Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 5274, 2022 09 07.
Article in English | MEDLINE | ID: mdl-36071059

ABSTRACT

Understanding how neural circuits underlie behaviour is challenging even in the connectome era because it requires a combination of anatomical and functional analyses. This is exemplified in the circuit underlying the light avoidance behaviour displayed by Drosophila melanogaster larvae. While this behaviour is robust and the nervous system relatively simple, the circuit is only partially delineated with some contradictions among studies. Here, we devise trans-Tango MkII, an offshoot of the transsynaptic circuit tracing tool trans-Tango, and implement it in anatomical tracing together with functional analysis. We use neuronal inhibition to test necessity of particular neuronal types in light avoidance and selective neuronal activation to examine sufficiency in rescuing light avoidance deficiencies exhibited by photoreceptor mutants. Our studies reveal a four-order circuit for light avoidance connecting the light-detecting photoreceptors with a pair of neuroendocrine cells via two types of clock neurons. This approach can be readily expanded to studying other circuits.


Subject(s)
Connectome , Drosophila , Animals , Drosophila/physiology , Drosophila melanogaster/physiology , Larva , Neural Pathways/physiology
2.
BMC Genomics ; 18(1): 522, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28693467

ABSTRACT

BACKGROUND: Repetitive elements are now known to have relevant cellular functions, including self-complementary sequences that form double stranded (ds) RNA. There are numerous pathways that determine the fate of endogenous dsRNA, and misregulation of endogenous dsRNA is a driver of autoimmune disease, particularly in the brain. Unfortunately, the alignment of high-throughput, short-read sequences to repeat elements poses a dilemma: Such sequences may align equally well to multiple genomic locations. In order to differentiate repeat elements, current alignment methods depend on sequence variation in the reference genome. Reads are discarded when no such variations are present. However, RNA hyper-editing, a possible fate for dsRNA, introduces enough variation to distinguish between repeats that are otherwise identical. RESULTS: To take advantage of this variation, we developed a new algorithm, RepProfile, that simultaneously aligns reads and predicts novel variations. RepProfile accurately aligns hyper-edited reads that other methods discard. In particular we predict hyper-editing of Drosophila melanogaster repeat elements in vivo at levels previously described only in vitro, and provide validation by Sanger sequencing sixty-two individual cloned sequences. We find that hyper-editing is concentrated in genes involved in cell-cell communication at the synapse, including some that are associated with neurodegeneration. We also find that hyper-editing tends to occur in short runs. CONCLUSIONS: Previous studies of RNA hyper-editing discarded ambiguously aligned reads, ignoring hyper-editing in long, perfect dsRNA - the perfect substrate for hyper-editing. We provide a method that simulation and Sanger validation show accurately predicts such RNA editing, yielding a superior picture of hyper-editing.


Subject(s)
Drosophila melanogaster/genetics , RNA Editing , Sequence Alignment , Algorithms , Animals , Gene Rearrangement , Polymorphism, Single Nucleotide , Repetitive Sequences, Nucleic Acid/genetics
3.
Front Genet ; 7: 100, 2016.
Article in English | MEDLINE | ID: mdl-27458478

ABSTRACT

Transcription of genetic information from archival DNA into RNA molecule working copies is vital for proper cellular function and is highly accurate. In turn, RNAs serve structural, enzymatic, and regulatory roles, as well as being informational templates for the ribosomal translation of proteins. Following RNA synthesis, maturing of RNA molecules occurs through various RNA processing events. One component of the collection of processes involving RNA species, broadly defined as RNA metabolism, is the RNA-editing pathway and is found in all animals. Acting specifically on RNA substrates with double-stranded character, RNA editing has been shown to regulate a plethora of genomic outputs, including gene recoding, RNA splicing, biogenesis and targeting actions of microRNAs and small interfering RNAs, and global gene expression. Recent evidence suggests that RNA modifications mediated via RNA editing influence the biogenesis of circular RNAs and safeguard against aberrant innate immune responses generated to endogenous RNA sources. These novel roles have the potential to contribute new insights into molecular mechanisms underlying pathogenesis mediated by mishandling of double-stranded RNA. Here, we discuss recent advances in the field, which highlight novel roles associated with the RNA-editing process and emphasize their importance during cellular RNA metabolism. In addition, we highlight the relevance of these newly discovered roles in the context of neurological disorders and the more general concept of innate recognition of self versus non-self.

4.
Methods Mol Biol ; 1358: 255-68, 2016.
Article in English | MEDLINE | ID: mdl-26463388

ABSTRACT

Adenosine (A)-to-inosine (I) RNA editing is a fundamental posttranscriptional modification that ensures the deamination of A-to-I in double-stranded (ds) RNA molecules. Intriguingly, the A-to-I RNA editing system is particularly active in the nervous system of higher eukaryotes, altering a plethora of noncoding and coding sequences. Abnormal RNA editing is highly associated with many neurological phenotypes and neurodevelopmental disorders. However, the molecular mechanisms underlying RNA editing-mediated pathogenesis still remain enigmatic and have attracted increasing attention from researchers. Over the last decade, methods available to perform genome-wide transcriptome analysis, have evolved rapidly. Within the RNA editing field researchers have adopted next-generation sequencing technologies to identify RNA-editing sites within genomes and to elucidate the underlying process. However, technical challenges associated with editing site discovery have hindered efforts to uncover comprehensive editing site datasets, resulting in the general perception that the collections of annotated editing sites represent only a small minority of the total number of sites in a given organism, tissue, or cell type of interest. Additionally to doubts about sensitivity, existing RNA-editing site lists often contain high percentages of false positives, leading to uncertainty about their validity and usefulness in downstream studies. An accurate investigation of A-to-I editing requires properly validated datasets of editing sites with demonstrated and transparent levels of sensitivity and specificity. Here, we describe a high signal-to-noise method for RNA-editing site detection using single-molecule sequencing (SMS). With this method, authentic RNA-editing sites may be differentiated from artifacts. Machine learning approaches provide a procedure to improve upon and experimentally validate sequencing outcomes through use of computationally predicted, iterative feedback loops. Subsequent use of extensive Sanger sequencing validations can generate accurate editing site lists. This approach has broad application and accurate genome-wide editing analysis of various tissues from clinical specimens or various experimental organisms is now a possibility.


Subject(s)
Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , RNA Editing/genetics , RNA/genetics , Databases, Genetic , Genome, Human , Humans , Transcriptome
5.
BMC Biol ; 13: 1, 2015 Jan 03.
Article in English | MEDLINE | ID: mdl-25555396

ABSTRACT

BACKGROUND: Adenosine-to-inosine RNA editing is a highly conserved process that post-transcriptionally modifies mRNA, generating proteomic diversity, particularly within the nervous system of metazoans. Transcripts encoding proteins involved in neurotransmission predominate as targets of such modifications. Previous reports suggest that RNA editing is responsive to environmental inputs in the form of temperature alterations. However, the molecular determinants underlying temperature-dependent RNA editing responses are not well understood. RESULTS: Using the poikilotherm Drosophila, we show that acute temperature alterations within a normal physiological range result in substantial changes in RNA editing levels. Our examination of particular sites reveals diversity in the patterns with which editing responds to temperature, and these patterns are conserved across five species of Drosophilidae representing over 10 million years of divergence. In addition, we show that expression of the editing enzyme, ADAR (adenosine deaminase acting on RNA), is dramatically decreased at elevated temperatures, partially, but not fully, explaining some target responses to temperature. Interestingly, this reduction in editing enzyme levels at elevated temperature is only partially reversed by a return to lower temperatures. Lastly, we show that engineered structural variants of the most temperature-sensitive editing site, in a sodium channel transcript, perturb thermal responsiveness in RNA editing profile for a particular RNA structure. CONCLUSIONS: Our results suggest that the RNA editing process responds to temperature alterations via two distinct molecular mechanisms: through intrinsic thermo-sensitivity of the RNA structures that direct editing, and due to temperature sensitive expression or stability of the RNA editing enzyme. Environmental cues, in this case temperature, rapidly reprogram the Drosophila transcriptome through RNA editing, presumably resulting in altered proteomic ratios of edited and unedited proteins.


Subject(s)
Drosophila melanogaster/genetics , RNA Editing/genetics , Temperature , Adenosine Deaminase/metabolism , Animals , Conserved Sequence , Drosophila Proteins/metabolism , Models, Molecular , Mutation/genetics , Protein Isoforms/genetics
6.
J Neurophysiol ; 112(4): 903-12, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24805083

ABSTRACT

Hundreds of mutations in the SCN1A sodium channel gene confer a wide spectrum of epileptic disorders, requiring efficient model systems to study cellular mechanisms and identify potential therapeutic targets. We recently demonstrated that Drosophila knock-in flies carrying the K1270T SCN1A mutation known to cause a form of genetic epilepsy with febrile seizures plus (GEFS+) exhibit a heat-induced increase in sodium current activity and seizure phenotype. To determine whether different SCN1A mutations cause distinct phenotypes in Drosophila as they do in humans, this study focuses on a knock-in line carrying a mutation that causes a more severe seizure disorder termed Dravet syndrome (DS). Introduction of the DS SCN1A mutation (S1231R) into the Drosophila sodium channel gene para results in flies that exhibit spontaneous and heat-induced seizures with distinct characteristics and lower onset temperature than the GEFS+ flies. Electrophysiological studies of GABAergic interneurons in the brains of adult DS flies reveal, for the first time in an in vivo model system, that a missense DS mutation causes a constitutive and conditional reduction in sodium current activity and repetitive firing. In addition, feeding with the serotonin precursor 5-HTP suppresses heat-induced seizures in DS but not GEFS+ flies. The distinct alterations of sodium currents in DS and GEFS+ GABAergic interneurons demonstrate that both loss- and gain-of-function alterations in sodium currents are capable of causing reduced repetitive firing and seizure phenotypes. The mutation-specific effects of 5-HTP on heat-induced seizures suggest the serotonin pathway as a potential therapeutic target for DS.


Subject(s)
Action Potentials , Epilepsies, Myoclonic/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Sodium/metabolism , 5-Hydroxytryptophan/metabolism , Animals , Brain/cytology , Brain/metabolism , Brain/physiopathology , Drosophila/genetics , Drosophila/metabolism , Drosophila/physiology , Epilepsies, Myoclonic/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Interneurons/metabolism , Interneurons/physiology , Mutation, Missense , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Phenotype , Serotonin/metabolism
7.
Genome Biol ; 15(1): 103, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24468094

ABSTRACT

A large comparative genomic sequence study has determined the extent of conservation between RNA editing sites within the mammalian evolutionary tree.


Subject(s)
Mammals/genetics , RNA Editing/genetics , RNA-Binding Proteins/genetics , Animals , Humans
8.
Nat Commun ; 4: 2745, 2013.
Article in English | MEDLINE | ID: mdl-24201902

ABSTRACT

Heterochromatin formation drives epigenetic mechanisms associated with silenced gene expression. Repressive heterochromatin is established through the RNA interference pathway, triggered by double-stranded RNAs (dsRNAs) that can be modified via RNA editing. However, the biological consequences of such modifications remain enigmatic. Here we show that RNA editing regulates heterochromatic gene silencing in Drosophila. We utilize the binding activity of an RNA-editing enzyme to visualize the in vivo production of a long dsRNA trigger mediated by Hoppel transposable elements. Using homologous recombination, we delete this trigger, dramatically altering heterochromatic gene silencing and chromatin architecture. Furthermore, we show that the trigger RNA is edited and that dADAR serves as a key regulator of chromatin state. Additionally, dADAR auto-editing generates a natural suppressor of gene silencing. Lastly, systemic differences in RNA editing activity generates interindividual variation in silencing state within a population. Our data reveal a global role for RNA editing in regulating gene expression.


Subject(s)
DNA Transposable Elements/physiology , Gene Silencing , Heterochromatin/genetics , RNA Editing/physiology , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , DNA Transposable Elements/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Protein Binding , RNA Editing/genetics , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism
9.
Nat Struct Mol Biol ; 20(11): 1333-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24077224

ABSTRACT

The accurate and thorough genome-wide detection of adenosine-to-inosine editing, a biologically indispensable process, has proven challenging. Here, we present a discovery pipeline in adult Drosophila, with 3,581 high-confidence editing sites identified with an estimated accuracy of 87%. The target genes and specific sites highlight global biological properties and functions of RNA editing, including hitherto-unknown editing in well-characterized classes of noncoding RNAs and 645 sites that cause amino acid substitutions, usually at conserved positions. The spectrum of functions that these gene targets encompass suggests that editing participates in a diverse set of cellular processes. Editing sites in Drosophila exhibit sequence-motif preferences and tend to be concentrated within a small subset of total RNAs. Finally, editing regulates expression levels of target mRNAs and strongly correlates with alternative splicing.


Subject(s)
Adenosine/genetics , Adenosine/metabolism , Inosine/genetics , Inosine/metabolism , RNA Editing , RNA/genetics , RNA/metabolism , Animals , Drosophila , Gene Expression , Genome, Insect
10.
Bioinformatics ; 29(22): 2844-51, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24048353

ABSTRACT

MOTIVATION: Validation and reproducibility of results is a central and pressing issue in genomics. Several recent embarrassing incidents involving the irreproducibility of high-profile studies have illustrated the importance of this issue and the need for rigorous methods for the assessment of reproducibility. RESULTS: Here, we describe an existing statistical model that is very well suited to this problem. We explain its utility for assessing the reproducibility of validation experiments, and apply it to a genome-scale study of adenosine deaminase acting on RNA (ADAR)-mediated RNA editing in Drosophila. We also introduce a statistical method for planning validation experiments that will obtain the tightest reproducibility confidence limits, which, for a fixed total number of experiments, returns the optimal number of replicates for the study. AVAILABILITY: Downloadable software and a web service for both the analysis of data from a reproducibility study and for the optimal design of these studies is provided at http://ccmbweb.ccv.brown.edu/reproducibility.html .


Subject(s)
Genomics/methods , Models, Statistical , Adenosine Deaminase , Animals , Drosophila/genetics , Genome , RNA Editing , Reproducibility of Results , Software
11.
Nat Commun ; 3: 790, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22531175

ABSTRACT

Auto-regulatory feedback loops are a common molecular strategy used to optimize protein function. In Drosophila, many messenger RNAs involved in neuro-transmission are re-coded at the RNA level by the RNA-editing enzyme, dADAR, leading to the incorporation of amino acids that are not directly encoded by the genome. dADAR also re-codes its own transcript, but the consequences of this auto-regulation in vivo are unclear. Here we show that hard-wiring or abolishing endogenous dADAR auto-regulation dramatically remodels the landscape of re-coding events in a site-specific manner. These molecular phenotypes correlate with altered localization of dADAR within the nuclear compartment. Furthermore, auto-editing exhibits sexually dimorphic patterns of spatial regulation and can be modified by abiotic environmental factors. Finally, we demonstrate that modifying dAdar auto-editing affects adaptive complex behaviours. Our results reveal the in vivo relevance of auto-regulatory control over post-transcriptional mRNA re-coding events in fine-tuning brain function and organismal behaviour.


Subject(s)
Adenosine Deaminase/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Homeostasis , RNA Editing , RNA, Messenger/genetics , Adenosine Deaminase/chemistry , Adenosine Deaminase/metabolism , Amino Acid Sequence , Animals , Cell Nucleus/enzymology , Cell Nucleus/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/chemistry , Gene Expression Regulation , Male , Molecular Sequence Data , RNA, Messenger/metabolism , RNA-Binding Proteins , Sequence Alignment
12.
Front Genet ; 3: 57, 2012.
Article in English | MEDLINE | ID: mdl-22539933

ABSTRACT

Perhaps no other topic in contemporary genomics has inspired such diverse viewpoints as the 95+% of the genome, previously known as "junk DNA," that does not code for proteins. Here, we present a theory in which dark matter RNA plays a role in the generation of a landscape of spatial micro-domains coupled to the information signaling matrix of the nuclear landscape. Within and between these micro-domains, dark matter RNAs additionally function to tether RNA interacting proteins and complexes of many different types, and by doing so, allow for a higher performance of the various processes requiring them at ultra-fast rates. This improves signal to noise characteristics of RNA processing, trafficking, and epigenetic signaling, where competition and differential RNA binding among proteins drives the computational decisions inherent in regulatory events.

