Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Genome Biol ; 24(1): 63, 2023 04 03.
Article in English | MEDLINE | ID: mdl-37013657

ABSTRACT

Transposable Element MOnitoring with LOng-reads (TrEMOLO) is a new software that combines assembly- and mapping-based approaches to robustly detect genetic elements called transposable elements (TEs). Using high- or low-quality genome assemblies, TrEMOLO can detect most TE insertions and deletions and estimate their allele frequency in populations. Benchmarking with simulated data revealed that TrEMOLO outperforms other state-of-the-art computational tools. TE detection and frequency estimation by TrEMOLO were validated using simulated and experimental datasets. Therefore, TrEMOLO is a comprehensive and suitable tool to accurately study TE dynamics. TrEMOLO is available under GNU GPL3.0 at https://github.com/DrosophilaGenomeEvolution/TrEMOLO .


Subject(s)
DNA Transposable Elements , Software , Gene Frequency , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA
2.
Cells ; 9(8)2020 07 25.
Article in English | MEDLINE | ID: mdl-32722451

ABSTRACT

Transposable elements (TEs) are the main components of genomes. However, due to their repetitive nature, they are very difficult to study using data obtained with short-read sequencing technologies. Here, we describe an efficient pipeline to accurately recover TE insertion (TEI) sites and sequences from long reads obtained by Oxford Nanopore Technology (ONT) sequencing. With this pipeline, we could precisely describe the landscapes of the most recent TEIs in wild-type strains of Drosophila melanogaster and Drosophila simulans. Their comparison suggests that this subset of TE sequences is more similar than previously thought in these two species. The chromosome assemblies obtained using this pipeline also allowed recovering piRNA cluster sequences, which was impossible using short-read sequencing. Finally, we used our pipeline to analyze ONT sequencing data from a D. melanogaster unstable line in which LTR transposition was derepressed for 73 successive generations. We could rely on single reads to identify new insertions with intact target site duplications. Moreover, the detailed analysis of TEIs in the wild-type strains and the unstable line did not support the trap model claiming that piRNA clusters are hotspots of TE insertions.


Subject(s)
DNA Transposable Elements/immunology , Drosophila melanogaster/immunology , Drosophila/immunology , Nanopores , Animals
3.
Nat Commun ; 11(1): 2818, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32499524

ABSTRACT

In eukaryotes, trimethylation of lysine 9 on histone H3 (H3K9) is associated with transcriptional silencing of transposable elements (TEs). In drosophila ovaries, this heterochromatic repressive mark is thought to be deposited by SetDB1 on TE genomic loci after the initial recognition of nascent transcripts by PIWI-interacting RNAs (piRNAs) loaded on the Piwi protein. Here, we show that the nucleosome remodeler Mi-2, in complex with its partner MEP-1, forms a subunit that is transiently associated, in a MEP-1 C-terminus-dependent manner, with known Piwi interactors, including a recently reported SUMO ligase, Su(var)2-10. Together with the histone deacetylase Rpd3, this module is involved in the piRNA-dependent TE silencing, correlated with H3K9 deacetylation and trimethylation. Therefore, drosophila piRNA-mediated transcriptional silencing involves three epigenetic effectors, a remodeler, Mi-2, an eraser, Rpd3 and a writer, SetDB1, in addition to the Su(var)2-10 SUMO ligase.


Subject(s)
Adenosine Triphosphatases/metabolism , Autoantigens/metabolism , Drosophila Proteins/metabolism , Heterochromatin/chemistry , Histone Deacetylase 1/metabolism , Nucleosomes/metabolism , RNA, Small Interfering/metabolism , Animals , Argonaute Proteins/metabolism , Drosophila melanogaster , Epigenesis, Genetic , Female , Gene Expression Regulation , Gene Silencing , Histones/chemistry , Ovary/metabolism , Protein Inhibitors of Activated STAT
4.
Nucleic Acids Res ; 46(18): 9524-9536, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30312469

ABSTRACT

Transposable elements (TEs) are parasitic DNA sequences that threaten genome integrity by replicative transposition in host gonads. The Piwi-interacting RNAs (piRNAs) pathway is assumed to maintain Drosophila genome homeostasis by downregulating transcriptional and post-transcriptional TE expression in the ovary. However, the bursts of transposition that are expected to follow transposome derepression after piRNA pathway impairment have not yet been reported. Here, we show, at a genome-wide level, that piRNA loss in the ovarian somatic cells boosts several families of the endogenous retroviral subclass of TEs, at various steps of their replication cycle, from somatic transcription to germinal genome invasion. For some of these TEs, the derepression caused by the loss of piRNAs is backed up by another small RNA pathway (siRNAs) operating in somatic tissues at the post transcriptional level. Derepressed transposition during 70 successive generations of piRNA loss exponentially increases the genomic copy number by up to 10-fold.


