Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
RNA Biol ; 19(1): 68-77, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34965182

RESUMO

DNA double-strand breaks are among the most toxic lesions that can occur in a genome and their faithful repair is thus of great importance. Recent findings have uncovered local transcription that initiates at the break and forms a non-coding transcript, called damage-induced long non-coding RNA (dilncRNA), which helps to coordinate the DNA transactions necessary for repair. We provide nascent RNA sequencing-based evidence that RNA polymerase II transcribes the dilncRNA in Drosophila and that this is more efficient for DNA breaks in an intron-containing gene, consistent with the higher damage-induced siRNA levels downstream of an intron. The spliceosome thus stimulates recruitment of RNA polymerase II to the break, rather than merely promoting the annealing of sense and antisense RNA to form the siRNA precursor. In contrast, RNA polymerase III nascent RNA libraries did not contain reads corresponding to the cleaved loci and selective inhibition of RNA polymerase III did not reduce the yield of damage-induced siRNAs. Finally, the damage-induced siRNA density was unchanged downstream of a T8 sequence, which terminates RNA polymerase III transcription. We thus found no evidence for a participation of RNA polymerase III in dilncRNA transcription in cultured Drosophila cells.


Assuntos
Quebras de DNA de Cadeia Dupla , Drosophila/genética , Drosophila/metabolismo , RNA Polimerase II/metabolismo , RNA Longo não Codificante/genética , Transcrição Gênica , Animais , Reparo do DNA , Regulação da Expressão Gênica , Íntrons , Ligação Proteica , RNA Polimerase III/metabolismo , Splicing de RNA , RNA Interferente Pequeno/genética , Análise de Sequência de DNA
2.
PLoS Genet ; 17(2): e1009318, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33600407

RESUMO

The generation of lineage-specific gene expression programmes that alter proliferation capacity, metabolic profile and cell type-specific functions during differentiation from multipotent stem cells to specialised cell types is crucial for development. During differentiation gene expression programmes are dynamically modulated by a complex interplay between sequence-specific transcription factors, associated cofactors and epigenetic regulators. Here, we study U-shaped (Ush), a multi-zinc finger protein that maintains the multipotency of stem cell-like hemocyte progenitors during Drosophila hematopoiesis. Using genomewide approaches we reveal that Ush binds to promoters and enhancers and that it controls the expression of three gene classes that encode proteins relevant to stem cell-like functions and differentiation: cell cycle regulators, key metabolic enzymes and proteins conferring specific functions of differentiated hemocytes. We employ complementary biochemical approaches to characterise the molecular mechanisms of Ush-mediated gene regulation. We uncover distinct Ush isoforms one of which binds the Nucleosome Remodeling and Deacetylation (NuRD) complex using an evolutionary conserved peptide motif. Remarkably, the Ush/NuRD complex specifically contributes to the repression of lineage-specific genes but does not impact the expression of cell cycle regulators or metabolic genes. This reveals a mechanism that enables specific and concerted modulation of functionally related portions of a wider gene expression programme. Finally, we use genetic assays to demonstrate that Ush and NuRD regulate enhancer activity during hemocyte differentiation in vivo and that both cooperate to suppress the differentiation of lamellocytes, a highly specialised blood cell type. Our findings reveal that Ush coordinates proliferation, metabolism and cell type-specific activities by isoform-specific cooperation with an epigenetic regulator.


Assuntos
Ciclo Celular/genética , Proteínas de Drosophila/metabolismo , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Hematopoese/genética , Hemócitos/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Animais , Linhagem Celular , Proliferação de Células/genética , Sobrevivência Celular/genética , Sequenciamento de Cromatina por Imunoprecipitação , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Elementos Facilitadores Genéticos , Ontologia Genética , Regiões Promotoras Genéticas , Isoformas de Proteínas , Interferência de RNA , RNA-Seq , Fatores de Transcrição/genética
3.
Nucleic Acids Res ; 48(7): 3906-3921, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32025726

RESUMO

RNA interference targets aberrant transcripts with cognate small interfering RNAs, which derive from double-stranded RNA precursors. Several functional screens have identified Drosophila blanks/lump (CG10630) as a facilitator of RNAi, yet its molecular function has remained unknown. The protein carries two dsRNA binding domains (dsRBD) and blanks mutant males have a spermatogenesis defect. We demonstrate that blanks selectively boosts RNAi triggered by dsRNA of nuclear origin. Blanks binds dsRNA via its second dsRBD in vitro, shuttles between nucleus and cytoplasm and the abundance of siRNAs arising at many sites of convergent transcription is reduced in blanks mutants. Since features of nascent RNAs - such as introns and transcription beyond the polyA site - contribute to the small RNA pool, we propose that Blanks binds dsRNA formed by cognate nascent RNAs in the nucleus and fosters its export to the cytoplasm for dicing. We refer to the resulting small RNAs as blanks exported siRNAs (bepsiRNAs). While bepsiRNAs were fully dependent on RNA binding to the second dsRBD of blanks in transgenic flies, male fertility was not. This is consistent with a previous report that linked fertility to the first dsRBD of Blanks. The role of blanks in spermatogenesis appears thus unrelated to its role in dsRNA export.


Assuntos
Proteínas de Drosophila/metabolismo , Precursores de RNA/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas Argonautas/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/química , Fertilidade/genética , Sequências Repetitivas Dispersas , Masculino , Mutação , Domínios Proteicos , RNA Helicases/metabolismo , Interferência de RNA , Transporte de RNA , RNA Antissenso , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/química , Ribonuclease III/metabolismo , Transcrição Gênica
4.
Int J Obes (Lond) ; 44(5): 1185-1190, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31776435

RESUMO

BACKGROUND: Obesity is a global rising problem with epidemiological dimension. Obese parents can have programming effects on their offspring leading to obesity and associated diseases in later life. This constitutes a vicious circle. Epidemiological data and studies in rodents demonstrated differential programming effects in male and female offspring, but the timing of their developmental origin is not known. METHODS: This study investigated if sex-specific programming effects of parental obesity can already be detected in the pre-implantation period. Diet-induced obese male or female mice were mated with normal-weight partners and blastocysts were recovered. RESULTS: Gene expression profiling revealed sex-specific responses of the blastocyst transcriptome to maternal and paternal obesity. The changes in the transcriptome of male blastocysts were more pronounced than those of female blastocysts, with a stronger impact of paternal than of maternal obesity. The sperm of obese mice revealed an increased abundance of several miRNAs compared with lean mice. CONCLUSIONS: Our study indicates that sex-specific programming effects of parental obesity already start in the pre-implantation period and reveals specific alterations of the sperm miRNA profile as mechanistic link to programming effects of paternal obesity.


Assuntos
Desenvolvimento Embrionário/genética , Obesidade/genética , Transcriptoma/genética , Animais , Blastocisto/metabolismo , Feminino , Masculino , Camundongos , Camundongos Obesos , Gravidez , Regulação para Cima/genética
5.
Nucleic Acids Res ; 45(21): 12536-12550, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29040648

RESUMO

RNA interference defends against RNA viruses and retro-elements within an organism's genome. It is triggered by duplex siRNAs, of which one strand is selected to confer sequence-specificity to the RNA induced silencing complex (RISC). In Drosophila, Dicer-2 (Dcr-2) and the double-stranded RNA binding domain (dsRBD) protein R2D2 form the RISC loading complex (RLC) and select one strand of exogenous siRNAs according to the relative thermodynamic stability of base-pairing at either end. Through genome editing we demonstrate that Loqs-PD, the Drosophila homolog of human TAR RNA binding protein (TRBP) and a paralog of R2D2, forms an alternative RLC with Dcr-2 that is required for strand choice of endogenous siRNAs in S2 cells. Two canonical dsRBDs in Loqs-PD bind to siRNAs with enhanced affinity compared to miRNA/miRNA* duplexes. Structural analysis, NMR and biophysical experiments indicate that the Loqs-PD dsRBDs can slide along the RNA duplex to the ends of the siRNA. A moderate but notable binding preference for the thermodynamically more stable siRNA end by Loqs-PD alone is greatly amplified in complex with Dcr-2 to initiate strand discrimination by asymmetry sensing in the RLC.


Assuntos
Proteínas de Drosophila/metabolismo , RNA Helicases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonuclease III/metabolismo , Animais , Proteínas Argonautas/metabolismo , Células Cultivadas , Drosophila/metabolismo , Ligação Proteica , Domínios Proteicos , RNA de Cadeia Dupla/metabolismo , RNA Interferente Pequeno/química , Proteínas de Ligação a RNA/química , Termodinâmica
6.
PLoS One ; 12(6): e0180135, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28658280

RESUMO

The prokaryotic phage defense CRISPR/cas-system has developed into a versatile toolbox for genome engineering and genetic studies in many organisms. While many efforts were spent on analyzing the consequences of off-target effects, only few studies addressed side-effects that occur due to the targeted manipulation of the genome. Here, we show that the CRISPR/cas9-mediated integration of an epitope tag in combination with a selection cassette can trigger an siRNA-mediated, epigenetic genome surveillance pathway in Drosophila melanogaster cells. After homology-directed insertion of the sequence coding for the epitope tag and the selection marker, a moderate level of siRNAs covering the inserted sequence and extending into the targeted locus was detected. This response affected protein levels less than two-fold and it persisted even after single cell cloning. However, removal of the selection cassette abolished the siRNA generation, demonstrating that this response is reversible. Consistently, marker-free genome engineering did not trigger the same surveillance mechanism. These two observations indicate that the selection cassette we employed induces an aberrant transcriptional arrangement and ultimately sets off the siRNA production. There have been prior concerns about undesirable effects induced by selection markers, but fortunately we were able to show that at least one of the epigenetic changes reverts as the marker gene is excised. Although the effects observed were rather weak (less than twofold de-repression upon ago2 or dcr-2 knock-down), we recommend that when selection markers are used during genome editing, a strategy for their subsequent removal should always be included.


Assuntos
Drosophila melanogaster/genética , Edição de Genes , Regulação da Expressão Gênica/genética , Animais , Sistemas CRISPR-Cas/genética , Linhagem Celular , Epitopos/genética , Edição de Genes/métodos , Mutagênese Insercional/métodos , RNA Interferente Pequeno/genética
7.
PLoS Genet ; 13(6): e1006861, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28628606

RESUMO

DNA double-strand breaks trigger the production of locus-derived siRNAs in fruit flies, human cells and plants. At least in flies, their biogenesis depends on active transcription running towards the break. Since siRNAs derive from a double-stranded RNA precursor, a major question is how broken DNA ends can generate matching sense and antisense transcripts. We performed a genome-wide RNAi-screen in cultured Drosophila cells, which revealed that in addition to DNA repair factors, many spliceosome components are required for efficient siRNA generation. We validated this observation through site-specific DNA cleavage with CRISPR-cas9 followed by deep sequencing of small RNAs. DNA breaks in intron-less genes or upstream of a gene's first intron did not efficiently trigger siRNA production. When DNA double-strand breaks were induced downstream of an intron, however, this led to robust siRNA generation. Furthermore, a downstream break slowed down splicing of the upstream intron and a detailed analysis of siRNA coverage at the targeted locus revealed that unspliced pre-mRNA contributes the sense strand to the siRNA precursor. Since splicing factors are stimulating the response but unspliced transcripts are entering the siRNA biogenesis, the spliceosome is apparently stalled in a pre-catalytic state and serves as a signaling hub. We conclude that convergent transcription at DNA breaks is stimulated by a splicing dependent control process. The resulting double-stranded RNA is converted into siRNAs that instruct the degradation of cognate mRNAs. In addition to a potential role in DNA repair, the break-induced transcription may thus be a means to cull improper RNAs from the transcriptome of Drosophila melanogaster. Since the splicing factors identified in our screen also stimulated siRNA production from high copy transgenes, it is possible that this surveillance mechanism serves in genome defense beyond DNA double-strand breaks.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , Splicing de RNA/genética , Transcrição Gênica , Animais , Sistemas CRISPR-Cas , Drosophila melanogaster/genética , Genoma de Inseto , Íntrons/genética , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/genética
8.
Nucleic Acids Res ; 44(17): 8261-71, 2016 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-27353331

RESUMO

Small interfering RNAs (siRNAs) defend the organism against harmful transcripts from exogenous (e.g. viral) or endogenous (e.g. transposons) sources. Recent publications describe the production of siRNAs induced by DNA double-strand breaks (DSB) in Neurospora crassa, Arabidopsis thaliana, Drosophila melanogaster and human cells, which suggests a conserved function. A current hypothesis is that break-induced small RNAs ensure efficient homologous recombination (HR). However, biogenesis of siRNAs is often intertwined with other small RNA species, such as microRNAs (miRNAs), which complicates interpretation of experimental results. In Drosophila, siRNAs are produced by Dcr-2 while miRNAs are processed by Dcr-1. Thus, it is possible to probe siRNA function without miRNA deregulation. We therefore examined DNA double-strand break repair after perturbation of siRNA biogenesis in cultured Drosophila cells as well as mutant flies. Our assays comprised reporters for the single-strand annealing pathway, homologous recombination and sensitivity to the DSB-inducing drug camptothecin. We could not detect any repair defects caused by the lack of siRNAs derived from the broken DNA locus. Since production of these siRNAs depends on local transcription, they may thus participate in RNA metabolism-an established function of siRNAs-rather than DNA repair.


Assuntos
Drosophila melanogaster/genética , RNA Interferente Pequeno/metabolismo , Reparo de DNA por Recombinação , Animais , Proteínas Argonautas/metabolismo , Camptotecina/farmacologia , Cromossomos de Insetos/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efeitos dos fármacos , Edição de Genes , Técnicas de Silenciamento de Genes , Genes Reporter , Mutação/genética , Reparo de DNA por Recombinação/efeitos dos fármacos , Zigoto/metabolismo
9.
G3 (Bethesda) ; 6(8): 2467-78, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27280787

RESUMO

Traditional loss-of-function studies in Drosophila suffer from a number of shortcomings, including off-target effects in the case of RNA interference (RNAi) or the stochastic nature of mosaic clonal analysis. Here, we describe minimal in vivo GFP interference (miGFPi) as a versatile strategy to characterize gene function and to conduct highly stringent, cell type-specific loss-of-function experiments in Drosophila miGFPi combines CRISPR/Cas9-mediated tagging of genes at their endogenous locus with an immunotag and an exogenous 21 nucleotide RNAi effector sequence with the use of a single reagent, highly validated RNAi line targeting this sequence. We demonstrate the utility and time effectiveness of this method by characterizing the function of the Polymerase I (Pol I)-associated transcription factor Tif-1a, and the previously uncharacterized gene MESR4, in the Drosophila female germline stem cell lineage. In addition, we show that miGFPi serves as a powerful technique to functionally characterize individual isoforms of a gene. We exemplify this aspect of miGFPi by studying isoform-specific loss-of-function phenotypes of the longitudinals lacking (lola) gene in neural stem cells. Altogether, the miGFPi strategy constitutes a generalized loss-of-function approach that is amenable to the study of the function of all genes in the genome in a stringent and highly time effective manner.


Assuntos
Sistemas CRISPR-Cas , Proteínas de Drosophila/genética , Drosophila/genética , Interferência de RNA , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Feminino , Células Germinativas , Proteínas de Fluorescência Verde/genética , Mutação , Isoformas de Proteínas/genética , RNA Guia de Cinetoplastídeos , Proteínas Repressoras/metabolismo , Células-Tronco
10.
G3 (Bethesda) ; 6(6): 1777-85, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27172193

RESUMO

Custom genome editing has become an essential element of molecular biology. In particular, the generation of fusion constructs with epitope tags or fluorescent proteins at the genomic locus facilitates the analysis of protein expression, localization, and interaction partners at physiologic levels. Following up on our initial publication, we now describe a considerably simplified, more efficient, and readily scalable experimental workflow for PCR-based genome editing in cultured Drosophila melanogaster cells. Our analysis at the act5C locus suggests that PCR-based homology arms of 60 bp are sufficient to reach targeting efficiencies of up to 80% after selection; extension to 80 bp (PCR) or 500 bp (targeting vector) did not further improve the yield. We have expanded our targeting system to N-terminal epitope tags; this also allows the generation of cell populations with heterologous expression control of the tagged locus via the copper-inducible mtnDE promoter. We present detailed, quantitative data on editing efficiencies for several genomic loci that may serve as positive controls or benchmarks in other laboratories. While our first PCR-based editing approach offered only blasticidin-resistance for selection, we now introduce puromycin-resistance as a second, independent selection marker; it is thus possible to edit two loci (e.g., for coimmunoprecipitation) without marker removal. Finally, we describe a modified FLP recombinase expression plasmid that improves the efficiency of marker cassette FLP-out. In summary, our technique and reagents enable a flexible, robust, and cloning-free genome editing approach that can be parallelized for scale-up.


Assuntos
Drosophila melanogaster/genética , Edição de Genes , Genoma , Genômica , Animais , Sequência de Bases , Sistemas CRISPR-Cas , Linhagem Celular , Proteínas de Drosophila/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Marcação de Genes/métodos , Genes Reporter , Loci Gênicos , Vetores Genéticos/genética , Genômica/métodos , Recombinação Homóloga
11.
Methods Mol Biol ; 1173: 33-49, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24920358

RESUMO

The small RNA silencing system is an important player in the control of gene expression. In particular analysis of the highly complex sequence repertoire of piRNAs and endogenous siRNAs directed against repetitive elements and transposons has been facilitated by the advent of next-generation sequencing tools. We are providing a detailed protocol for the creation of deep sequencing libraries in combination with a chemical modification step (periodate oxidation and ß-elimination) that enriches for 3'-terminal modified small RNAs. This step can thus facilitate the detection and-since incorporation of the small RNA into the effector complex precedes the terminal methylation-determine the Argonaute-loading state of certain small RNA species. Both, the oxidation as well as the deep sequencing library preparation are adaptations of popular, previously published protocols. The sequencing libraries described here can be sequenced on all flow cell types (i.e., single-end and paired-end) of the Illumina sequencing systems and are compatible with Illumina index reads. It is therefore also possible to analyze the libraries on the MiSeq system, for which currently only paired-end flow cells are available, and combine several libraries in multiplexed experiments. The chapter concludes with considerations concerning quality control and data analysis.


Assuntos
Drosophila/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , Animais , Sequência de Bases , Oxirredução
12.
Nucleic Acids Res ; 42(11): e89, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24748663

RESUMO

The ability to edit the genome is essential for many state-of-the-art experimental paradigms. Since DNA breaks stimulate repair, they can be exploited to target site-specific integration. The clustered, regularly interspaced, short palindromic repeats (CRISPR)/cas9 system from Streptococcus pyogenes has been harnessed into an efficient and programmable nuclease for eukaryotic cells. We thus combined DNA cleavage by cas9, the generation of homologous recombination donors by polymerase chain reaction (PCR) and transient depletion of the non-homologous end joining factor lig4. Using cultured Drosophila melanogaster S2-cells and the phosphoglycerate kinase gene as a model, we reached targeted integration frequencies of up to 50% in drug-selected cell populations. Homology arms as short as 29 nt appended to the PCR primer resulted in detectable integration, slightly longer extensions are beneficial. We confirmed established rules for S. pyogenes cas9 sgRNA design and demonstrate that the complementarity region allows length variation and 5'-extensions. This enables generation of U6-promoter fusion templates by overlap-extension PCR with a standardized protocol. We present a series of PCR template vectors for C-terminal protein tagging and clonal Drosophila S2 cell lines with stable expression of a myc-tagged cas9 protein. The system can be used for epitope tagging or reporter gene knock-ins in an experimental setup that can in principle be fully automated.


Assuntos
Proteínas Associadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Desoxirribonucleases/metabolismo , Recombinação Homóloga , Reação em Cadeia da Polimerase , Animais , Linhagem Celular , Cromossomos de Insetos , Clivagem do DNA , Drosophila melanogaster/citologia , Mutação Puntual , Regiões Promotoras Genéticas , RNA Nuclear Pequeno/genética , Streptococcus pyogenes/enzimologia , Pequeno RNA não Traduzido
13.
PLoS One ; 9(1): e84994, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24454776

RESUMO

Transposable elements are a serious threat for genome integrity and their control via small RNA mediated silencing pathways is an ancient strategy. The fruit fly Drosophila melanogaster has two silencing activities that target transposons: endogenous siRNAs (esiRNAs or endo-siRNAs) and Piwi-interacting small RNAs (piRNAs). The biogenesis of endo-siRNAs involves the Dicer-2 co-factors Loqs-PD, which acts predominantly during processing of dsRNA by Dcr-2, and R2D2, which primarily helps to direct siRNAs into the RNA interference effector Ago2. Nonetheless, loss of either protein is not sufficient to produce a phenotype comparable with a dcr-2 mutation. We provide further deep sequencing evidence supporting the notion that R2D2 and Loqs-PD have partially overlapping function. Certain transposons display a preference for either dsRBD-protein during production or loading; this appeared to correlate neither with overall abundance, classification of the transposon or a specific site of genomic origin. The endo-siRNA biogenesis pathway in germline operates according to the same principles as the existing model for the soma, and its impairment does not significantly affect piRNAs. Expanding the analysis, we confirmed the occurrence of somatic piRNA-like RNAs (pilRNAs) that show a ping-pong signature. We detected expression of the Piwi-family protein mRNAs only barely above background, indicating that the somatic pilRNAs may arise from a small sub-population of somatic cells that express a functional piRNA pathway.


Assuntos
Elementos de DNA Transponíveis/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Feminino , Biblioteca Gênica , Células Germinativas/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Mutação/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
RNA Biol ; 10(6): 1042-56, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23669073

RESUMO

Development, growth and adult survival are coordinated with available metabolic resources, ascertaining that the organism responds appropriately to environmental conditions. MicroRNAs are short (21-23 nt) regulatory RNAs that confer specificity on the RNA-induced silencing complex (RISC) to inhibit a given set of mRNA targets. We profiled changes in miRNA expression during adult life in Drosophila melanogaster and determined that miR-277 is downregulated during adult life. Molecular analysis revealed that this miRNA controls branched-chain amino acid (BCAA) catabolism and as a result it can modulate the activity of the TOR kinase, a central growth regulator, in cultured cells. Metabolite analysis in cultured cells as well as flies suggests that the mechanistic basis may be an accumulation of branched-chain α-keto-acids (BCKA), rather than BCAAs, thus avoiding potentially detrimental consequences of increased branched chain amino acid levels on e.g., translational fidelity. Constitutive miR-277 expression shortens lifespan and is synthetically lethal with reduced insulin signaling, indicating that metabolic control underlies this phenotype. Transgenic inhibition with a miRNA sponge construct also shortens lifespan, in particular on protein-rich food. Thus, optimal metabolic adaptation appears to require tuning of cellular BCAA catabolism by miR-277.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Drosophila melanogaster/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Envelhecimento , Animais , Animais Geneticamente Modificados , Células Cultivadas , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Insulina/metabolismo , Longevidade , Análise de Sequência de RNA
15.
Genetics ; 192(4): 1543-52, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23051640

RESUMO

Animal microRNAs (miRNA) are implicated in the control of nearly all cellular functions. Due to high sequence redundancy within the miRNA gene pool, loss of most of these 21- to 24-bp long RNAs individually does not cause a phenotype. Thus, only very few miRNAs have been associated with clear functional roles. We constructed a transgenic UAS-miRNA library in Drosophila melanogaster that contains 180 fly miRNAs. This library circumvents the redundancy issues by facilitating the controlled misexpression of individual miRNAs and is a useful tool to complement loss-of-function approaches. Demonstrating the effectiveness of our library, 78 miRNAs induced clear phenotypes. Most of these miRNAs were previously unstudied. Furthermore, we present a simple system to create GFP sensors to monitor miRNA expression and test direct functional interactions in vivo. Finally, we focus on the miR-92 family and identify a direct target gene that is responsible for the specific wing phenotype induced by the misexpression of miR-92 family members.


Assuntos
Drosophila melanogaster/genética , Biblioteca Gênica , MicroRNAs , Animais , Animais Geneticamente Modificados , Clonagem Molecular , Proteínas de Drosophila/genética , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Proteínas de Fluorescência Verde/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas dos Microfilamentos/genética , Fenótipo , Asas de Animais/fisiologia
16.
Nucleic Acids Res ; 40(19): 9596-603, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22848104

RESUMO

Small RNAs have been implicated in numerous cellular processes, including effects on chromatin structure and the repression of transposons. We describe the generation of a small RNA response at DNA ends in Drosophila that is analogous to the recently reported double-strand break (DSB)-induced RNAs or Dicer- and Drosha-dependent small RNAs in Arabidopsis and vertebrates. Active transcription in the vicinity of the break amplifies this small RNA response, demonstrating that the normal messenger RNA contributes to the endogenous small interfering RNAs precursor. The double-stranded RNA precursor forms with an antisense transcript that initiates at the DNA break. Breaks are thus sites of transcription initiation, a novel aspect of the cellular DSB response. This response is specific to a double-strand break since nicked DNA structures do not trigger small RNA production. The small RNAs are generated independently of the exact end structure (blunt, 3'- or 5'-overhang), can repress homologous sequences in trans and may therefore--in addition to putative roles in repair--exert a quality control function by clearing potentially truncated messages from genes in the vicinity of the break.


Assuntos
Quebras de DNA de Cadeia Dupla , Drosophila/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Animais , Células Cultivadas , DNA/metabolismo , Quebras de DNA de Cadeia Simples , Reparo do DNA , RNA Interferente Pequeno/biossíntese , Transcrição Gênica
17.
RNA Biol ; 9(5): 633-43, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22614836

RESUMO

Pur-α was identified as a DNA-binding protein with high affinity for the single-stranded PUR-motif (GGN)n. Bound to DNA, Pur-α can both activate and repress transcription. In addition, Pur-α binds to RNA and may participate in nuclear RNA export as well as transport of cytoplasmic neuronal mRNP granules. The heritable trinucleotide-repeat expansion disease Fragile X associated Tremor and Ataxia Syndrome (FXTAS) leads to interaction of Pur-α with mutant, abnormally long r(CGG)n stretches, which appears to titrate the protein away from its physiologic mRNA targets into nuclear RNA-protein aggregates. We examined the function of Drosophila Pur-α and demonstrate that the protein accumulates in the growing oocyte early in oogenesis. Co-purifying proteins reveal that Pur-α is part of transported mRNP complexes, analogous to its reported role in nerve cells. We analyzed the subcellular localization of mutant GFP-Pur-α fusion proteins where either nucleic acid binding or dimerization, or both, were prevented. We propose that association with mRNAs occurs in the nucleus and is required for nuclear export of the complex. Furthermore, efficient translocation into the oocyte also requires RNA binding as well as dimerization. RNA binding assays demonstrate that recombinant Drosophila Pur-α can bind r(CGG) 4 with higher affinity than previously thought. Related sequences, such as r(CAG) 4 and the consensus sequence of the opa-repeat r(CAG) 3CAA, can also associate with Pur-α in vitro and in vivo. The mRNA target spectrum of Pur-α may therefore be larger than previously anticipated.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Oócitos/metabolismo , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo , Repetições de Trinucleotídeos , Transporte Ativo do Núcleo Celular , Animais , Sequência de Bases , Linhagem Celular , Polaridade Celular , Sequência Consenso , Drosophila melanogaster/citologia , Epitélio/metabolismo , Feminino , Oogênese , Folículo Ovariano/citologia , Ligação Proteica , Ribonucleoproteínas/metabolismo
18.
J Gen Virol ; 93(Pt 7): 1537-1547, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22442111

RESUMO

One goal of virus infection is to reprogramme the host cell to optimize virus replication. As part of this process, viral microRNAs (miRNAs) may compete for components of the miRNA/small interfering RNA pathway, as well as regulate cellular targets. Murine cytomegalovirus (MCMV) has been described to generate large numbers of viral miRNAs during lytic infection and was therefore used to analyse the impact of viral miRNAs on the host-cell small-RNA system, as well as to check for sorting of viral small RNAs into specific Argonaute (Ago) proteins. Deep-sequencing analysis of MCMV-infected cells revealed that viral miRNAs represented only ~13% of all detected miRNAs. All previously described MCMV miRNAs with the exception of miR-m88-1* were confirmed, and for the MCMV miR-m01-1 hairpin, an additional miRNA, designated miR-m01-1-3p, was found. Its presence was confirmed by quantitative real-time PCR and Northern blotting. Deep sequencing after RNA-induced silencing complex (RISC) immunoprecipitation with antibodies specific for either Ago1 or Ago2 showed that all MCMV miRNAs were loaded into both RISCs. The ratio of MCMV to mouse miRNAs was not increased after immunoprecipitation of Ago proteins. Viral miRNAs therefore did not overwhelm the host miRNA processing system, nor were they incorporated preferentially into RISCs. Three mouse miRNAs were found that showed altered expression as a result of MCMV infection. Downregulation of miR-27a, as described previously, could be confirmed. In addition, miR-26a was downregulated, and upregulation of miR-7a dependent on viral protein expression could be observed.


Assuntos
Expressão Gênica , Interações Hospedeiro-Patógeno , MicroRNAs/biossíntese , Muromegalovirus/crescimento & desenvolvimento , Animais , Northern Blotting , Células Cultivadas , Imunoprecipitação da Cromatina , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase em Tempo Real
19.
PLoS One ; 7(1): e28907, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22253700

RESUMO

AU-rich elements (AREs) are regulatory sequences located in the 3' untranslated region of many short-lived mRNAs. AREs are recognized by ARE-binding proteins and cause rapid mRNA degradation. Recent reports claimed that the function of AREs may be--at least in part--relayed through the miRNA pathway. We have revisited this hypothesis using dicer knock-out mouse embryonic fibroblasts and cultured Drosophila cells. In contrast to the published results, we find no evidence for a general requirement of the miRNA pathway in the function of AREs. Endogenous ier3 mRNA, which is known to contain a functional ARE, was degraded rapidly at indistinguishable rates in wild type and dicer knock-out mouse embryonic fibroblasts. In cultured Drosophila cells, both ARE-containing GFP reporter mRNAs and the endogenous cecA1 mRNA were resistant to depletion of the mi/siRNA factors dcr-1, dcr-2, ago1 and ago2. Furthermore, the Drosophila miRNA originally proposed to recognize AU-rich elements, miR-289, is not detectably expressed in flies or cultured S2 cells. Even our attempts to overexpress this miRNA from its genomic hairpin sequence failed. Thus, this sequence cannot serve as link between the miRNA and the AU-rich element mediated silencing pathways. Taken together, our studies in mammalian and Drosophila cells strongly argue that AREs can function independently of miRNAs.


Assuntos
Drosophila melanogaster/genética , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , MicroRNAs/genética , Estabilidade de RNA/genética , Sequências Reguladoras de Ácido Ribonucleico/genética , Animais , Sequência de Bases , Linhagem Celular , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/metabolismo , Drosophila melanogaster/citologia , Fibroblastos/enzimologia , Genes Reporter/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , MicroRNAs/metabolismo , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonuclease III/deficiência , Ribonuclease III/metabolismo
20.
Nucleic Acids Res ; 39(9): 3836-51, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21245036

RESUMO

In Drosophila, siRNAs are classified as endo- or exo-siRNAs based on their origin. Both are processed from double-stranded RNA precursors by Dcr-2 and then loaded into the Argonaute protein Ago2. While exo-siRNAs serve to defend the cell against viruses, endo-siRNAs restrict the spread of selfish DNA in somatic cells, analogous to piRNAs in the germ line. Endo- and exo-siRNAs display a differential requirement for double-stranded RNA binding domain proteins (dsRBPs): R2D2 is needed to load exo-siRNAs into Ago2 while the PD isoform of Loquacious (Loqs-PD) stimulates Dcr-2 during the nucleolytic processing of hairpin-derived endo-siRNAs. In cell culture assays, R2D2 antagonizes Loqs-PD in endo-siRNA silencing and Loqs-PD is an inhibitor of RNA interference. Loqs-PD can interact via the C-terminus unique to this isoform with the DExH/D-helicase domain of Drosophila Dcr-2, where binding of R2D2 has also been localized. Separation of the two pathways is not complete; rather, the dicing and Ago2-loading steps appear uncoupled, analogous to the corresponding steps in miRNA biogenesis. Analysis of deep sequencing data further demonstrates that in r2d2 mutant flies, siRNAs can be loaded into Ago2 but not all siRNA classes are equally proficient for this. Thus, the canonical Ago2-RISC loading complex can be bypassed under certain circumstances.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Complexo de Inativação Induzido por RNA/metabolismo , Animais , Proteínas Argonautas , Drosophila/metabolismo , Proteínas de Drosophila/química , Isoformas de Proteínas/metabolismo , Multimerização Proteica , Estrutura Terciária de Proteína , RNA Helicases/química , RNA Helicases/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/química , Ribonuclease III/química , Ribonuclease III/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA