Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.114
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Int J Mol Sci ; 25(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891830

ABSTRACT

The dynamic process of Drosophila spermatogenesis involves asymmetric division, mitosis, and meiosis, which ultimately results in the production of mature spermatozoa. Disorders of spermatogenesis can lead to infertility in males. ADAR (adenosine deaminase acting on RNA) mutations in Drosophila cause male infertility, yet the causative factors remain unclear. In this study, immunofluorescence staining was employed to visualize endogenous ADAR proteins and assess protein levels via fluorescence-intensity analysis. In addition, the early differentiation disorders and homeostatic alterations during early spermatogenesis in the testes were examined through quantification of transit-amplifying region length, counting the number of GSCs (germline stem cells), and fertility experiments. Our findings suggest that deletion of ADAR causes testicular tip transit-amplifying cells to accumulate and become infertile in older male Drosophila. By overexpressing ADAR in early germline cells, male infertility can be partially rescued. Transcriptome analysis showed that ADAR maintained early spermatogenesis homeostasis through the bone-morphogenetic-protein (BMP) signaling pathway. Taken together, these findings have the potential to help explore the role of ADAR in early spermatogenesis.


Subject(s)
Adenosine Deaminase , Bone Morphogenetic Proteins , Drosophila Proteins , Drosophila melanogaster , Signal Transduction , Spermatogenesis , Animals , Male , Spermatogenesis/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Infertility, Male/genetics , Infertility, Male/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Testis/metabolism
7.
Int J Mol Sci ; 25(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892173

ABSTRACT

A-to-I RNA editing, catalyzed by the ADAR protein family, significantly contributes to the diversity and adaptability of mammalian RNA signatures, aligning with developmental and physiological needs. Yet, the functions of many editing sites are still to be defined. The Unc80 gene stands out in this context due to its brain-specific expression and the evolutionary conservation of its codon-altering editing event. The precise biological functions of Unc80 and its editing, however, are still largely undefined. In this study, we first demonstrated that Unc80 editing occurs in an ADAR2-dependent manner and is exclusive to the brain. By employing the CRISPR/Cas9 system to generate Unc80 knock-in mouse models that replicate the natural editing variations, our findings revealed that mice with the "gain-of-editing" variant (Unc80G/G) exhibit heightened basal neuronal activity in critical olfactory regions, compared to the "loss-of-editing" (Unc80S/S) counterparts. Moreover, an increase in glutamate levels was observed in the olfactory bulbs of Unc80G/G mice, indicating altered neurotransmitter dynamics. Behavioral analysis of odor detection revealed distinctive responses to novel odors-both Unc80 deficient (Unc80+/-) and Unc80S/S mice demonstrated prolonged exploration times and heightened dishabituation responses. Further elucidating the olfactory connection of Unc80 editing, transcriptomic analysis of the olfactory bulb identified significant alterations in gene expression that corroborate the behavioral and physiological findings. Collectively, our research advances the understanding of Unc80's neurophysiological functions and the impact of its editing on the olfactory sensory system, shedding light on the intricate molecular underpinnings of olfactory perception and neuronal activity.


Subject(s)
Adenosine Deaminase , Olfactory Perception , RNA Editing , Animals , Mice , Olfactory Perception/physiology , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Olfactory Bulb/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Neurons/metabolism , CRISPR-Cas Systems , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
8.
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
9.
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
10.
Oncoimmunology ; 13(1): 2371051, 2024.
Article in English | MEDLINE | ID: mdl-38915783

ABSTRACT

Improving cancer immunotherapy efficacy hinges on identifying key T-cell populations critical for tumor control and response to Immune Checkpoint Blockade (ICB). We have recently reported that while the co-expression of PD-1 and CD28 is associated with impaired functionality in peripheral blood, it significantly enhances T-cell fitness in the tumor site of non-small cell lung cancer (NSCLC) patients. To uncover the underlying mechanisms, we explored the role of CD26, a key player in T-cell activation through its interaction with adenosine deaminase (ADA), a crucial intra/extracellular enzyme able to neutralize local adenosine (ADO). We found that an autocrine ADA/CD26 axis enhances CD8+PD-1+CD28+ T-cell function, particularly within an immunosuppressive environment marked by CD39 expression. Then, we interrogated the TCGA and OAK datasets to gain insight into the prognostic/predictive potential of our findings. We identified a signature predicting overall survival (OS) in LUAD patients and response to atezolizumab in advanced LUAD cases. These findings suggest promising avenues for therapeutic intervention targeting the ADA/CD26 axis.


