Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Neurosci ; 39(12): 2347-2364, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30659087

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease for which the origin and underlying cellular defects are not fully understood. Although motor neuron degeneration is the signature feature of ALS, it is not clear whether motor neurons or other cells of the motor circuit are the site of disease initiation. To better understand the contribution of multiple cell types in ALS, we made use of a Drosophila Sod1G85R knock-in model, in which all cells harbor the disease allele. End-stage dSod1G85R animals of both sexes exhibit severe motor deficits with clear degeneration of motor neurons. Interestingly, earlier in dSod1G85R larvae, motor function is also compromised, but their motor neurons exhibit only subtle morphological and electrophysiological changes that are unlikely to cause the observed decrease in locomotion. We analyzed the intact motor circuit and identified a defect in sensory feedback that likely accounts for the altered motor activity of dSod1G85R We found cell-autonomous activation of bone morphogenetic protein signaling in proprioceptor sensory neurons which are critical for the relay of the contractile status of muscles back to the central nerve cord, completely rescues early-stage motor defects and partially rescue late-stage motor function to extend lifespan. Identification of a defect in sensory feedback as a potential initiating event in ALS motor dysfunction, coupled with the ability of modified proprioceptors to alleviate such motor deficits, underscores the critical role that nonmotor neurons play in disease progression and highlights their potential as a site to identify early-stage ALS biomarkers and for therapeutic intervention.SIGNIFICANCE STATEMENT At diagnosis, many cellular processes are already disrupted in the amyotrophic lateral sclerosis (ALS) patient. Identifying the initiating cellular events is critical for achieving an earlier diagnosis to slow or prevent disease progression. Our findings indicate that neurons relaying sensory information underlie early stage motor deficits in a Drosophila knock-in model of ALS that best replicates gene dosage in familial ALS (fALS). Importantly, studies on intact motor circuits revealed defects in sensory feedback before evidence of motor neuron degeneration. These findings strengthen our understanding of how neural circuit dysfunctions lead to neurodegeneration and, coupled with our demonstration that the activation of bone morphogenetic protein signaling in proprioceptors alleviates both early and late motor dysfunction, underscores the importance of considering nonmotor neurons as therapeutic targets.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Retroalimentação Sensorial/fisiologia , Neurônios Motores/metabolismo , Degeneração Neural/metabolismo , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila , Proteínas de Drosophila/genética , Feminino , Técnicas de Introdução de Genes , Locomoção , Masculino , Propriocepção/fisiologia , Transdução de Sinais , Superóxido Dismutase/genética
2.
BMC Genomics ; 18(1): 522, 2017 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-28693467

RESUMO

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.


Assuntos
Drosophila melanogaster/genética , Edição de RNA , Alinhamento de Sequência , Algoritmos , Animais , Rearranjo Gênico , Polimorfismo de Nucleotídeo Único , Sequências Repetitivas de Ácido Nucleico/genética
3.
Proc Natl Acad Sci U S A ; 111(1): 337-42, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24367095

RESUMO

PIWI proteins and their bound PIWI-interacting RNAs (piRNAs) are found in animal germlines and are essential for fertility, but their functions outside of the gonad are not well understood. The cnidarian Hydra is a simple metazoan with well-characterized stem/progenitor cells that provides a unique model for analysis of PIWI function. Here we report that Hydra has two PIWI proteins, Hydra PIWI (Hywi) and Hydra PIWI-like (Hyli), both of which are expressed in all Hydra stem/progenitor cells, but not in terminally differentiated cells. We identified ∼15 million piRNAs associated with Hywi and/or Hyli and found that they exhibit the ping-pong signature of piRNA biogenesis. Hydra PIWI proteins are strictly cytoplasmic and thus likely act as posttranscriptional regulators. To explore this function, we generated a Hydra transcriptome for piRNA mapping. piRNAs map to transposons with a 25- to 35-fold enrichment compared with the abundance of transposon transcripts. By sequencing the small RNAs specific to the interstitial, ectodermal, and endodermal lineages, we found that the targeting of transposons appears to be largely restricted to the interstitial lineage. We also identified putative nontransposon targets of the pathway unique to each lineage. Finally we demonstrate that hywi function is essential in the somatic epithelial lineages. This comprehensive analysis of the PIWI-piRNA pathway in the somatic stem/progenitor cells of a nonbilaterian animal suggests that this pathway originated with broader stem cell functionality.


Assuntos
Proteínas Argonautas/metabolismo , Regulação da Expressão Gênica , Hydra/genética , Hydra/metabolismo , RNA Interferente Pequeno/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Separação Celular , Citoplasma/metabolismo , Células Epiteliais/citologia , Citometria de Fluxo , Filogenia , RNA/metabolismo , Interferência de RNA , Processamento Pós-Transcricional do RNA , Proteínas Recombinantes/química , Especificidade da Espécie , Transcriptoma , Transgenes
4.
BMC Biol ; 13: 1, 2015 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-25555396

RESUMO

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.


Assuntos
Drosophila melanogaster/genética , Edição de RNA/genética , Temperatura , Adenosina Desaminase/metabolismo , Animais , Sequência Conservada , Proteínas de Drosophila/metabolismo , Modelos Moleculares , Mutação/genética , Isoformas de Proteínas/genética
5.
Semin Cell Dev Biol ; 23(3): 281-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22178616

RESUMO

Post-transcriptional modifications such as RNA editing and splicing diversify the proteome while limiting the necessary size of the genome. Although splicing globally rearranges existing information within the transcript, the conserved process of adenosine-to-inosine RNA editing recodes the message through single nucleotide changes, often at very specific locations. Because inosine is interpreted as guanosine by the cellular machineries, editing effectively results in the substitution of a guanosine for an adenosine in the primary RNA sequence. Precise control of editing is dictated by duplex structures in the transcript, formed between the exonic region surrounding the editing site and cis regulatory elements often localized in a nearby intron, suggesting that editing must precede splicing. However, the precise relationship between these post-transcriptional processes remains unclear. Here we present general commonalities of RNA editing substrates and consequential predictions regarding the interaction between editing and splicing. We also discuss anomalies and interesting cases of RNA editing that confound our understanding of the relationship between these post-transcriptional processes.


Assuntos
Conformação de Ácido Nucleico , Edição de RNA , Splicing de RNA/genética , RNA , Animais , Humanos , RNA/química , RNA/genética , RNA/metabolismo
6.
Nat Methods ; 9(2): 189-94, 2011 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-22198342

RESUMO

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.


Assuntos
Adenosina Desaminase/metabolismo , Adenosina/genética , Inosina/genética , Sistema Nervoso/metabolismo , Edição de RNA , Adenosina Desaminase/química , Animais , Drosophila melanogaster , Genes Reporter , Proteínas de Fluorescência Verde/genética
7.
J Biol Chem ; 286(10): 8325-8337, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21078670

RESUMO

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.


Assuntos
Adenosina Desaminase/metabolismo , Comportamento Animal/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Loci Gênicos/fisiologia , Neurônios/metabolismo , Edição de RNA/fisiologia , Adenosina Desaminase/genética , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Feminino , Locomoção/fisiologia , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Proteínas de Ligação a RNA , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Nature ; 434(7031): 409-13, 2005 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-15772668

RESUMO

Most RNA editing systems are mechanistically diverse, informationally restorative, and scattershot in eukaryotic lineages. In contrast, genetic recoding by adenosine-to-inosine RNA editing seems common in animals; usually, altering highly conserved or invariant coding positions in proteins. Here I report striking variation between species in the recoding of synaptotagmin I (sytI). Fruitflies, mosquitoes and butterflies possess shared and species-specific sytI editing sites, all within a single exon. Honeybees, beetles and roaches do not edit sytI. The editing machinery is usually directed to modify particular adenosines by information stored in intron-mediated RNA structures. Combining comparative genomics of 34 species with mutational analysis reveals that complex, multi-domain, pre-mRNA structures solely determine species-appropriate RNA editing. One of these is a previously unreported long-range pseudoknot. I show that small changes to intronic sequences, far removed from an editing site, can transfer the species specificity of editing between RNA substrates. Taken together, these data support a phylogeny of sytI gene editing spanning more than 250 million years of hexapod evolution. The results also provide models for the genesis of RNA editing sites through the stepwise addition of structural domains, or by short walks through sequence space from ancestral structures.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Evolução Molecular , Genômica , Glicoproteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Edição de RNA/genética , Adenosina/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Éxons/genética , Inosina/genética , Íntrons/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Filogenia , Precursores de RNA/genética , Precursores de RNA/metabolismo , Especificidade da Espécie , Sinaptotagmina I , Sinaptotagminas , Fatores de Tempo
9.
J Biol Chem ; 284(45): 31391-400, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19759011

RESUMO

Adenosine deaminases acting on RNA (ADARs) catalyze the deamination of adenosine to inosine in double-stranded RNA templates, a process known as RNA editing. In Drosophila, multiple ADAR isoforms are generated from a single locus (dAdar) via post-transcriptional modifications. Collectively, these isoforms act to edit a wide range of transcripts involved in neuronal signaling, as well as the precursors of endogenous small interfering RNAs. The phenotypic consequences of a loss of dADAR activity have been well characterized and consist of profound behavioral defects manifested at the adult stage, including extreme uncoordination, seizures, and temperature-sensitive paralysis. However, the spatio-temporal requirements of adenosine to inosine editing for correct behavior are unclear. Using transgenic RNA interference, we show that network-wide editing in the nervous system is required for normal adult locomotion. Regulated restoration of editing activity demonstrates that the neuronal requirement of dADAR activity has a significant adult stage component. Furthermore we show that in relation to behavior there are no observable genetic interactions between dAdar and several loci encoding RNA interference components, suggesting that editing of neuronal transcripts is the key mode of ADAR activity for normal behavior in Drosophila.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/fisiologia , Edição de RNA , Adenosina/genética , Adenosina/metabolismo , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Animais , Comportamento Animal , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Feminino , Inosina/genética , Inosina/metabolismo , Locomoção , Masculino , Sistema Nervoso/crescimento & desenvolvimento , Sistema Nervoso/metabolismo
10.
Biochim Biophys Acta ; 1779(8): 459-70, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18086576

RESUMO

Adenosine to inosine RNA editing, catalyzed by Adenosine Deaminases Acting on RNA (ADARs), represents an evolutionary conserved post-transcriptional mechanism which harnesses RNA structures to produce proteins that are not literally encoded in the genome. The species-specific alteration of functionally important residues in a multitude of neuronal ion channels and pre-synaptic proteins through RNA editing has been shown to have profound importance for normal nervous system function in a wide range of invertebrate and vertebrate model organisms. ADARs have also been shown to regulate neuronal gene expression through a remarkable variety of disparate processes, including modulation of the RNAi pathway, the creation of alternative splice sites, and the abolition of stop codons. In addition, ADARs have recently been revealed to have a novel role in the primate lineage: the widespread editing of Alu elements, which comprise approximately 10% of the human genome. Thus, as well as enabling the cell-specific regulation of RNAi and selfish genetic elements, the unshackling of the proteome from the constraints of the genome through RNA editing may have been fundamental to the evolution of complex behavior.


Assuntos
Adenosina Desaminase/fisiologia , Encéfalo/metabolismo , Regulação da Expressão Gênica , Edição de RNA/fisiologia , Adenosina/metabolismo , Elementos Alu , Sequência de Aminoácidos , Animais , Genoma Humano , Humanos , Inosina/metabolismo , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Conformação de Ácido Nucleico , Edição de RNA/genética , Interferência de RNA , Proteínas de Ligação a RNA
11.
Neuroscience ; 396: A3-A20, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30594291

RESUMO

Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease that has significant overlap with frontotemporal dementia (FTD). Mutations in specific genes have been identified that can cause and/or predispose patients to ALS. However, the clinical variability seen in ALS patients suggests that additional genes impact pathology, susceptibility, severity, and/or progression of the disease. To identify molecular pathways involved in ALS, we undertook a meta-analysis of published genetic modifiers both in patients and in model organisms, and undertook bioinformatic pathway analysis. From 72 published studies, we generated a list of 946 genes whose perturbation (1) impacted ALS in patient populations, (2) altered defects in laboratory models, or (3) modified defects caused by ALS gene ortholog loss of function. Herein, these are all called modifier genes. We found 727 modifier genes that encode proteins with human orthologs. Of these, 43 modifier genes were identified as modifiers of more than one ALS gene/model, consistent with the hypothesis that shared genes and pathways may underlie ALS. Further, we used a gene ontology-based bioinformatic analysis to identify pathways and associated genes that may be important in ALS. To our knowledge this is the first comprehensive survey of ALS modifier genes. This work suggests that shared molecular mechanisms may underlie pathology caused by different ALS disease genes. Surprisingly, few ALS modifier genes have been tested in more than one disease model. Understanding genes that modify ALS-associated defects will help to elucidate the molecular pathways that underlie ALS and provide additional targets for therapeutic intervention.


Assuntos
Esclerose Lateral Amiotrófica/genética , Genes Modificadores/genética , Transdução de Sinais/genética , Animais , Biologia Computacional , Predisposição Genética para Doença/genética , Humanos
12.
Cell Rep ; 23(7): 1915-1921, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29768192

RESUMO

Transcriptional modulation of the process of autophagy involves the transcription factor HLH-30/TFEB. In order to systematically determine the regulatory network of HLH-30/TFEB, we performed a genome-wide RNAi screen in C. elegans and found that silencing the nuclear export protein XPO-1/XPO1 enhances autophagy by significantly enriching HLH-30 in the nucleus, which is accompanied by proteostatic benefits and improved longevity. Lifespan extension via xpo-1 silencing requires HLH-30 and autophagy, overlapping mechanistically with several established longevity models. Selective XPO1 inhibitors recapitulated the effect on autophagy and lifespan observed by silencing xpo-1 and protected ALS-afflicted flies from neurodegeneration. XPO1 inhibition in HeLa cells enhanced TFEB nuclear localization, autophagy, and lysosome biogenesis without affecting mTOR activity, revealing a conserved regulatory mechanism for HLH-30/TFEB. Altogether, our study demonstrates that altering the nuclear export of HLH-30/TFEB can regulate autophagy and establishes the rationale of targeting XPO1 to stimulate autophagy in order to prevent neurodegeneration.


Assuntos
Autofagia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Núcleo Celular/metabolismo , Longevidade , Transporte Ativo do Núcleo Celular , Animais , Caenorhabditis elegans/metabolismo , Inativação Gênica , Células HeLa , Humanos
13.
Methods Enzymol ; 424: 245-64, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17662844

RESUMO

Adenosine-to-inosine (A-to-I) RNA editing is the enzymatic deamination of A-to-I catalyzed by ADAR (adenosine deaminase acting on RNA). Adenosine is read by ribosomes as guanosine causing a codon change and potentially protein recoding. A-to-I RNA editing can be either promiscuous, where the editing is nonspecific, or site specific, which requires a complex target RNA secondary structure formed by intramolecular base pairings between editing sequences and intronic or exonic editing site complementary sequences (ECSs). The most numerous editing sites have been found in noncoding regions containing Alu repeats, such as 3' untranslated regions, while specific editing sites are mostly found in transcripts involved in the transmission of neuronal signals. Previously A-to-I RNA editing sites were discovered by chance, but recently investigators have used comparative genomic and bioinformatics methods to identify novel sites. In this chapter, we discuss these approaches to identifying new editing sites.


Assuntos
Adenosina/química , Biologia Computacional/métodos , Genômica/métodos , Inosina/química , Animais , Sequência de Bases , Sítios de Ligação , Evolução Molecular , Técnicas Genéticas , Humanos , Dados de Sequência Molecular , Primatas , Edição de RNA , RNA Mensageiro/metabolismo , Especificidade por Substrato
14.
Methods Enzymol ; 424: 265-87, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17662845

RESUMO

Increasing proteomic diversity via the hydrolytic deamination of adenosine to inosine (A-to-I) in select mRNA templates appears crucial to the correct functioning of the nervous system in several model organisms, including Drosophila, Caenorabditis elegans, and mice. The genome of the fruitfly, Drosophila melanogaster, contains a single gene encoding the enzyme responsible for deamination, termed ADAR (for adenosine deaminase acting on RNA). The mRNAs that form the substrates for ADAR primarily function in neuronal signaling, and, correspondingly, deletion of ADAR leads to severe nervous system defects. While several ADAR enzymes are present in mice, the presence of a single ADAR in Drosophila, combined with the diverse genetic toolkit available to researchers and the wide range of ADAR target mRNAs identified to date, make Drosophila an ideal organism to study the genetic basis of A-to-I RNA editing. This chapter describes a variety of methods for genetically manipulating Drosophila A-to-I editing both in time and space, as well as techniques to study the molecular basis of ADAR-mRNA interactions. A prerequisite for experiments in this field is the ability to quantify the levels of editing in a given mRNA. Therefore, several commonly used methods for the quantification of editing levels will also be described.


Assuntos
Adenosina Desaminase/genética , Adenosina/química , Técnicas Genéticas , Inosina/química , Edição de RNA/genética , Animais , Linhagem Celular , Primers do DNA/química , Enzimas de Restrição do DNA/metabolismo , Drosophila , Genômica/métodos , Genótipo , Modelos Genéticos , Conformação de Ácido Nucleico , Saccharomyces cerevisiae/metabolismo
15.
Peptides ; 28(1): 51-6, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17141372

RESUMO

We have developed a model genetic system for analyzing the function of peptide toxins from animal venoms. We engineered and propagated strains of Drosophila melanogaster expressing heat-inducible transgenes encoding either kappa-ACTX-Hv1c or omega-ACTX-Hv1a, two insect-specific neurotoxic peptides found in the venom of the Australian funnel-web spider Hadronyche versuta. Heat induction of transgene expression for 20 min was sufficient to kill all transgenic flies, indicating that the ion channels targeted by these toxins are viable insecticide targets. The unusual phenotype of flies induced to express omega-ACTX-Hv1a recapitulates that of a hypomorphic allele of the high-voltage-activated calcium channel Dmca1D, suggesting that this is likely to be the target of omega-ACTX-Hv1a.


Assuntos
Peptídeos/fisiologia , Venenos de Aranha/genética , Toxinas Biológicas/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Viúva Negra/genética , Viúva Negra/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Expressão Gênica , Vetores Genéticos/genética , Modelos Biológicos , Dados de Sequência Molecular , Neurotoxinas/genética , Neurotoxinas/metabolismo , Peptídeos/genética , Alinhamento de Sequência , Venenos de Aranha/metabolismo , Toxinas Biológicas/genética , Transformação Genética
16.
Methods Mol Biol ; 1358: 255-68, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26463388

RESUMO

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.


Assuntos
Perfilação da Expressão Gênica/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Edição de RNA/genética , RNA/genética , Bases de Dados Genéticas , Genoma Humano , Humanos , Transcriptoma
17.
Front Genet ; 7: 100, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27458478

RESUMO

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.

18.
Genome Biol ; 15(1): 103, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24468094

RESUMO

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


Assuntos
Mamíferos/genética , Edição de RNA/genética , Proteínas de Ligação a RNA/genética , Animais , Humanos
19.
Aging (Albany NY) ; 6(1): 58-69, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24519859

RESUMO

Natural selection acts to maximize reproductive fitness. However, antagonism between life span and reproductive success frequently poses a dilemma pitting the cost of fecundity against longevity. Here, we show that natural populations of Drosophila melanogaster harbor a Hoppel transposon insertion variant in the longevity gene Indy (I'm not dead yet), which confers both increased reproduction and longevity through metabolic changes. Heterozygosity for this natural long-lived variant has been maintained in isolates despite long-term inbreeding under laboratory conditions and advantageously confers increased fecundity. DNA sequences of variant chromosome isolates show evidence of selective sweep acting on the advantageous allele, suggesting that natural selection acts to maintain this variant. The transposon insertion also regulates Indy expression level, which has experimentally been shown to affect life span and fecundity. Thus, in the wild, evolution reaffirms that the mechanism of heterozygote advantage has acted upon the Indy gene to assure increased reproductive fitness and, coincidentally, longer life span through regulatory transposon mutagenesis.


Assuntos
Elementos de DNA Transponíveis , Transportadores de Ácidos Dicarboxílicos/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Aptidão Genética , Longevidade/genética , Polimorfismo Genético , Seleção Genética/genética , Simportadores/genética , Animais , Transportadores de Ácidos Dicarboxílicos/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Fertilidade/genética , Regulação da Expressão Gênica , Genótipo , Heterozigoto , Fenótipo , RNA Mensageiro/metabolismo , Simportadores/metabolismo
20.
Genetics ; 193(3): 771-84, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23267055

RESUMO

Diploid sexual reproduction involves segregation of allelic pairs, ensuring equal representation of genotypes in the gamete pool. Some genes, however, are able to "cheat" the system by promoting their own transmission. The Segregation distorter (Sd) locus in Drosophila melanogaster males is one of the best-studied examples of this type of phenomenon. In this system the presence of Sd on one copy of chromosome 2 results in dysfunction of the non-Sd-bearing (Sd(+)) sperm and almost exclusive transmission of Sd to the next generation. The mechanism by which Sd wreaks such selective havoc has remained elusive. However, its effect requires a target locus on chromosome 2 known as Responder (Rsp). The Rsp locus comprises repeated copies of a satellite DNA sequence and Rsp copy number correlates with sensitivity to Sd. Under distorting conditions during spermatogenesis, nuclei with chromosomes containing greater than several hundred Rsp repeats fail to condense chromatin and are eliminated. Recently, Rsp sequences were found as small RNAs in association with Argonaute family proteins Aubergine (Aub) and Argonaute3 (AGO3). These proteins are involved in a germline-specific RNAi mechanism known as the Piwi-interacting RNA (piRNA) pathway, which specifically suppresses transposon activation in the germline. Here, we evaluate the role of piRNAs in segregation distortion by testing the effects of mutations to piRNA pathway components on distortion. Further, we specifically targeted mutations to the aub locus of a Segregation Distorter (SD) chromosome, using ends-out homologous recombination. The data herein demonstrate that mutations to piRNA pathway components act as enhancers of SD.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas Ativadoras de GTPase/genética , Meiose/genética , Fatores de Iniciação de Peptídeos/genética , RNA Interferente Pequeno/genética , Sequência de Aminoácidos , Animais , Cromatina , Cromossomos , DNA Satélite , Proteínas de Drosophila/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Dosagem de Genes , Loci Gênicos , Recombinação Homóloga , Dados de Sequência Molecular , Mutação , Fatores de Iniciação de Peptídeos/metabolismo , RNA Interferente Pequeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa