Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
1.
J Biol Chem ; 295(52): 18199-18212, 2020 12 25.
Article in English | MEDLINE | ID: mdl-33100268

ABSTRACT

Post-transcriptional modifications of pre-mRNAs expand the diversity of proteomes in higher eukaryotes. In the brain, these modifications diversify the functional output of many critical neuronal signal molecules. In this study, we identified a brain-specific A-to-I RNA editing that changed glutamine to arginine (Q/R) at exon 20 and an alternative splicing of exon 4 in Tmem63b, which encodes a ubiquitously expressed osmosensitive cation channel. The channel isoforms lacking exon 4 occurred in ∼80% of Tmem63b mRNAs in the brain but were not detected in other tissues, suggesting a brain-specific splicing. We found that the Q/R editing was catalyzed by Adar2 (Adarb1) and required an editing site complementary sequence located in the proximal 5' end of intron 20. Moreover, the Q/R editing was almost exclusively identified in the splicing isoform lacking exon 4, indicating a coupling between the editing and the splicing. Elimination of the Q/R editing in brain-specific Adar2 knockout mice did not affect the splicing efficiency of exon 4. Furthermore, transfection with the splicing isoform containing exon 4 suppressed the Q/R editing in primary cultured cerebellar granule neurons. Thus, our study revealed a coupling between an RNA editing and a distant alternative splicing in the Tmem63b pre-mRNA, in which the splicing plays a dominant role. Finally, physiological analysis showed that the splicing and the editing coordinately regulate Ca2+ permeability and osmosensitivity of channel proteins, which may contribute to their functions in the brain.


Subject(s)
Adenosine Deaminase/physiology , Alternative Splicing , Brain/metabolism , Calcium Channels/genetics , Exons , RNA Editing , RNA Precursors/genetics , RNA-Binding Proteins/physiology , Animals , Calcium Channels/metabolism , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
PLoS Biol ; 16(11): e2006577, 2018 11.
Article in English | MEDLINE | ID: mdl-30496178

ABSTRACT

The interferon (IFN)-mediated innate immune response is the first line of defense against viruses. However, an IFN-stimulated gene, the adenosine deaminase acting on RNA 1 (ADAR1), favors the replication of several viruses. ADAR1 binds double-stranded RNA and converts adenosine to inosine by deamination. This form of editing makes duplex RNA unstable, thereby preventing IFN induction. To better understand how ADAR1 works at the cellular level, we generated cell lines that express exclusively either the IFN-inducible, cytoplasmic isoform ADAR1p150, the constitutively expressed nuclear isoform ADAR1p110, or no isoform. By comparing the transcriptome of these cell lines, we identified more than 150 polymerase II transcripts that are extensively edited, and we attributed most editing events to ADAR1p150. Editing is focused on inverted transposable elements, located mainly within introns and untranslated regions, and predicted to form duplex RNA structures. Editing of these elements occurs also in primary human samples, and there is evidence for cross-species evolutionary conservation of editing patterns in primates and, to a lesser extent, in rodents. Whereas ADAR1p150 rarely edits tightly encapsidated standard measles virus (MeV) genomes, it efficiently edits genomes with inverted repeats accidentally generated by a mutant MeV. We also show that immune activation occurs in fully ADAR1-deficient (ADAR1KO) cells, restricting virus growth, and that complementation of these cells with ADAR1p150 rescues virus growth and suppresses innate immunity activation. Finally, by knocking out either protein kinase R (PKR) or mitochondrial antiviral signaling protein (MAVS)-another protein controlling the response to duplex RNA-in ADAR1KO cells, we show that PKR activation elicits a stronger antiviral response. Thus, ADAR1 prevents innate immunity activation by cellular transcripts that include extensive duplex RNA structures. The trade-off is that viruses take advantage of ADAR1 to elude innate immunity control.


Subject(s)
Adenosine Deaminase/physiology , RNA Viruses/genetics , RNA-Binding Proteins/physiology , Adenosine/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , HeLa Cells , Humans , Immunity, Innate/physiology , Interferons/metabolism , Protein Isoforms , Proviruses/genetics , Proviruses/immunology , RNA Viruses/metabolism , RNA, Double-Stranded/physiology , RNA, Viral/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcriptome/genetics , Virion/genetics
3.
Int J Mol Sci ; 22(21)2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34768866

ABSTRACT

Adenosine deaminase acting on RNA 1 (ADAR1) is an enzyme responsible for double-stranded RNA (dsRNA)-specific adenosine-to-inosine RNA editing, which is estimated to occur at over 100 million sites in humans. ADAR1 is composed of two isoforms transcribed from different promoters: p150 and N-terminal truncated p110. Deletion of ADAR1 p150 in mice activates melanoma differentiation-associated protein 5 (MDA5)-sensing pathway, which recognizes endogenous unedited RNA as non-self. In contrast, we have recently demonstrated that ADAR1 p110-mediated RNA editing does not contribute to this function, implying that a unique Z-DNA/RNA-binding domain α (Zα) in the N terminus of ADAR1 p150 provides specific RNA editing, which is critical for preventing MDA5 activation. In addition, a mutation in the Zα domain is identified in patients with Aicardi-Goutières syndrome (AGS), an inherited encephalopathy characterized by overproduction of type I interferon. Accordingly, we and other groups have recently demonstrated that Adar1 Zα-mutated mice show MDA5-dependent type I interferon responses. Furthermore, one such mutant mouse carrying a W197A point mutation in the Zα domain, which inhibits Z-RNA binding, manifests AGS-like encephalopathy. These findings collectively suggest that Z-RNA binding by ADAR1 p150 is essential for proper RNA editing at certain sites, preventing aberrant MDA5 activation.


Subject(s)
Adenosine Deaminase/metabolism , Adenosine Deaminase/physiology , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Adenosine , Animals , DNA, Z-Form/metabolism , DNA, Z-Form/physiology , Humans , Inosine , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Mice , Protein Isoforms/metabolism , RNA Editing/physiology , RNA, Double-Stranded
4.
Brain ; 142(3): 586-605, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30698736

ABSTRACT

As clinical evidence supports a negative impact of dysfunctional energy metabolism on the disease progression in amyotrophic lateral sclerosis, it is vital to understand how the energy metabolic pathways are altered and whether they can be restored to slow disease progression. Possible approaches include increasing or rerouting catabolism of alternative fuel sources to supplement the glycolytic and mitochondrial pathways such as glycogen, ketone bodies and nucleosides. To analyse the basis of the catabolic defect in amyotrophic lateral sclerosis we used a novel phenotypic metabolic array. We profiled fibroblasts and induced neuronal progenitor-derived human induced astrocytes from C9orf72 amyotrophic lateral sclerosis patients compared to normal controls, measuring the rates of production of reduced nicotinamide adenine dinucleotides from 91 potential energy substrates. This approach shows for the first time that C9orf72 human induced astrocytes and fibroblasts have an adenosine to inosine deamination defect caused by reduction of adenosine deaminase, which is also observed in induced astrocytes from sporadic patients. Patient-derived induced astrocyte lines were more susceptible to adenosine-induced toxicity, which could be mimicked by inhibiting adenosine deaminase in control lines. Furthermore, adenosine deaminase inhibition in control induced astrocytes led to increased motor neuron toxicity in co-cultures, similar to the levels observed with patient derived induced astrocytes. Bypassing metabolically the adenosine deaminase defect by inosine supplementation was beneficial bioenergetically in vitro, increasing glycolytic energy output and leading to an increase in motor neuron survival in co-cultures with induced astrocytes. Inosine supplementation, in combination with modulation of the level of adenosine deaminase may represent a beneficial therapeutic approach to evaluate in patients with amyotrophic lateral sclerosis.


Subject(s)
Adenosine Deaminase/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Motor Neurons/metabolism , Adenosine Deaminase/physiology , Adult , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Astrocytes/metabolism , C9orf72 Protein/metabolism , Cell Death , Cell Survival , Cells, Cultured , Coculture Techniques , Disease Progression , Energy Metabolism/physiology , Female , Fibroblasts/metabolism , Humans , Inosine/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Rats , Rats, Sprague-Dawley , Stem Cells/metabolism
5.
Genet Sel Evol ; 52(1): 35, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32611306

ABSTRACT

Base editing has the potential to improve important economic traits in agriculture and can precisely convert single nucleotides in DNA or RNA sequences into minimal double-strand DNA breaks (DSB). Adenine base editors (ABE) have recently emerged as a base editing tool for the conversion of targeted A:T to G:C, but have not yet been used in sheep. ABEmax is one of the latest versions of ABE, which consists of a catalytically-impaired nuclease and a laboratory-evolved DNA-adenosine deaminase. The Booroola fecundity (FecBB) mutation (g.A746G, p.Q249R) in the bone morphogenetic protein receptor 1B (BMPR1B) gene influences fecundity in many sheep breeds. In this study, by using ABEmax we successfully obtained lambs with defined point mutations that result in an amino acid substitution (p.Gln249Arg). The efficiency of the defined point mutations was 75% in newborn lambs, since six lambs were heterozygous at the FecBB mutation site (g.A746G, p.Q249R), and two lambs were wild-type. We did not detect off-target mutations in the eight edited lambs. Here, we report the validation of the first gene-edited sheep generated by ABE and highlight its potential to improve economically important traits in livestock.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/genetics , Fertility/genetics , Gene Editing/methods , Adenine/metabolism , Adenosine Deaminase/metabolism , Adenosine Deaminase/physiology , Animals , Breeding , Female , Genetic Engineering/methods , Genotype , Heterozygote , Litter Size/genetics , Male , Mutation , Phenotype , Polymorphism, Single Nucleotide , Pregnancy , Sheep/genetics
6.
Bioessays ; 40(2)2018 02.
Article in English | MEDLINE | ID: mdl-29280160

ABSTRACT

RNA editing is a major post-transcriptional mechanism that changes specific nucleotides at the RNA level. The most common RNA editing type in humans is adenosine (A) to inosine (I) editing, which is mediated by ADAR enzymes. RNA editing events can not only change amino acids in proteins, but also affect the functions of non-coding RNAs such as miRNAs. Recent studies have characterized thousands of miRNA RNA editing events across different cancer types. Importantly, individual cases of miRNA editing have been reported to play a role in cancer development. In this review, we summarize the current knowledge of miRNA editing in cancer, and discuss the mechanisms on how miRNA-related editing events modulate the initiation and progression of human cancer. Finally, we discuss the challenges and future directions of studying miRNA editing in cancer.


Subject(s)
MicroRNAs/genetics , Neoplasms/genetics , RNA Editing/genetics , Adenosine/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/physiology , Animals , Humans , Inosine/genetics , MicroRNAs/physiology , RNA-Binding Proteins/physiology
7.
RNA Biol ; 15(7): 863-867, 2018.
Article in English | MEDLINE | ID: mdl-30071181

ABSTRACT

DNA harbors the blueprint for life. However, the instructions stored in the DNA could be altered at the RNA level before they are executed. One of these processes is RNA editing, which was shown to modify RNA sequences in many organisms. The most abundant modification is the deamination of adenosine (A) into inosine (I). In turn, inosine can be identified as a guanosine (G) by the ribosome and other cellular machineries such as reverse transcriptase. In multicellular organisms, enzymes from the ADAR (adenosine deaminase acting on RNA) family mediate RNA editing in mRNA, whereas enzymes from the ADAT family mediate A-to-I editing on tRNAs. In bacteria however, until recently, only one editing site was described, in tRNAArg, but never in mRNA. The tRNA site was shown to be modified by tadA (tRNA specific adenosine deaminase) which is believed to be the ancestral enzyme for the RNA editing family of enzymes. In our recent work, we have shown for the first time, editing on multiple sites in bacterial mRNAs and identified tadA as the enzyme responsible for this editing activity. Focusing on one of the identified targets - the self-killing toxin hokB, we found that editing is physiologically regulated and that it increases protein activity. Here we discuss possible modes of regulation on hokB editing, potential roles of RNA editing in bacteria, possible implications, and future research directions.


Subject(s)
Adenosine Deaminase/physiology , Klebsiella pneumoniae/enzymology , RNA Editing/physiology , RNA, Messenger/metabolism , Yersinia enterocolitica/enzymology , Adenosine/genetics , Bacterial Toxins/metabolism , Deamination/physiology , Drug Resistance, Bacterial/physiology , Inosine/genetics , RNA, Transfer/metabolism , Toxin-Antitoxin Systems/physiology
8.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 43(8): 904-911, 2018 Aug 28.
Article in Zh | MEDLINE | ID: mdl-30197321

ABSTRACT

RNA editing, especially A-to-I RNA editing, is a common post-transcriptional modification in mammals. Adenosine deaminase acting on RNA (ADAR) is a key protein for A-to-I editing, which converts the adenosine group of a double-stranded RNA to creatinine group by deaminating it, resulting in a change of nucleotide sequence. There are 3 types of ADARs (ADAR1, ADAR2, ADAR3) that have been found in recent years. The abnormalities of ADARs are closely related to many human diseases such as viral infections, metabolic diseases, nervous system diseases, and tumors.