13.
Genome Biol ; 13(12): 252, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23273215

ABSTRACT

Adenosine to inosine (A-to-I) RNA editing is a post-transcriptional process by which adenosines are selectively converted to inosines in double-stranded RNA (dsRNA) substrates. A highly conserved group of enzymes, the adenosine deaminase acting on RNA (ADAR) family, mediates this reaction. All ADARs share a common domain architecture consisting of a variable number of amino-terminal dsRNA binding domains (dsRBDs) and a carboxy-terminal catalytic deaminase domain. ADAR family members are highly expressed in the metazoan nervous system, where these enzymes predominantly localize to the neuronal nucleus. Once in the nucleus, ADARs participate in the modification of specific adenosines in pre-mRNAs of proteins involved in electrical and chemical neurotransmission, including pre-synaptic release machineries, and voltage- and ligand-gated ion channels. Most RNA editing sites in these nervous system targets result in non-synonymous codon changes in functionally important, usually conserved, residues and RNA editing deficiencies in various model organisms bear out a crucial role for ADARs in nervous system function. Mutation or deletion of ADAR genes results in striking phenotypes, including seizure episodes, extreme uncoordination, and neurodegeneration. Not only does the process of RNA editing alter important nervous system peptides, but ADARs also regulate gene expression through modification of dsRNA substrates that enter the RNA interference (RNAi) pathway and may then act at the chromatin level. Here, we present a review on the current knowledge regarding the ADAR protein family, including evolutionary history, key structural features, localization, function and mechanism.


Subject(s)
Adenosine Deaminase/genetics , Adenosine Deaminase/physiology , Adenosine Deaminase/analysis , Adenosine Deaminase/chemistry , Animals , Evolution, Molecular , Humans , Multigene Family , RNA Editing , RNA Splicing
14.
Nat Methods ; 9(2): 189-94, 2011 Dec 25.
Article in English | MEDLINE | ID: mdl-22198342

ABSTRACT

Informational recoding by adenosine-to-inosine RNA editing diversifies neuronal proteomes by chemically modifying structured mRNAs. However, techniques for analyzing editing activity on substrates in defined neurons in vivo are lacking. Guided by comparative genomics, here we reverse-engineered a fluorescent reporter sensitive to Drosophila melanogaster adenosine deaminase that acts on RNA (dADAR) activity and alterations in dADAR autoregulation. Using this artificial dADAR substrate, we visualized variable patterns of RNA-editing activity in the Drosophila nervous system between individuals. Our results demonstrate the feasibility of structurally mimicking ADAR substrates as a method to regulate protein expression and, potentially, therapeutically repair mutant mRNAs. Our data suggest variable RNA editing as a credible molecular mechanism for mediating individual-to-individual variation in neuronal physiology and behavior.


Subject(s)
Adenosine Deaminase/metabolism , Adenosine/genetics , Inosine/genetics , Nervous System/metabolism , RNA Editing , Adenosine Deaminase/chemistry , Animals , Drosophila melanogaster , Genes, Reporter , Green Fluorescent Proteins/genetics
15.
Nat Neurosci ; 14(12): 1517-24, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-22037499

ABSTRACT

Loss of FMR1 gene function results in fragile X syndrome, the most common heritable form of intellectual disability. The protein encoded by this locus (FMRP) is an RNA-binding protein that is thought to primarily act as a translational regulator; however, recent studies have implicated FMRP in other mechanisms of gene regulation. We found that the Drosophila fragile X homolog (dFMR1) biochemically interacted with the adenosine-to-inosine RNA-editing enzyme dADAR. Adar and Fmr1 mutant larvae exhibited distinct morphological neuromuscular junction (NMJ) defects. Epistasis experiments based on these phenotypic differences revealed that Adar acts downstream of Fmr1 and that dFMR1 modulates dADAR activity. Furthermore, sequence analyses revealed that a loss or overexpression of dFMR1 affects editing efficiency on certain dADAR targets with defined roles in synaptic transmission. These results link dFMR1 with the RNA-editing pathway and suggest that proper NMJ synaptic architecture requires modulation of dADAR activity by dFMR1.


Subject(s)
Adenosine Deaminase/metabolism , Drosophila Proteins/metabolism , Fragile X Mental Retardation Protein/metabolism , Neuromuscular Junction/metabolism , RNA Editing/genetics , Adenosine Deaminase/genetics , Analysis of Variance , Animals , Animals, Genetically Modified , Cell Line, Transformed , Drosophila , Drosophila Proteins/genetics , Fragile X Mental Retardation Protein/genetics , Gene Expression Regulation, Developmental , Immunoprecipitation , Larva , Mass Spectrometry , Microscopy, Confocal , Mutation , Neuromuscular Junction/cytology , Neuromuscular Junction/genetics , RNA-Binding Proteins , Synaptic Transmission , Transfection
16.
J Biol Chem ; 286(10): 8325-8337, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21078670

ABSTRACT

Select proteins involved in electrical and chemical neurotransmission are re-coded at the RNA level via the deamination of particular adenosines to inosine by adenosine deaminases acting on RNA (ADARs). It has been hypothesized that this process, termed RNA editing, acts to "fine-tune" neurophysiological properties in animals and potentially downstream behavioral outputs. However, the extreme phenotypes resulting from deletions of adar loci have precluded investigations into the relationship between ADAR levels, target transcripts, and complex behaviors. Here, we engineer Drosophila hypomorphic for ADAR expression using homologous recombination. A substantial reduction in ADAR activity (>80%) leads to altered circadian motor patterns and abnormal male courtship, although surprisingly, general locomotor coordination is spared. The altered phenotypic landscape in our adar hypomorph is paralleled by an unexpected dichotomous response of ADAR target transcripts, i.e. certain adenosines are minimally affected by dramatic ADAR reduction, whereas editing of others is severely curtailed. Furthermore, we use a novel reporter to map RNA editing activity across the nervous system, and we demonstrate that knockdown of editing in fruitless-expressing neurons is sufficient to modify the male courtship song. Our data demonstrate that network-wide temporal and spatial regulation of ADAR activity can tune the complex system of RNA-editing sites and modulate multiple ethologically relevant behavioral modalities.


Subject(s)
Adenosine Deaminase/metabolism , Behavior, Animal/physiology , Gene Expression Regulation, Enzymologic/physiology , Genetic Loci/physiology , Neurons/metabolism , RNA Editing/physiology , Adenosine Deaminase/genetics , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Locomotion/physiology , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , RNA-Binding Proteins , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Neurosci Lett ; 466(2): 89-98, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19751800

ABSTRACT

The depth and complexity of the non-coding transcriptome in nervous system tissues provides a rich substrate for adenosine de-amination acting on RNA (ADAR). Non-coding RNAs (ncRNAs) serve diverse regulatory and computational functions, coupling signal flow from the environment to evolutionarily coded analog and digital information elements within the transcriptome. We present a perspective of ADARs interaction with the non-coding transcriptome as a computational matrix, enhancing the information processing power of the cell, adding flexibility, rapid response, and fine tuning to critical pathways. Dramatic increases in ADAR activity during stress response and inflammation result in powerful information processing events that change the functional state of the cell. This review examines the pathways and mechanisms of ADAR interaction with the non-coding transcriptome, and their functional consequences for information processing in nervous system tissues.


Subject(s)
Adenosine Deaminase/metabolism , Adenosine/metabolism , RNA Editing/physiology , RNA, Untranslated/physiology , Signal Transduction/physiology , Adenosine Deaminase/genetics , Animals , Gene Expression Profiling , Humans , Models, Molecular , Nervous System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...