Subject(s)
Adenosine Deaminase , CD28 Antigens , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung , Dipeptidyl Peptidase 4 , Immune Checkpoint Inhibitors , Lung Neoplasms , Programmed Cell Death 1 Receptor , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD28 Antigens/metabolism , Prognosis , Programmed Cell Death 1 Receptor/metabolism , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl Peptidase 4/genetics , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/administration & dosage , Female , Male , Apyrase/metabolism
11.
Biochem Biophys Res Commun ; 726: 150289, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-38917633

ABSTRACT

Among the various RNA modifications, adenosine-to-inosine RNA editing, catalyzed by adenosine deaminase acting on RNA (ADAR) family, ADAR1 and ADAR2, is the most common nucleotide conversion in mammalian cells. The pathological relevance of ADAR expression has been highlighted in recent human genetic studies. Low expression of the ADAR2 gene is correlated with a poor prognosis in breast cancer patients, but the underlying mechanism remains enigmatic. In this study, we constructed Adar2-knockdown (Adar2-KD) murine breast cancer 4T1 cells and observed their reduced susceptibility to chemotherapeutic drug doxorubicin. Downregulation of ADAR2 induced the expression of P-glycoprotein (P-gp), leading to a reduction in the intracellular accumulation of doxorubicin. The upregulation of P-gp occurred at the post-transcriptional level due to the decreased miR-195a-3p function. The search for the underlying cause of the induction of P-gp expression in Adar2-KD 4T1 cells led to the identification of circular RNA (circRNA) circHif1a as a sponge for miR-195a-3p. The enhanced expression of circHif1a inhibited miR-195a-3p function, resulting in the upregulation of P-gp expression. These results suggest that ADAR2 acts as a suppressor of circHif1a biogenesis and then allows miR-195a-3p to interfere with P-gp translation. Our findings may help to improve drug efficacy by clarifying the mechanism of chemoresistance in breast cancer.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Adenosine Deaminase , Doxorubicin , Gene Expression Regulation, Neoplastic , MicroRNAs , RNA Editing , RNA, Circular , Animals , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Female , RNA, Circular/genetics , RNA, Circular/metabolism , Doxorubicin/pharmacology , Cell Line, Tumor , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Antibiotics, Antineoplastic/pharmacology
12.
Nat Commun ; 15(1): 5366, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926387

ABSTRACT

Adenosine-to-inosine (A-to-I) editing is a prevalent post-transcriptional RNA modification within the brain. Yet, most research has relied on postmortem samples, assuming it is an accurate representation of RNA biology in the living brain. We challenge this assumption by comparing A-to-I editing between postmortem and living prefrontal cortical tissues. Major differences were found, with over 70,000 A-to-I sites showing higher editing levels in postmortem tissues. Increased A-to-I editing in postmortem tissues is linked to higher ADAR and ADARB1 expression, is more pronounced in non-neuronal cells, and indicative of postmortem activation of inflammation and hypoxia. Higher A-to-I editing in living tissues marks sites that are evolutionarily preserved, synaptic, developmentally timed, and disrupted in neurological conditions. Common genetic variants were also found to differentially affect A-to-I editing levels in living versus postmortem tissues. Collectively, these discoveries offer more nuanced and accurate insights into the regulatory mechanisms of RNA editing in the human brain.


Subject(s)
Adenosine Deaminase , Adenosine , Autopsy , Brain , Inosine , RNA Editing , RNA-Binding Proteins , Humans , Adenosine/metabolism , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Brain/metabolism , Inosine/metabolism , Inosine/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Prefrontal Cortex/metabolism , Postmortem Changes , Male
13.
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
14.
Nucleic Acids Res ; 52(12): 6733-6747, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38828787

ABSTRACT

Adenosine Deaminases Acting on RNA (ADARs) are enzymes that catalyze the conversion of adenosine to inosine in RNA duplexes. These enzymes can be harnessed to correct disease-causing G-to-A mutations in the transcriptome because inosine is translated as guanosine. Guide RNAs (gRNAs) can be used to direct the ADAR reaction to specific sites. Chemical modification of ADAR guide strands is required to facilitate delivery, increase metabolic stability, and increase the efficiency and selectivity of the editing reaction. Here, we show the ADAR reaction is highly sensitive to ribose modifications (e.g. 4'-C-methylation and Locked Nucleic Acid (LNA) substitution) at specific positions within the guide strand. Our studies were enabled by the synthesis of RNA containing a new, ribose-modified nucleoside analog (4'-C-methyladenosine). Importantly, the ADAR reaction is potently inhibited by LNA or 4'-C-methylation at different positions in the ADAR guide. While LNA at guide strand positions -1 and -2 block the ADAR reaction, 4'-C-methylation only inhibits at the -2 position. These effects are rationalized using high-resolution structures of ADAR-RNA complexes. This work sheds additional light on the mechanism of ADAR deamination and aids in the design of highly selective ADAR guide strands for therapeutic editing using chemically modified RNA.


Subject(s)
Adenosine Deaminase , RNA Editing , Ribose , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/chemistry , Ribose/chemistry , Ribose/metabolism , Humans , Oligonucleotides/chemistry , Oligonucleotides/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , Methylation , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/chemistry , Nucleosides/chemistry , Nucleosides/metabolism , RNA/metabolism , RNA/chemistry , Inosine/metabolism , Inosine/chemistry
15.
Commun Biol ; 7(1): 615, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777862

ABSTRACT

Deficiency of adenosine deaminase 2 (DADA2) is an inborn error of immunity caused by loss-of-function mutations in the adenosine deaminase 2 (ADA2) gene. Clinical manifestations of DADA2 include vasculopathy and immuno-hematological abnormalities, culminating in bone marrow failure. A major gap exists in our knowledge of the regulatory functions of ADA2 during inflammation and hematopoiesis, mainly due to the absence of an ADA2 orthologue in rodents. Exploring these mechanisms is essential for understanding disease pathology and developing new treatments. Zebrafish possess two ADA2 orthologues, cecr1a and cecr1b, with the latter showing functional conservation with human ADA2. We establish a cecr1b-loss-of-function zebrafish model that recapitulates the immuno-hematological and vascular manifestations observed in humans. Loss of Cecr1b disrupts hematopoietic stem cell specification, resulting in defective hematopoiesis. This defect is caused by induced inflammation in the vascular endothelium. Blocking inflammation, pharmacological modulation of the A2r pathway, or the administration of the recombinant human ADA2 corrects these defects, providing insights into the mechanistic link between ADA2 deficiency, inflammation and immuno-hematological abnormalities. Our findings open up potential therapeutic avenues for DADA2 patients.


Subject(s)
Adenosine Deaminase , Hematopoiesis , Hematopoietic Stem Cells , Inflammation , Zebrafish , Animals , Zebrafish/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Adenosine Deaminase/deficiency , Hematopoietic Stem Cells/metabolism , Inflammation/genetics , Inflammation/metabolism , Hematopoiesis/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Humans , Signal Transduction , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism
16.
Commun Biol ; 7(1): 594, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760406

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a chronic disease caused by hepatic steatosis. Adenosine deaminases acting on RNA (ADARs) catalyze adenosine to inosine RNA editing. However, the functional role of ADAR2 in NAFLD is unclear. ADAR2+/+/GluR-BR/R mice (wild type, WT) and ADAR2-/-/GluR-BR/R mice (ADAR2 KO) mice are fed with standard chow or high-fat diet (HFD) for 12 weeks. ADAR2 KO mice exhibit protection against HFD-induced glucose intolerance, insulin resistance, and dyslipidemia. Moreover, ADAR2 KO mice display reduced liver lipid droplets in concert with decreased hepatic TG content, improved hepatic insulin signaling, better pyruvate tolerance, and increased glycogen synthesis. Mechanistically, ADAR2 KO effectively mitigates excessive lipid production via AMPK/Sirt1 pathway. ADAR2 KO inhibits hepatic gluconeogenesis via the AMPK/CREB pathway and promotes glycogen synthesis by activating the AMPK/GSK3ß pathway. These results provide evidence that ADAR2 KO protects against NAFLD progression through the activation of AMPK signaling pathways.


Subject(s)
Adenosine Deaminase , Diet, High-Fat , Mice, Knockout , Non-alcoholic Fatty Liver Disease , RNA-Binding Proteins , Signal Transduction , Animals , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/deficiency , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/etiology , Diet, High-Fat/adverse effects , Male , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Insulin Resistance , Mice, Obese , Obesity/metabolism , Obesity/genetics , Mice, Inbred C57BL , Liver/metabolism
17.
Neoplasma ; 71(2): 180-192, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38766853

ABSTRACT

It has been demonstrated that calreticulin (CALR) is expressed abnormally in various tumors and is involved in the occurrence and development of tumors. In this study, CALR and EIF2AK2 expression was measured in the clinical specimens of 39 patients with melanoma. Then, we constructed knockdown and overexpression cell models of CALR and EIF2AK2 and used wound healing and Transwell assays to observe cell migration and invasion. Apoptosis, EDU, and ROS assays were used to measure cell apoptosis and proliferation, as well as ROS levels. The effect of CALR on endoplasmic reticulum stress was detected using endoplasmic reticulum fluorescent probes. Western blotting was used to detect protein levels of CALR, EIF2AK2, ADAR1, and MMP14. The results indicated that CALR and EIF2AK2 expression levels were significantly higher in human melanoma tissues than in adjacent non-tumor tissue. In addition, we found a correlation between CALR and the expression of EIF2AK2 and MMP14, and the experimental results indicated that overexpression of CALR significantly upregulated the expression of EIF2AK2, MMP14, and ADAR1, while knockdown of CALR inhibited their expression. Notably, the knockdown of EIF2AK2 in the CALR overexpression group blocked the upregulation of MMP14 and ADAR1 expression by CALR, and the knockdown of both CALR and EIF2AK2 significantly inhibited MMP14 and ADAR1 expression. In conclusion, CALR and EIF2AK2 play a promoting role in melanoma progression, and knockdown of CALR and EIF2AK2 may be an effective anti-tumor target, and its mechanism may be through MMP14, ADAR1 signaling.


Subject(s)
Adenosine Deaminase , Calreticulin , Cell Proliferation , Matrix Metalloproteinase 14 , Melanoma , RNA-Binding Proteins , Signal Transduction , eIF-2 Kinase , Humans , Adenosine Deaminase/metabolism , Adenosine Deaminase/genetics , Melanoma/pathology , Melanoma/metabolism , Melanoma/genetics , eIF-2 Kinase/metabolism , eIF-2 Kinase/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Calreticulin/genetics , Calreticulin/metabolism , Cell Line, Tumor , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 14/genetics , Cell Movement , Apoptosis , Endoplasmic Reticulum Stress , Female , Disease Progression , Male , Gene Expression Regulation, Neoplastic , Middle Aged
18.
BMC Pulm Med ; 24(1): 241, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750432

ABSTRACT

BACKGROUND: Pleural fluid is one of the common complications of thoracic diseases, and tuberculous pleural effusion (TPE) is the most common cause of pleural effusion in TB-endemic areas and the most common type of exudative pleural effusion in China. In clinical practice, distinguishing TPE from pleural effusion caused by other reasons remains a relatively challenging issue. The objective of present study was to explore the clinical significance of the pleural fluid lactate dehydrogenase/adenosine deaminase ratio (pfLDH/pfADA) in the diagnosis of TPE. METHODS: The clinical data of 618 patients with pleural effusion were retrospectively collected, and the patients were divided into 3 groups: the TPE group (412 patients), the parapneumonic pleural effusion (PPE) group (106 patients), and the malignant pleural effusion (MPE) group (100 patients). The differences in the ratios of pleural effusion-related and serology-related indicators were compared among the three groups, and receiver operating characteristic curves were drawn to analyze the sensitivity and specificity of the parameter ratios of different indicators for the diagnosis of TPE. RESULTS: The median serum ADA level was higher in the TPE group (13 U/L) than in the PPE group (10 U/L, P < 0.01) and MPE group (10 U/L, P < 0.001). The median pfADA level in the TPE group was 41 (32, 52) U/L; it was lowest in the MPE group at 9 (7, 12) U/L and highest in the PPE group at 43 (23, 145) U/L. The pfLDH level in the PPE group was 2542 (1109, 6219) U/L, which was significantly higher than that in the TPE group 449 (293, 664) U/L. In the differential diagnosis between TPE and non-TPE, the AUC of pfLDH/pfADA for diagnosing TPE was the highest at 0.946 (0.925, 0.966), with an optimal cutoff value of 23.20, sensitivity of 93.9%, specificity of 87.0%, and Youden index of 0.809. In the differential diagnosis of TPE and PPE, the AUC of pfLDH/pfADA was the highest at 0.964 (0.939, 0.989), with an optimal cutoff value of 24.32, sensitivity of 94.6%, and specificity of 94.4%; this indicated significantly better diagnostic efficacy than that of the single index of pfLDH. In the differential diagnosis between TPE and MPE, the AUC of pfLDH/pfADA was 0.926 (0.896, 0.956), with a sensitivity of 93.4% and specificity of 80.0%; this was not significantly different from the diagnostic efficacy of pfADA. CONCLUSIONS: Compared with single biomarkers, pfLDH/pfADA has higher diagnostic value for TPE and can identify patients with TPE early, easily, and economically.


Subject(s)
Adenosine Deaminase , L-Lactate Dehydrogenase , Pleural Effusion , ROC Curve , Sensitivity and Specificity , Tuberculosis, Pleural , Humans , Adenosine Deaminase/analysis , Adenosine Deaminase/blood , Adenosine Deaminase/metabolism , Male , Female , Retrospective Studies , Middle Aged , Pleural Effusion/diagnosis , L-Lactate Dehydrogenase/analysis , Tuberculosis, Pleural/diagnosis , Adult , Aged , China , Diagnosis, Differential , Pleural Effusion, Malignant/diagnosis , Biomarkers/analysis , Biomarkers/blood , Clinical Relevance
19.
Fish Shellfish Immunol ; 150: 109620, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38740229

ABSTRACT

Adenosine deaminases acting on RNA 1 (ADAR1) is a dsRNA adenosine (A)-to-inosine (I) editing enzyme that regulates the innate immune response against virus invasion. In the present study, a novel CgADAR1 was identified from the oyster Crassostrea gigas. The open reading frame (ORF) of CgADAR1 was of 3444 bp encoding a peptide of 1147 amino acid residues with two Zα domains, one dsRNA binding motif (DSRM) and one RNA adenosine deaminase domain (ADEAMc). The mRNA transcripts of CgADAR1 were detected in all the examined tissues, with higher expression levels in mantle and gill, which were 7.11-fold and 4.90-fold (p < 0.05) of that in labial palp, respectively. The mRNA transcripts of CgADAR1 in haemocytes were significantly induced at 24 h and 36 h after Poly (A: U) stimulation, which were 6.03-fold (p < 0.01) and 1.37-fold (p < 0.001) of that in control group, respectively. At 48 h after Poly (A:U) stimulation, the mRNA expression of CgRIG-Ⅰ, CgIRF8 and CgIFNLP significantly increased, which were 4.36-fold (p < 0.001), 1.82-fold (p < 0.05) and 1.92-fold (p < 0.05) of that in control group. After CgADAR1 expression was inhibited by RNA interference (RNAi), the mRNA expression levels of CgMDA5, CgRIG-Ⅰ, CgTBK1, CgIRF8 and CgIFNLP were significantly increased, which were 11.88-fold, 11.51-fold, 2.22-fold, 2.85-fold and 2.52-fold of that in control group (p < 0.001), and the phosphorylation level of CgTBK1 was also significantly increased. These results suggested that CgADAR1 played a regulation role in the early stages of viral infection by inhibiting the synthesis of interferon-like protein.


Subject(s)
Crassostrea , Gene Expression Regulation , Immunity, Innate , Interferons , Animals , Crassostrea/immunology , Crassostrea/genetics , Immunity, Innate/genetics , Gene Expression Regulation/immunology , Interferons/genetics , Interferons/immunology , Amino Acid Sequence , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Phylogeny , Gene Expression Profiling , Sequence Alignment , Base Sequence
20.
Artif Intell Med ; 153: 102886, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749310

ABSTRACT

Tuberculous pleural effusion poses a significant threat to human health due to its potential for severe disease and mortality. Without timely treatment, it may lead to fatal consequences. Therefore, early identification and prompt treatment are crucial for preventing problems such as chronic lung disease, respiratory failure, and death. This study proposes an enhanced differential evolution algorithm based on colony predation and dispersed foraging strategies. A series of experiments conducted on the IEEE CEC 2017 competition dataset validated the global optimization capability of the method. Additionally, a binary version of the algorithm is introduced to assess the algorithm's ability to address feature selection problems. Comprehensive comparisons of the effectiveness of the proposed algorithm with 8 similar algorithms were conducted using public datasets with feature sizes ranging from 10 to 10,000. Experimental results demonstrate that the proposed method is an effective feature selection approach. Furthermore, a predictive model for tuberculous pleural effusion is established by integrating the proposed algorithm with support vector machines. The performance of the proposed model is validated using clinical records collected from 140 tuberculous pleural effusion patients, totaling 10,780 instances. Experimental results indicate that the proposed model can identify key correlated indicators such as pleural effusion adenosine deaminase, temperature, white blood cell count, and pleural effusion color, aiding in the clinical feature analysis of tuberculous pleural effusion and providing early warning for its treatment and prediction.


Subject(s)
Algorithms , Pleural Effusion , Support Vector Machine , Tuberculosis, Pleural , Humans , Pleural Effusion/microbiology , Tuberculosis, Pleural/diagnosis , Adenosine Deaminase/metabolism , Leukocyte Count
SELECTION OF CITATIONS
SEARCH DETAIL
...