Subject(s)
DNA Transposable Elements/genetics , Drosophila melanogaster/genetics , Germ Cells/metabolism , Ovary/metabolism , RNA, Small Interfering/genetics , Aneuploidy , Animals , Drosophila melanogaster/cytology , Female , Gene Silencing , Genome, Insect/genetics , Germ Cells/cytology , Ovary/cytology , Signal Transduction/genetics
5.
Genome Biol Evol ; 9(6): 1450-1470, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28854624

ABSTRACT

Interspecific hybridization is a genomic stress condition that leads to the activation of transposable elements (TEs) in both animals and plants. In hybrids between Drosophila buzzatii and Drosophila koepferae, mobilization of at least 28 TEs has been described. However, the molecular mechanisms underlying this TE release remain poorly understood. To give insight on the causes of this TE activation, we performed a TE transcriptomic analysis in ovaries (notorious for playing a major role in TE silencing) of parental species and their F1 and backcrossed (BC) hybrids. We find that 15.2% and 10.6% of the expressed TEs are deregulated in F1 and BC1 ovaries, respectively, with a bias toward overexpression in both cases. Although differences between parental piRNA (Piwi-interacting RNA) populations explain only partially these results, we demonstrate that piRNA pathway proteins have divergent sequences and are differentially expressed between parental species. Thus, a functional divergence of the piRNA pathway between parental species, together with some differences between their piRNA pools, might be at the origin of hybrid instabilities and ultimately cause TE misregulation in ovaries. These analyses were complemented with the study of F1 testes, where TEs tend to be less expressed than in D. buzzatii. This can be explained by an increase in piRNA production, which probably acts as a defence mechanism against TE instability in the male germline. Hence, we describe a differential impact of interspecific hybridization in testes and ovaries, which reveals that TE expression and regulation are sex-biased.


Subject(s)
DNA Transposable Elements , Drosophila/genetics , Evolution, Molecular , RNA, Small Interfering/genetics , Animals , Drosophila/classification , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Hybridization, Genetic , Male , Ovary/metabolism , Phylogeny , RNA, Small Interfering/metabolism
6.
Mol Cell ; 66(3): 411-419.e4, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28457744

ABSTRACT

Most piRNAs in the Drosophila female germline are transcribed from heterochromatic regions called dual-strand piRNA clusters. Histone 3 lysine 9 trimethylation (H3K9me3) is required for licensing piRNA production by these clusters. However, it is unclear when and how they acquire this permissive heterochromatic state. Here, we show that transient Piwi depletion in Drosophila embryos results in H3K9me3 decrease at piRNA clusters in ovaries. This is accompanied by impaired biogenesis of ovarian piRNAs, accumulation of transposable element transcripts, and female sterility. Conversely, Piwi depletion at later developmental stages does not disturb piRNA cluster licensing. These results indicate that the identity of piRNA clusters is epigenetically acquired in a Piwi-dependent manner during embryonic development, which is reminiscent of the widespread genome reprogramming occurring during early mammalian zygotic development.


Subject(s)
Argonaute Proteins/metabolism , DNA Methylation , DNA Transposable Elements , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epigenetic Repression , Heterochromatin/metabolism , Ovary/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Age Factors , Animals , Argonaute Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Female , Fertility , Gene Expression Regulation, Developmental , Heterochromatin/genetics , Histones/metabolism , Infertility, Female/genetics , Infertility, Female/metabolism , Infertility, Female/physiopathology , Methylation , Morphogenesis , Ovary/embryology , Protein Binding , RNA, Small Interfering/genetics
7.
Sci Rep ; 7: 40618, 2017 01 16.
Article in English | MEDLINE | ID: mdl-28091568

ABSTRACT

Crosses between close species can lead to genomic disorders, often considered to be the cause of hybrid incompatibility, one of the initial steps in the speciation process. How these incompatibilities are established and what are their causes remain unclear. To understand the initiation of hybrid incompatibility, we performed reciprocal crosses between two species of Drosophila (D. mojavensis and D. arizonae) that diverged less than 1 Mya. We performed a genome-wide transcriptomic analysis on ovaries from parental lines and on hybrids from reciprocal crosses. Using an innovative procedure of co-assembling transcriptomes, we show that parental lines differ in the expression of their genes and transposable elements. Reciprocal hybrids presented specific gene categories and few transposable element families misexpressed relative to the parental lines. Because TEs are mainly silenced by piwi-interacting RNAs (piRNAs), we hypothesize that in hybrids the deregulation of specific TE families is due to the absence of such small RNAs. Small RNA sequencing confirmed our hypothesis and we therefore propose that TEs can indeed be major players of genome differentiation and be implicated in the first steps of genomic incompatibilities through small RNA regulation.


Subject(s)
DNA Transposable Elements/genetics , Drosophila/genetics , Gene Expression Regulation , Hybridization, Genetic , Animals , Conserved Sequence/genetics , Female , Gene Ontology , Genes, Insect , Geography , Inheritance Patterns/genetics , Male , Mexico , RNA, Small Interfering/metabolism , Species Specificity , Transcriptome/genetics , United States
8.
PLoS Genet ; 11(5): e1005194, 2015 May.
Article in English | MEDLINE | ID: mdl-25993106

ABSTRACT

RNA interference-related silencing mechanisms concern very diverse and distinct biological processes, from gene regulation (via the microRNA pathway) to defense against molecular parasites (through the small interfering RNA and the Piwi-interacting RNA pathways). Small non-coding RNAs serve as specificity factors that guide effector proteins to ribonucleic acid targets via base-pairing interactions, to achieve transcriptional or post-transcriptional regulation. Because of the small sequence complementarity required for microRNA-dependent post-transcriptional regulation, thousands of microRNA (miRNA) putative targets have been annotated in Drosophila. In Drosophila somatic ovarian cells, genomic parasites, such as transposable elements (TEs), are transcriptionally repressed by chromatin changes induced by Piwi-interacting RNAs (piRNAs) that prevent them from invading the germinal genome. Here we show, for the first time, that a functional miRNA pathway is required for the piRNA-mediated transcriptional silencing of TEs in this tissue. Global miRNA depletion, caused by tissue- and stage-specific knock down of drosha (involved in miRNA biogenesis), AGO1 or gawky (both responsible for miRNA activity), resulted in loss of TE-derived piRNAs and chromatin-mediated transcriptional de-silencing of TEs. This specific TE de-repression was also observed upon individual titration (by expression of the complementary miRNA sponge) of two miRNAs (miR-14 and miR-34) as well as in a miR-14 loss-of-function mutant background. Interestingly, the miRNA defects differentially affected TE- and 3' UTR-derived piRNAs. To our knowledge, this is the first indication of possible differences in the biogenesis or stability of TE- and 3' UTR-derived piRNAs. This work is one of the examples of detectable phenotypes caused by loss of individual miRNAs in Drosophila and the first genetic evidence that miRNAs have a role in the maintenance of genome stability via piRNA-mediated TE repression.


Subject(s)
DNA Transposable Elements , Drosophila Proteins/metabolism , Drosophila/genetics , MicroRNAs/metabolism , Ovarian Follicle/metabolism , RNA Interference , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Female , Gene Expression Regulation , Gene Silencing , MicroRNAs/genetics , Ovarian Follicle/cytology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
9.
Methods ; 67(1): 91-101, 2014 May 01.
Article in English | MEDLINE | ID: mdl-23727218

ABSTRACT

The discovery of the small regulatory RNAs has changed our vision of cellular regulations. Indeed, when loaded on Argonaute proteins they form ribonucleoprotein complexes (RNPs) that target complementary sequences to achieve widespread silencing mechanisms conserved in most eukaryotes. The recent development of deep sequencing approaches highly contributed to their detection. Small RNA isolation from cells and/or tissues remains a crucial stage to generate robust and relevant sequencing data. In 2006, a novel strategy based on anion-exchange chromatography has been proposed as an alternative to the standard size-isolation purification procedure. Using bioinformatic comparative analysis, we here demonstrate that anion-exchange chromatographic RNP purification prior to small RNA extraction unbiasedly enriches datasets in bona fide reads (small regulatory RNA sequences) and depletes endogenous contaminants (ribosomal RNAs and degradation RNA products). The resulting increase in sequencing depth provides a major benefit to study rare populations. We then developed a fast and basic manual procedure to purify such small non-coding RNAs using anion-exchange chromatography at the bench. We validated the efficiency of this new method and used this strategy to purify small RNAs from various tissues and organisms. We moreover determined that our manual purification increases the output of the previously described anion-exchange chromatography procedure.


Subject(s)
RNA, Small Untranslated/isolation & purification , Animals , Chromatography, Ion Exchange , Drosophila/genetics , Female , Genes, Insect , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Ovary/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Sequence Analysis, RNA , Testis/metabolism
10.
Genome Res ; 22(10): 1877-88, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22555593

ABSTRACT

The maintenance of genome integrity is an essential trait to the successful transmission of genetic information. In animal germ cells, piRNAs guide PIWI proteins to silence transposable elements (TEs) in order to maintain genome integrity. In insects, most TE silencing in the germline is achieved by secondary piRNAs that are produced by a feed-forward loop (the ping-pong cycle), which requires the piRNA-directed cleavage of two types of RNAs: mRNAs of functional euchromatic TEs and heterochromatic transcripts that contain defective TE sequences. The first cleavage that initiates such an amplification loop remains poorly understood. Taking advantage of the existence of strains that are devoid of functional copies of the LINE-like I-element, we report here that in such Drosophila ovaries, the initiation of a ping-pong cycle is exclusively achieved by secondary I-element piRNAs that are produced in the ovary and deposited in the embryonic germline. This unusual secondary piRNA biogenesis, detected in the absence of functional I-element copies, results from the processing of sense and antisense transcripts of several different defective I-element. Once acquired, for instance after ancestor aging, this capacity to produce heterochromatic-only secondary piRNAs is partially transmitted through generations via maternal piRNAs. Furthermore, such piRNAs acting as ping-pong initiators in a chromatin-independent manner confer to the progeny a high capacity to repress the I-element mobility. Our study explains, at the molecular level, the basis for epigenetic memory of maternal immunity that protects females from hybrid dysgenesis caused by transposition of paternally inherited functional I-element.


Subject(s)
DNA Transposable Elements , Drosophila/genetics , Quantitative Trait, Heritable , RNA, Small Interfering/genetics , Aging/genetics , Animals , Chromatin , Female , Gene Silencing , Male , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Small Interfering/metabolism , Transcription, Genetic
11.
Development ; 138(11): 2315-23, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21558379

ABSTRACT

Homeodomain transcription factors classically exert their morphogenetic activities through the cell-autonomous regulation of developmental programs. In vertebrates, several homeoproteins have also been shown to have direct non-cell-autonomous activities in the developing nervous system. We present the first in vivo evidence for homeoprotein signaling in Drosophila. Focusing on wing development as a model, we first demonstrate that the homeoprotein Engrailed (En) is secreted. Using single-chain anti-En antibodies expressed under the control of a variety of promoters, we delineate the wing territories in which secreted En acts. We show that En is a short-range signaling molecule that participates in anterior crossvein development, interacting with the Dpp signaling pathway. This report thus suggests that direct signaling with homeoproteins is an evolutionarily conserved phenomenon that is not restricted to neural tissues and involves interactions with bona fide signal transduction pathways.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/embryology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Wings, Animal/embryology , Animals , Animals, Genetically Modified , Antibodies , Drosophila/genetics , Drosophila/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Homeodomain Proteins/immunology , Humans , Signal Transduction , Transcription Factors/immunology , Wings, Animal/metabolism
12.
Hum Mol Genet ; 17(22): 3601-16, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18718937

ABSTRACT

Huntington's disease (HD) is caused by the expansion of the polyglutamine (polyQ) tract in the human Huntingtin (hHtt) protein (polyQ-hHtt). Although this mutation behaves dominantly, htt loss of function may also contribute to HD pathogenesis. Using a Drosophila model of HD, we found that Engrailed (EN), a transcriptional activator of endogenous Drosophila htt (dhtt), is able to prevent aggregation of polyQ-hHtt. To interpret these findings, we tested and identified a protective role of N-terminal fragments of both Drosophila and Human wild-type Htt onto polyQ-hHtt-induced cellular defects. In addition, N-terminal parts of normal hHtt were also able to rescue eye degeneration due to the loss of Drosophila endogenous dhtt function. Thus, our data indicate that Drosophila and Human Htt share biological properties, and confirm a model whereby EN activates endogenous dhtt, which in turn prevents polyQ-hHtt-induced phenotypes. The protective role of wild-type hHtt N-terminal parts, specifically onto polyQ-hHtt-induced cellular toxicity suggests that the HD may be considered as a dominant negative disease rather than solely dominant.


Subject(s)
Homeodomain Proteins/physiology , Huntington Disease/metabolism , Serotonin Plasma Membrane Transport Proteins , Transcription Factors/physiology , Animals , Animals, Genetically Modified , Chromatin Immunoprecipitation , Chromosome Mapping , Drosophila Proteins , Eye/pathology , Homeodomain Proteins/genetics , Humans , Models, Animal , Peptides/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Transcription Factors/genetics
13.
Dev Biol ; 301(2): 542-54, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17126316

ABSTRACT

In Drosophila, the ventral nerve cord (VNC) architecture is built from neuroblasts that are specified during embryonic development, mainly by transcription factors. Here we show that Engrailed, a homeodomain transcription factor known to be involved in the establishment of neuroblast identity, is also directly implicated in the regulation of axonal guidance cues. Posterior commissures (PC) are missing in engrailed mutant embryos, and axonal pathfinding defects are observed when Engrailed is ectopically expressed at early stages, prior to neuronal specification. We also show that frazzled, enabled, and trio, all of which are potential direct targets of Engrailed and are involved in axonal navigation, interact genetically with engrailed to form posterior commissures in the developing VNC. The regulation of frazzled expression in engrailed-expressing neuroblasts contributes significantly to the formation of the posterior commissures by acting on axon growth. Finally, we identified a small genomic fragment within intron 1 of frazzled that can mediate activation by Engrailed in vivo when fused to a GFP reporter. These results indicate that Engrailed's function during the segregation of the neuroblasts is crucial for regulating different actors that are later involved in axon guidance.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Neurons/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism , Alleles , Animals , Cell Differentiation , Chromosomes/genetics , Drosophila Proteins , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Homeodomain Proteins/genetics , Mutation/genetics , Netrin Receptors , Neurons/cytology , Phenotype , Receptors, Cell Surface/genetics , Time Factors , Transcription Factors/genetics
14.
RNA ; 9(3): 299-308, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12592004

ABSTRACT

RNA interference (RNAi) designates the multistep process by which double-stranded RNA induces the silencing of homologous endogenous genes. Some aspects of RNAi appear to be conserved throughout evolution, including the processing of trigger dsRNAs into small 21-23-bp siRNAs and their use to guide the degradation of complementary mRNAs. Two remarkable features of RNAi were uncovered in plants and Caenorhabditid elegans. First, RNA-dependent RNA polymerase activities allow the synthesis of siRNA complementary to sequences upstream of or downstream from the initial trigger region in the target mRNA, leading to a transitive RNAi with sequences that had not been initially targeted. Secondly, systemic RNAi may cause the targeting of gene silencing in one tissue to spread to other tissues. Using transgenes expressing dsRNA, we investigated whether transitive and systemic RNAi occur in Drosophila. DsRNA-producing transgenes targeted RNAi to specific regions of alternative mRNA species of one gene without transitive effect directed to sequences downstream from or upstream of the initial trigger region. Moreover, specific expression of a dsRNA, using either cell-specific GAL4 drivers or random clonal activation of a GAL4 driver, mediated a cell-autonomous RNAi. Together, our results provide evidence that transitive and systemic aspects of RNAi are not conserved in Drosophila and demonstrate that dsRNA-producing transgenes allow powerful reverse genetic approaches to be conducted in this model organism, by knocking down gene functions at the resolution of a single-cell type and of a single isoform.


Subject(s)
Drosophila/genetics , RNA Interference , Animals , Animals, Genetically Modified , DNA Probes , Drosophila Proteins/genetics , Gene Expression Regulation , Green Fluorescent Proteins , Luminescent Proteins/genetics , Microscopy, Confocal , Protein Isoforms , Receptors, Steroid/genetics , Repetitive Sequences, Nucleic Acid , Transcription Factors/genetics , Transgenes
15.
Development ; 130(7): 1243-54, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12588842

ABSTRACT

Chromatin immunoprecipitation after UV crosslinking of DNA/protein interactions was used to construct a library enriched in genomic sequences that bind to the Engrailed transcription factor in Drosophila embryos. Sequencing of the clones led to the identification of 203 Engrailed-binding fragments localized in intergenic or intronic regions. Genes lying near these fragments, which are considered as potential Engrailed target genes, are involved in different developmental pathways, such as anteroposterior patterning, muscle development, tracheal pathfinding or axon guidance. We validated this approach by in vitro and in vivo tests performed on a subset of Engrailed potential targets involved in these various pathways. Finally, we present strong evidence showing that an immunoprecipitated genomic DNA fragment corresponds to a promoter region involved in the direct regulation of frizzled2 expression by engrailed in vivo.


Subject(s)
DNA/metabolism , Drosophila/genetics , Drosophila/metabolism , Homeodomain Proteins/metabolism , Transcription Factors , Animals , Electrophoretic Mobility Shift Assay , Gene Expression Regulation , Larva/metabolism , Receptors, Cell Surface/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...