Subject(s)
Adenosine Deaminase/physiology , Disease/etiology , RNA Editing/physiology , RNA-Binding Proteins/physiology , Adenosine/metabolism , Base Sequence , Creatinine/metabolism , Deamination , Humans , RNA, Double-Stranded
9.
J Biol Chem ; 291(12): 6158-68, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26817845

ABSTRACT

Adenosine deaminases acting on double-stranded RNA (ADARs) catalyze the deamination of adenosine (A) to produce inosine (I) in double-stranded (ds) RNA structures, a process known as A-to-I RNA editing. dsRNA is an important trigger of innate immune responses, including interferon (IFN) production and action. We examined the role of A-to-I RNA editing by two ADARs, ADAR1 and ADAR2, in the sensing of self-RNA in the absence of pathogen infection, leading to activation of IFN-induced, RNA-mediated responses in mouse embryo fibroblasts. IFN treatment of Adar1(-/-) cells lacking both the p110 constitutive and p150 IFN-inducible ADAR1 proteins induced formation of stress granules, whereas neither wild-type (WT) nor Adar2(-/-) cells displayed a comparable stress granule response following IFN treatment. Phosphorylation of protein synthesis initiation factor eIF2α at serine 51 was increased in IFN-treated Adar1(-/-) cells but not in either WT or Adar2(-/-) cells following IFN treatment. Analysis by deep sequencing of mouse exonic loci containing A-to-I-editing sites revealed that the majority of editing in mouse embryo fibroblasts was carried out by ADAR1. IFN treatment increased editing in both WT and Adar2(-/-) cells but not in either Adar1(-/-) or Adar1(-/-) (p150) cells or Stat1(-/-) or Stat2(-/-) cells. Hyper-edited sites found in predicted duplex structures showed strand bias of editing for some RNAs. These results implicate ADAR1 p150 as the major A-to-I editor in mouse embryo fibroblasts, acting as a feedback suppressor of innate immune responses otherwise triggered by self-RNAs possessing regions of double-stranded character.


Subject(s)
Adenosine Deaminase/physiology , Immunity, Innate , RNA Editing , RNA, Double-Stranded/metabolism , Animals , Cells, Cultured , Cytoplasmic Granules/metabolism , Deamination , Eukaryotic Initiation Factor-2/metabolism , Fibroblasts/metabolism , Immune Tolerance , Interferon-alpha/physiology , Mice, Knockout , Phosphorylation , Protein Processing, Post-Translational , RNA, Double-Stranded/genetics , RNA-Binding Proteins/physiology , Signal Transduction
11.
Am J Pathol ; 185(12): 3224-37, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26453800

ABSTRACT

Adenosine deaminase acting on RNA 1 (ADAR1) is an essential protein for embryonic liver development. ADAR1 loss is embryonically lethal because of severe liver damage. Although ADAR1 is required in adult livers to prevent liver cell death, as demonstrated by liver-specific conditional knockout (Alb-ADAR1(KO)) mice, the mechanism remains elusive. We systematically analyzed Alb-ADAR1(KO) mice for liver damage. Differentiation genes and inflammatory pathways were examined in hepatic tissues from Alb-ADAR1(KO) and littermate controls. Inducible ADAR1 KO mice were used to validate regulatory effects of ADAR1 on inflammatory cytokines. We found that Alb-ADAR1(KO) mice showed dramatic growth retardation and high mortality because of severe structural and functional damage to the liver, which showed overwhelming inflammation, cell death, fibrosis, fatty change, and compensatory regeneration. Simultaneously, Alb-ADAR1(KO) showed altered expression of key differentiation genes and significantly higher levels of hepatic inflammatory cytokines, especially type I interferons, which was also verified by inducible ADAR1 knockdown in primary hepatocyte cultures. We conclude that ADAR1 is an essential molecule for maintaining adult liver homeostasis and, in turn, morphological and functional integrity. It inhibits the production of type I interferons and other inflammatory cytokines. Our findings may provide novel insight in the pathogenesis of liver diseases caused by excessive inflammatory responses, including autoimmune hepatitis.


Subject(s)
Adenosine Deaminase/physiology , Hepatitis/metabolism , Hepatocytes/metabolism , Interferons/biosynthesis , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Blood Proteins/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Cytokines/biosynthesis , Gene Expression Regulation/physiology , Gene Knockout Techniques/methods , Hepatitis/pathology , Hepatitis/physiopathology , Lipids/blood , Mice, Knockout
12.
Adv Exp Med Biol ; 907: 189-213, 2016.
Article in English | MEDLINE | ID: mdl-27256387

ABSTRACT

RNA editing is a cellular process used to expand and diversify the RNA transcripts produced from a generally immutable genome. In animals, the most prevalent type of RNA editing is adenosine (A) to inosine (I) deamination catalyzed by the ADAR family. Throughout development, A-to-I editing levels increase while ADAR expression is constant, suggesting cellular mechanisms to regulate A-to-I editing exist. Furthermore, in several disease states, ADAR expression levels are similar to the normal state, but A-to-I editing levels are altered. Therefore, understanding how these enzymes are regulated in normal tissues and misregulated in disease states is of profound importance. This chapter will both discuss how to identify A-to-I editing sites across the transcriptome and explore the mechanisms that regulate ADAR editing activity, with particular focus on the diverse types of RNA-binding proteins implicated in regulating A-to-I editing in vivo.


Subject(s)
Adenosine Deaminase/physiology , Adenosine/metabolism , Inosine/metabolism , RNA Editing , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/physiology , Anemia, Macrocytic/enzymology , Anemia, Macrocytic/genetics , Animals , Base Pairing , Caenorhabditis elegans Proteins/physiology , Chromosome Deletion , Chromosomes, Human, Pair 5/enzymology , Chromosomes, Human, Pair 5/genetics , Drosophila Proteins/physiology , Humans , Mice , Nervous System Diseases/enzymology , Nervous System Diseases/genetics , Nucleic Acid Conformation , RNA Splicing , Spliceosomes/physiology , Transcriptome
14.
Nucleic Acids Res ; 41(5): 3362-72, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23355609

ABSTRACT

In all organisms, transfer RNAs (tRNAs) undergo extensive post-transcriptional modifications. Although base modifications in the anticodon are known to alter decoding specificity or improve decoding accuracy, much less is known about the functional relevance of modifications in other positions of tRNAs. Here, we report the identification of an A-to-I tRNA editing enzyme that modifies the tRNA-Ala(AGC) in the model plant Arabidopsis thaliana. The enzyme is homologous to Tad1p, a yeast tRNA-specific adenosine deaminase, and it selectively deaminates the adenosine in the position 3'-adjacent to the anticodon (A37) to inosine. We show that the AtTAD1 protein is exclusively localized in the nucleus. The tad1 loss-of-function mutants isolated in Arabidopsis show normal accumulation of the tRNA-Ala(AGC), suggesting that the loss of the I37 modification does not affect tRNA stability. The tad1 knockout mutants display no discernible phenotype under standard growth conditions, but produce less biomass under environmental stress conditions. Our results provide the first evidence in support of a physiological relevance of the A37-to-I modification in eukaryotes.


Subject(s)
Adenosine Deaminase/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Cold-Shock Response , RNA Editing , RNA, Transfer, Ala/metabolism , Adenosine/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/physiology , Arabidopsis/cytology , Arabidopsis/physiology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/physiology , Base Sequence , Cell Nucleus/metabolism , Inosine/metabolism , Mitochondria , Mutagenesis, Insertional , Nucleic Acid Conformation , Phenotype , Plant Leaves/cytology , Plant Leaves/enzymology , Plant Leaves/physiology , Plant Roots/cytology , Plant Roots/enzymology , Plant Roots/physiology , Protein Transport , RNA Stability
15.
Mediators Inflamm ; 2015: 349215, 2015.
Article in English | MEDLINE | ID: mdl-26549939

