sites at lower and higher temperatures were also observed for hADAR2 previously in budding yeast, which was likely due to the influence of RNA folding at these different temperatures, among many other factors. We noticed examples of longer lengths of dsRNA around the editing sites that induced editing at 20 or 35 °C but were absent at the other two temperature conditions. We found genes' functions can be greatly affected by editing of their transcripts, for which over 50% of RNA editing sites for both hADAR1 and hADAR2 in S. pombe were in coding sequences (CDS), with more than 60% of them resulting in amino acid changes in protein products. This study revealed the extensive differences in substrate selectivity between the two active human ADARS, i.e., ADAR1 and ADAR2, and provided novel insight when utilizing the two different enzymes for in vivo treatment of human genetic diseases using the RNA editing approach.


Subject(s)
Adenosine Deaminase , RNA Editing , RNA-Binding Proteins , Schizosaccharomyces , Schizosaccharomyces/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , RNA Editing/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Humans , Substrate Specificity , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Adenosine/metabolism , Adenosine/genetics , Inosine/genetics , Inosine/metabolism
18.
PLoS Pathog ; 20(6): e1012238, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38843141

ABSTRACT

Although lack of ADAR (adenosine deaminase acting on RNA) orthologs, genome-wide A-to-I editing occurs specifically during sexual reproduction in a number of filamentous ascomycetes, including Fusarium graminearum and Neurospora crassa. Unlike ADAR-mediated editing in animals, fungal A-to-I editing has a strong preference for hairpin loops and U at -1 position, which leads to frequent editing of UAG and UAA stop codons. Majority of RNA editing events in fungi are in the coding region and cause amino acid changes. Some of these editing events have been experimentally characterized for providing heterozygote and adaptive advantages in F. graminearum. Recent studies showed that FgTad2 and FgTad3, 2 ADAT (adenosine deaminase acting on tRNA) enzymes that normally catalyze the editing of A34 in the anticodon of tRNA during vegetative growth mediate A-to-I mRNA editing during sexual reproduction. Stage specificity of RNA editing is conferred by stage-specific expression of short transcript isoforms of FgTAD2 and FgTAD3 as well as cofactors such as AME1 and FIP5 that facilitate the editing of mRNA in perithecia. Taken together, fungal A-to-I RNA editing during sexual reproduction is catalyzed by ADATs and it has the same sequence and structural preferences with editing of A34 in tRNA.


Subject(s)
Adenosine Deaminase , RNA Editing , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Ascomycota/genetics , RNA, Fungal/genetics , RNA, Fungal/metabolism , Adenosine/metabolism , Adenosine/genetics , Inosine/metabolism , Inosine/genetics , Fusarium/genetics , Neurospora crassa/genetics
19.
Virus Res ; 346: 199413, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38848818

ABSTRACT

The conversion of Adenosine (A) to Inosine (I), by Adenosine Deaminases Acting on RNA or ADARs, is an essential post-transcriptional modification that contributes to proteome diversity and regulation in metazoans including humans. In addition to its transcriptome-regulating role, ADARs also play a major part in immune response to viral infection, where an interferon response activates interferon-stimulated genes, such as ADARp150, in turn dynamically regulating host-virus interactions. A previous report has shown that infection from reoviruses, despite strong activation of ADARp150, does not influence the editing of some of the major known editing targets, while likely editing others, suggesting a potentially nuanced editing pattern that may depend on different factors. However, the results were based on a handful of selected editing sites and did not cover the entire transcriptome. Thus, to determine whether and how reovirus infection specifically affects host ADAR editing patterns, we analyzed a publicly available deep-sequenced RNA-seq dataset, from murine fibroblasts infected with wild-type and mutant reovirus strains that allowed us to examine changes in editing patterns on a transcriptome-wide scale. To the best of our knowledge, this is the first transcriptome-wide report on host editing changes after reovirus infection. Our results demonstrate that reovirus infection induces unique nuanced editing changes in the host, including introducing sites uniquely edited in infected samples. Genes with edited sites are overrepresented in pathways related to immune regulation, cellular signaling, metabolism, and growth. Moreover, a shift in editing targets has also been observed, where the same genes are edited in infection and control conditions but at different sites, or where the editing rate is increased for some and decreased for other differential targets, supporting the hypothesis of dynamic and condition-specific editing by ADARs.


Subject(s)
Adenosine Deaminase , Fibroblasts , Inosine , RNA Editing , Transcriptome , Animals , Mice , Fibroblasts/virology , Fibroblasts/metabolism , Inosine/metabolism , Inosine/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Adenosine/metabolism , Adenosine/genetics , Reoviridae Infections/virology , Reoviridae Infections/genetics , Host-Pathogen Interactions , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reoviridae/genetics , Reoviridae/physiology
20.
BMC Biol ; 22(1): 132, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38835016

ABSTRACT

BACKGROUND: ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, is thought to play a significant role both in tumor suppression and tumor initiation, which is highly dependent upon context. Previous studies have suggested that ARID1A deficiency may contribute to cancer development. The specific mechanisms of whether ARID1A loss affects tumorigenesis by RNA editing remain unclear. RESULTS: Our findings indicate that the deficiency of ARID1A leads to an increase in RNA editing levels and alterations in RNA editing categories mediated by adenosine deaminases acting on RNA 1 (ADAR1). ADAR1 edits the CDK13 gene at two previously unidentified sites, namely Q113R and K117R. Given the crucial role of CDK13 as a cyclin-dependent kinase, we further observed that ADAR1 deficiency results in changes in the cell cycle. Importantly, the sensitivity of ARID1A-deficient tumor cells to SR-4835, a CDK12/CDK13 inhibitor, suggests a promising therapeutic approach for individuals with ARID1A-mutant tumors. Knockdown of ADAR1 restored the sensitivity of ARID1A deficient cells to SR-4835 treatment. CONCLUSIONS: ARID1A deficiency promotes RNA editing of CDK13 by regulating ADAR1.


Subject(s)
Adenosine Deaminase , Cyclin-Dependent Kinases , DNA-Binding Proteins , RNA Editing , RNA-Binding Proteins , Transcription Factors , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/genetics , Cell Line, Tumor , CDC2 Protein Kinase
SELECTION OF CITATIONS
SEARCH DETAIL