ABSTRACT

The functions of genes involved in idiopathic portal hypertension (IPH) remain unidentified. The present study was undertaken to identify the functions of genes expressed in blood samples from patients with IPH through comprehensive analysis of gene expression using DNA microarrays. The data were compared with data from healthy individuals to explore the functions of genes showing increased or decreased expression in patients with IPH. In cluster analysis, no dominant probe group was shown to differ between patients with IPH and healthy controls. In functional annotation analysis using the Database for Annotation Visualization and Integrated Discovery tool, clusters showing dysfunction in patients with IPH involved gene terms related to the immune system. Analysis using network-based pathways revealed decreased expression of adenosine deaminase, ectonucleoside triphosphate diphosphohydrolase 4, ATP-binding cassette, subfamily C, member 1, transforming growth factor-ß, and prostaglandin E receptor 2; increased expression of cytochrome P450, family 4, subfamily F, polypeptide 3, and glutathione peroxidase 3; and abnormalities in the immune system, nucleic acid metabolism, arachidonic acid/leukotriene pathways, and biological processes. These results suggested that IPH involved compromised function of immunocompetent cells and that such dysfunction may be associated with abnormalities in nucleic acid metabolism and arachidonic acid/leukotriene-related synthesis/metabolism.


Subject(s)
Gene Regulatory Networks , Hypertension, Portal/genetics , Liver Cirrhosis/genetics , Oligonucleotide Array Sequence Analysis/methods , Pancytopenia/genetics , Splenomegaly/genetics , Adenosine Deaminase/physiology , Cluster Analysis , Humans , Receptors, Atrial Natriuretic Factor/physiology , Idiopathic Noncirrhotic Portal Hypertension
16.
Proc Natl Acad Sci U S A ; 109(48): E3295-304, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23129636

ABSTRACT

Adenosine deaminases that act on RNA (ADARs) deaminate adenosines in dsRNA to produce inosines. ADARs are essential in mammals and are particularly important in the nervous system. Altered levels of adenosine-to-inosine (A-to-I) editing are observed in several diseases. The extent to which an adenosine is edited depends on sequence context. Human ADAR2 (hADAR2) has 5' and 3' neighbor preferences, but which amino acids mediate these preferences, and by what mechanism, is unknown. We performed a screen in yeast to identify mutations in the hADAR2 catalytic domain that allow editing of an adenosine within a disfavored triplet. Binding affinity, catalytic rate, base flipping, and preferences were monitored to understand the effects of the mutations on ADAR reactivity. Our data provide information on the amino acids that affect preferences and point to a conserved loop as being of key importance. Unexpectedly, our data suggest that hADAR2's preferences derive from differential base flipping rather than from direct recognition of neighboring bases. Our studies set the stage for understanding the basis of altered editing levels in disease and for developing therapeutic reagents.


Subject(s)
Adenosine Deaminase/physiology , RNA Editing , Adenosine Deaminase/chemistry , Amino Acid Sequence , Animals , Binding Sites , Catalytic Domain , Models, Molecular , Molecular Sequence Data , Mutation , RNA-Binding Proteins , Sequence Homology, Amino Acid
17.
J Biol Chem ; 288(2): 826-36, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23209284

ABSTRACT

Somatic mutations accumulate in senescent cells. Bcl6, which functions as a transcriptional repressor, has been identified as a potent inhibitor of cell senescence, but a role of Bcl6 in the accumulation of somatic mutations has remained unclear. Ig class-switch recombination simultaneously induces somatic mutations in an IgM class-switch (Ig-Sµ) region of IgG B cells. Surprisingly, mutations were detected in the Ig-Sµ region of Bcl6-deficient IgM B cells without class-switch recombination, and these mutations were mainly generated by conversion of adenosine to guanosine, suggesting a novel DNA mutator in the B cells. The ADAR1 (adenosine deaminase acting on RNA1) gene was overexpressed in Bcl6-deficient cells, and its promoter analysis revealed that ADAR1 is a molecular target of Bcl6. Exogenous ADAR1 induced adenosine-targeted DNA mutations in IgM B cells from ADAR1-transgenic mice and in wild-type mouse embryonic fibroblasts (MEFs). These mutations accumulated in senescent MEFs accompanied with endogenous ADAR1 expression, and the frequency in senescent Bcl6-deficient MEFs was higher than senescent wild-type MEFs. Thus, Bcl6 protects senescent cells from accumulation of adenosine-targeted DNA mutations induced by ADAR1.


Subject(s)
Adenosine Deaminase/physiology , Adenosine/metabolism , DNA-Binding Proteins/physiology , DNA/genetics , Mutation , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6 , RNA-Binding Proteins
18.
Curr Top Microbiol Immunol ; 353: 221-36, 2012.
Article in English | MEDLINE | ID: mdl-21761288

ABSTRACT

Drosophila melanogaster has a single Adar gene encoding a protein related to mammalian ADAR2 that edits transcripts encoding glutamate receptor subunits. We describe the structure of the Drosophila Adar locus and use ModENCODE information to supplement published data on Adar gene transcription, and splicing. We discuss the roles of ADAR in Drosophila in terms of the two main types of RNA molecules edited and roles of ADARs as RNA-binding proteins. Site-specific RNA editing events in transcripts encoding ion channel subunits were initially found serendipitously and subsequent directed searches for editing sites and transcriptome sequencing have now led to 972 edited sites being identified in 597 transcripts. Four percent of D. melanogaster transcripts are site-specifically edited and these encode a wide range of largely membrane-associated proteins expressed particularly in CNS. Electrophysiological studies on the effects of specific RNA editing events on ion channel subunits do not suggest that loss of RNA editing events in ion channels consistently produce a particular outcome such as making Adar mutant neurons more excitable. This possibility would have been consistent with neurodegeneration seen in Adar mutant fly brains. A further set of ADAR targets are dsRNA intermediates in siRNA generation, derived from transposons and from structured RNA loci. Transcripts with convergent overlapping 3' ends are also edited and the first discovered instance of RNA editing in Drosophila, in the Rnp4F transcript, is an example. There is no evidence yet to show that Adar antagonizes RNA interference in Drosophila. Evidence has been obtained that catalytically inactive ADAR proteins exert effects on microRNA generation and RNA interference. Whether all effects of inactive ADARs are due to RNA-binding or to even further roles of these proteins remains to be determined.


Subject(s)
Adenosine Deaminase/physiology , Drosophila Proteins/physiology , Adenosine Deaminase/genetics , Animals , Drosophila , Drosophila Proteins/genetics , RNA Editing , RNA Interference , Transcription, Genetic
19.
Curr Top Microbiol Immunol ; 353: 91-109, 2012.
Article in English | MEDLINE | ID: mdl-21761289

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression by preventing the translation of specific messenger RNAs. Adenosine deaminases acting on RNAs (ADARs) catalyze adenosine-to-inosine (A-to-I) RNA editing, the conversion of adenosines into inosines, in double-stranded RNAs. Because inosine preferentially base pairs with cytidine, this conversion is equivalent to an adenosine to guanosine change. Over the past seven years, an increasing number of edited adenosines have been identified in miRNAs. Editing of miRNAs affects their biogenesis, causes their degradation or alters the set of messenger RNAs that they regulate. Recently, ADARs have been shown to also affect the miRNA phenomenon by sequestering miRNAs or by editing the messenger RNAs they regulate. This article reviews the recent attempts to identify miRNA editing sites and elucidate the effects of ADARs on miRNA expression and function.


Subject(s)
Adenosine Deaminase/physiology , MicroRNAs/physiology , Animals , Gene Expression Regulation , Humans , MicroRNAs/analysis , RNA Editing , RNA-Binding Proteins , Ribonuclease III/physiology
20.
Curr Top Microbiol Immunol ; 353: 197-220, 2012.
Article in English | MEDLINE | ID: mdl-21725896

ABSTRACT

RNA editing by deamination of adenosine to inosine (A-to-I editing) is a physiologically important posttranscriptional mechanism that can regulate expression of genes by modifying their transcripts. A-to-I editing is mediated by adenosine deaminases acting on RNA (ADAR) that can catalytically exchange adenosines to inosines, with varying efficiency, depending on the structure of the RNA substrates. Significant progress in understanding the biological function of mammalian ADARs has been made in the past decade by the creation and analysis of gene-targeted mice with disrupted or modified ADAR alleles. These studies have revealed important roles of ADARs in neuronal and hematopoietic tissue during embryonic and postnatal stages of mouse development.


Subject(s)
Adenosine Deaminase/physiology , Embryonic Development , Alu Elements , Amyotrophic Lateral Sclerosis/etiology , Animals , Hematopoiesis , Mice , MicroRNAs/genetics , RNA Editing , RNA-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL