The Interactive Fly

Transposons

  • Transposable elements in Drosophila
  • Mechanisms of LTR-Retroelement Transposition: Lessons from Drosophila melanogaster
  • Active retrotransposons help maintain pericentromeric heterochromatin required for faithful cell division
  • Unique transposon landscapes are pervasive across Drosophila melanogaster genomes
  • Reconstructing the Invasion Route of the P-Element in Drosophila melanogaster Using Extant Population Samples
  • Conserved noncoding elements influence the transposable element landscape in Drosophila
  • Extensive exchange of transposable elements in the Drosophila pseudoobscura group
  • Stress affects the epigenetic marks added by natural transposable element insertions in Drosophila melanogaster
  • Epigenetic and chromosomal features drive transposon insertion in Drosophila melanogaster
  • Phenotypic and Transcriptomic Responses to Stress Differ According to Population Geography in an Invasive Species
  • A robust transposon-endogenizing response from germline stem cells
  • Population specific dynamics and selection patterns of transposable element insertions in European natural populations
  • SQuIRE reveals locus-specific regulation of interspersed repeat expression
  • Dynamic Interactions between the genome and an endogenous retrovirus: Tirant in Drosophila simulans wild-type strains
  • Stress response, behavior, and development are shaped by transposable element-induced mutations in Drosophila
  • Transposable element landscape in Drosophila populations selected for longevity
  • Diverse families of transposable elements affect the transcriptional regulation of stress-response genes in Drosophila melanogaster
  • The Hsp70 chaperone is a major player in stress-induced transposable element activation
  • Patterns of piRNA Regulation in Drosophila Revealed through Transposable Element Clade Inference
  • High frequency of horizontal transfer in Jockey families (LINE order) of drosophilids
  • Special vulnerability of somatic niche cells to transposable element activation in Drosophila larval ovaries
  • Unraveling the features of somatic transposition in the Drosophila intestine
  • Transposable elements contribute to the genomic response to insecticides in Drosophila melanogaster
  • A Transposon Story: From TE Content to TE Dynamic Invasion of Drosophila Genomes Using the Single-Molecule Sequencing Technology from Oxford Nanopore
  • A Forward Genetic Approach to Mapping a P-Element Second Site Mutation Identifies DCP2 as a Novel Tumour Suppressor in Drosophila melanogaster
  • Epigenetic conflict on a degenerating Y chromosome increases mutational burden in Drosophila males
  • Toxic Y chromosome: Increased repeat expression and age-associated heterochromatin loss in male Drosophila with a young Y chromosome
  • The prolyl-isomerase PIN1 is essential for nuclear Lamin-B structure and function and protects heterochromatin under mechanical stress
  • Evidence of the Physical Interaction between Rpl22 and the Transposable Element Doc5, a Heterochromatic Transposon of Drosophila melanogaster
  • Transposon expression in the Drosophila brain is driven by neighboring genes and diversifies the neural transcriptome
  • Involvement of DNA Repair Genes and System of Radiation-Induced Activation of Transposons in Formation of Transgenerational Effects
  • piRNA-mediated gene regulation and adaptation to sex-specific transposon expression in D. melanogaster male germline
  • Infections by Transovarially Transmitted DMelSV in Drosophila Have No Impact on Ovarian Transposable Element Transcripts but Increase Their Amounts in the Soma
  • Regulatory regions in natural transposable element insertions drive interindividual differences in response to immune challenges in Drosophila
  • Retrotransposon activation during Drosophila metamorphosis conditions adult antiviral responses
  • Impaired function of rDNA transcription initiation machinery leads to derepression of ribosomal genes with insertions of R2 retrotransposon
  • Histone H3.3 K27M and K36M mutations de-repress transposable elements through perturbation of antagonistic chromatin marks
  • Transposable element profiles reveal cell line identity and loss of heterozygosity in Drosophila cell culture
  • Transposable element landscapes in aging Drosophila
  • Expression of retrotransposons contributes to aging in Drosophila Genetics
  • The effects of transpositions of functional I retrotransposons depend on the conditions and dose of parental exposure
  • Contribution of transposable elements to transgenerational effects of chronic radioactive exposure of natural populations of Drosophila melanogaster living for a long time in the zone of the Chernobyl nuclear disaster
  • Population-scale long-read sequencing uncovers transposable elements associated with gene expression variation and adaptive signatures in Drosophila

    Flamenco Transposon
  • Production of small non-coding RNAs from the flamenco locus is regulated by the gypsy retrotransposon of Drosophila melanogaster
  • Distribution, evolution, and diversity of retrotransposons at the flamenco locus reflect the regulatory properties of piRNA cluster
  • Retrotransposons Down- and Up-Regulation in Aging Somatic Tissues

    Gypsy Transposon
  • In between: gypsy in Drosophila melanogaster reveals new insights into endogenous retrovirus evolution.
  • The centrosomal protein CP190 is a component of the gypsy chromatin insulator
  • Production of small non-coding RNAs from the flamenco locus is regulated by the gypsy retrotransposon of Drosophila melanogaster
  • Cellular labeling of endogenous retrovirus replication (CLEVR) reveals de novo insertions of the gypsy retrotransposable element in cell culture and in both neurons and glial cells of aging fruit flies
  • Intercellular viral spread and intracellular transposition of Drosophila gypsy

    Roo Transposon
  • Different structural variants of roo retrotransposon are active in Drosophila melanogaster
  • Interrogating the 5'UTR tandem repeats of retrotransposon roo of Drosophila about horizontal transfer
  • Chromatin remodelling and retrotransposons activities during regeneration in Drosophila

    P element
  • The transposition rate has little influence on the plateauing level of the P-element

    Transposons and telomeres
  • see the Telomere page of The Interactive Fly
  • Subcellular localization and Egl-mediated transport of telomeric retrotransposon HeT-A ribonucleoprotein particles in the Drosophila germline and early embryogenesis
  • Telomeric TART elements target the piRNA machinery in Drosophila
  • Paramutation-like Epigenetic Conversion by piRNA at the Telomere of Drosophila virilis

    Evolution of Transposons
  • Double insertion of transposable elements provides a substrate for the evolution of satellite DNA
  • Evolutionary history and classification of Micropia retroelements in Drosophilidae species
  • Nanopore sequencing and Hi-C scaffolding provide insight into the evolutionary dynamics of transposable elements and piRNA production in wild strains of Drosophila melanogaster
  • Transposon activity, local duplications and propagation of structural variants across haplotypes drive the evolution of the Drosophila S2 cell line
  • Dynamics and Impacts of Transposable Element Proliferation in the Drosophila nasuta Species Group Radiation
  • Transposable element accumulation drives size differences among polymorphic Y Chromosomes in Drosophila

    Transposons and hybrid incompatability
  • Drosophila IRBP bZIP heterodimer binds P-element DNA and affects hybrid dysgenesis
  • Spontaneous gain of susceptibility suggests a novel mechanism of resistance to hybrid dysgenesis in Drosophila virilis
  • Hybrid dysgenesis in Drosophila virilis results in clusters of mitotic recombination and loss-of-heterozygosity but leaves meiotic recombination unaltered
  • Paternally inherited P-element copy number affects the magnitude of hybrid dysgenesis in Drosophila simulans and D. melanogaster
  • piRNAs are associated with diverse transgenerational effects on gene and transposon expression in a hybrid dysgenic syndrome of D. virilis
  • Defective satellite DNA clustering into chromocenters underlies hybrid incompatibility in Drosophila
  • Hybrid incompatibility between Drosophila virilis and D. lummei is stronger in the presence of transposable elements

    RNAi pathway and transposons
  • The somatic piRNA pathway controls germline transposition over generations
  • The Drosophila fragile X mental retardation protein participates in the piRNA pathway
  • Antisense transcription of retrotransposons in Drosophila: The origin of endogenous small interfering RNA precursors
  • Natural variation of piRNA expression affects immunity to transposable elements
  • piRNA-mediated regulation of transposon alternative splicing in the soma and germ line
  • Antisense transcription of retrotransposons in Drosophila: The origin of endogenous small interfering RNA precursors
  • Pathogenic tau-induced piRNA depletion promotes neuronal death through transposable element dysregulation in neurodegenerative tauopathies
  • Maelstrom represses canonical Polymerase II transcription within bi-directional piRNA clusters in Drosophila melanogaster
  • The integrity of piRNA clusters is abolished by insulators in the Drosophila germline
  • Dynamics of transposable element invasions with piRNA clusters
  • Trapping a somatic endogenous retrovirus into a germline piRNA cluster immunizes the germline against further invasion
  • Specialization of the Drosophila nuclear export family protein Nxf3 for piRNA precursor export
  • The evolutionary arms race between transposable elements and piRNAs in Drosophila melanogaster
  • Viral infection impacts transposable element transcript amounts in Drosophila

    Transposons and silencing of transposable elements and structural genes
  • RNA editing regulates transposon-mediated heterochromatic gene silencing
  • Drosophila small ovary gene is required for transposon silencing and heterochromatin organisation and ensures germline stem cell maintenance and differentiation
  • Transposon silencing in the Drosophila female germline is essential for genome stability in progeny embryos
  • Requirements for multivalent Yb body assembly in transposon silencing in Drosophila
  • The evolution of small-RNA-mediated silencing of an invading transposable element
  • Channel Nuclear Pore Complex subunits are required for transposon silencing in Drosophila
  • Maternally inherited piRNAs direct transient heterochromatin formation at active transposons during early Drosophila embryogenesis
  • Establishment of H3K9me3-dependent heterochromatin during embryogenesis in Drosophila miranda
  • Piwi-piRNA complexes induce stepwise changes in nuclear architecture at target loci. EMBO J: e108345
  • piRNA-independent transposon silencing by the Drosophila THO complex
  • Vasa sequesters transposon mRNAs in nuage via phase separation requiring RNA binding and self-association<


  • The centrosomal protein CP190 is a component of the gypsy chromatin insulator

    Chromatin insulators, or boundary elements, affect promoter-enhancer interactions and buffer transgenes from position effects. The gypsy insulator of Drosophila, a component of the gypsy long terminal repeat (LTR) retrotransposon, is bound by a protein complex with two characterized components, the zinc finger protein Suppressor of Hairy-wing [Su(Hw)] and Mod(mdg4)2.2, which is one of the multiple spliced variants encoded by the modifier of mdg4 [mod(mdg4)] gene. A genetic screen for dominant enhancers of the mod(mdg4) phenotype identified the Centrosomal Protein 190 (CP190) as an essential constituent of the gypsy insulator. The function of the centrosome is not affected in CP190 mutants whereas gypsy insulator activity is impaired. CP190 associates physically with both Su(Hw) and Mod(mdg4)2.2 and colocalizes with both proteins on polytene chromosomes. CP190 does not interact directly with insulator sequences present in the gypsy retrotransposon but binds to a previously characterized endogenous insulator, and it is necessary for the formation of insulator bodies. The results suggest that endogenous gypsy insulators contain binding sites for CP190, which is essential for insulator function, and may or may not contain binding sites for Su(Hw) and Mod(mdg4)2.2 (Pai, 2004).

    A genetic screen for dominant enhancers of mod(mdg4) has resulted in the identification of CP190 as a third component of the gypsy insulator. CP190 is present at gypsy retrotransposon insulator sites and overlaps extensively with Su(Hw) and Mod(mdg4)2.2 at presumed endogenous insulators. CP190 displays a specific distribution pattern on polytene chromosomes, showing significant overlap with Su(Hw) and Mod(mdg4)2.2 at the junctions between transcriptionally inert bands and transcriptionally active interbands. Similar localization patterns have been reported for other insulators. For example, the faswb insulator at the notch locus and the BEAF-32 protein of the scs' insulator are also present at the boundaries between bands and interbands. Results suggest that CP190 can bind DNA on its own or can be tethered to the chromosome through interactions with Su(Hw). Mutations in the CP190 gene impair the function of the insulator present in the gypsy retrotransposon without affecting the presence of Su(Hw) and Mod(mdg4)2.2, suggesting an essential task for CP190 in the activity of this insulator. In addition, the lethality of CP190 mutants suggests a critical role for the CP190 protein in the function of gypsy endogenous insulators. This essential role may be a consequence of the requirement of CP190 for the formation of insulator bodies in the nuclei of diploid cells (Pai, 2004).

    The insulator present in the gypsy retrotransposon contains only Su(Hw) binding sites, and CP190 is present in this insulator through direct interactions with Su(Hw). The gypsy insulator contains 12 Su(Hw) binding sites, and at least four are needed for insulator activity. However, clusters of three or more Su(Hw) binding sites are rare in the genome. Therefore, a critical question is whether the sites of Su(Hw) and Mod(mdg4)2.2 localization present throughout the genome truly function as insulators. The presence of CP190 at these sites and its ability to bind DNA might explain this apparent paradox. For example, the endogenous insulator present in the yellow-achaete region has only two binding sites for Su(Hw). Nevertheless, the y454 fragment containing this insulator is able to bind CP190, suggesting that this protein might act in concert with Su(Hw) to confer insulator activity. It is therefore possible that endogenous gypsy insulators are composed of binding sites for Su(Hw) and/or for CP190 and, together with Mod(mdg4)2.2, form a complex. Endogenous gypsy insulators may have few or no Su(Hw) binding sites, and they may rely on CP190 to bind DNA and tether other insulator components such as Mod(mdg4)2.2 via protein-protein interactions (Pai, 2004).

    Previous studies have suggested that gypsy insulators separated at a distance in the genome may come together and form large insulator bodies in the nucleus during interphase. These aggregates represent higher order structures of chromatin and are implicated in the regulation of gene expression by compartmentalizing the genome into transcriptionally independent domains. The formation of these aggregates appears to require Mod(mdg4) function because the large aggregates are missing in mod(mdg4) mutants. The formation of gypsy insulator bodies is severely impaired also in CP190 mutants, suggesting that CP190 plays an essential role in the formation of these bodies and in the establishment of the chromatin domain organization mediated by gypsy endogenous insulators. It is possible that the BTB/POZ protein-protein interaction domains of both CP190 and Mod(mdg4)2.2 are required for and contribute to the stability of the interactions among insulator sites. In vitro-expressed CP190 lacking the BTB/POZ domain is soluble, whereas the wt protein is not, further suggesting that CP190 might exist as a complex with itself or other proteins in vivo, and the formation of this complex is likely mediated by the BTB/POZ domain. However, because CP190 is present at the gypsy insulator in the absence of Mod(mdg4)2.2 protein, the interaction between these two proteins may not be crucial for CP190 recruitment to the insulator (Pai, 2004).

    Previous studies have identified CP190 as a centrosome-specific protein during mitosis that also associates with chromatin during interphase. Although many of these studies have focused on the possible role of CP190 during cell division, the current results suggest that centrosomal function and cell division are not affected in CP190 mutants. This conclusion is supported by independent studies of CP190 function during the cell cycle. The main function of CP190 might then be to regulate chromosome-related processes during interphase. Several lines of evidence suggest that this role is related to the function of the gypsy insulator: mutations in CP190 alter gypsy-induced phenotypes; CP190 colocalizes with Su(Hw) and Mod(mdg4)2.2 on polytene chromosomes and in diploid cell nuclei, and CP190 associates physically with gypsy insulator components in vitro and in vivo. However, the centrosomal localization of CP190 might also be important for its role in the gypsy insulator despite being unnecessary for cell cycle progression. The centrosome could either be a temporary storage site for CP190 during mitosis, or a site for a mitosis-specific modification that could be important for CP190 reassociation with chromosomes later in the cell cycle. The presence of CP190 in the centrosome could also be related to the regulation of the level of this protein in the cell. In fact, it has been shown that some chromatin-binding proteins are targeted to the centrosome for degradation. Alternatively, the presence of CP190 at the centrosome might be related to a possible role in the ubiquitin modification pathway. Recent findings have linked BTB/POZ domain proteins to ubiquitin E3 ligase function, some of which are known to be present at the centrosome. CP190 may be involved in similar types of interactions as an adaptor for ubiquitin E3 ligases and might target associated insulator proteins to the centrosome during mitosis for ubiquitination and/or degradation, which in turn may be required for properly reestablishing chromosome domain boundaries after mitosis (Pai, 2004).

    Distribution, evolution, and diversity of retrotransposons at the flamenco locus reflect the regulatory properties of piRNA cluster

    Most understanding of Drosophila heterochromatin structure and evolution has come from the annotation of heterochromatin from the isogenic y; cn bw sp strain. However, almost nothing is known about the heterochromatin's structural dynamics and evolution. This study has focused on a 180-kb heterochromatic locus producing Piwi-interacting RNAs (piRNA cluster), the flamenco (flam) locus, known to be responsible for the control of at least three transposable elements (TEs). Its detailed structure is reported in three different Drosophila lines chosen according to their capacity to repress or not to repress the expression of two retrotransposons named ZAM and Idefix, and they were shown to display high structural diversity. Numerous rearrangements due to homologous and nonhomologous recombination, deletions and segmental duplications, and loss and gain of TEs are diverse sources of active genomic variation at this locus. Notably, a correlation is evidenced between the presence of ZAM and Idefix in this piRNA cluster and their silencing. They are absent from flam in the strain where they are derepressed. It was shown that, unexpectedly, more than half of the flam locus results from recent TE insertions and that most of the elements concerned are prone to horizontal transfer between species of the melanogaster subgroup. A model is built showing how such high and constant dynamics of a piRNA master locus open the way to continual emergence of new patterns of piRNA biogenesis leading to changes in the level of transposition control (Zanni, 2013).

    The piRNA pathway plays a crucial role in TE silencing and is conserved among species. However, the mechanism by which this system adapts to new mobile elements is still obscure. The current data show a high insertion rate of recent TEs in the flam piRNA cluster far exceeding that previously suspected. A model has been developed in which Rhino protein might interact with the integration machinery of TEs to direct their integration into heterochromatin and, more specifically, into piRNA clusters. The current results concerning ZAM and Idefix highlight how the presence or absence of retrotransposons in piRNA-producing loci makes some Drosophila lines more susceptible to TE invasions than others, and thus how piRNA clusters affect the genomic TE distribution. A strict correlation was observed between the presence of ZAM and Idefix in the flam locus and their silencing. Consistently, their deletion from the flam locus observed in the Rev line is correlated with their activation, characterized by high mobilization, instability, and copy accumulation. A deeper analysis of the flam structure revealed that deletions occur frequently in the locus. Mostly, they affect internal segments of TEs, ranging from several base pairs up to several kilobases, affecting both ancient TEs, remaining as vestiges in the locus, and recently inserted TEs. The longest internal deletions affecting retrotransposons are due to homologous recombination between LTRs leading to the complete elimination of internal sequences. Moreover, large deletions may eliminate several TEs within one mutational event, as seen for ZAM and Idefix in the Rev line. At the same time, insertions also occur within the flam locus, as exemplified by the high proportion of recently inserted TEs, the recent insertions of 412 and Stalker2 in the Iso1A strain, and short and long segmental duplications. Such genetic dynamics of a piRNA master locus open the way to a constant emergence of new patterns of piRNA biogenesis potentially leading to changes in the level of transposition control (Zanni, 2013).

    The present data fit well with a model of TE invasion and its subsequent control by the invaded species as follows. The best genetic background for a TE to invade a genome and have full activity should be a 'virgin' genome devoid of any related copy. The best chance to find a virgin genome is to invade another species by horizontal transfer. In this genome, the incoming TE is not silenced, and is thus able to transpose at high frequency. A period of instability of the newly acquired TE results in its increased copy number. Insertions into piRNA clusters like flam are then highly probable because the current data evidence high content of recent TEs in such loci. These insertions would be associated with production of corresponding piRNAs and silencing of homologous elements. Thus, as soon as one copy of the TE is inserted in a piRNA cluster, a time of stability follows. This suggests that TEs regulated by a certain piRNA cluster should be present only once in this locus, as is seen for most TEs within flam. One or several deletion events can then lead to elimination of TE copies from the locus. A new period of activity of the remaining functional elements in the genome starts. Because deletion events may delete several elements from the locus, transposition bursts may happen involving several different TEs at the same time. This new period of instability for the TEs offers the opportunity to insert into a piRNA cluster again. When this occurs, stability is regained. Thus, transposition bursts, periods of stability, and periods of instability shaping the Drosophila genome would be directly correlated to the mutational events that affect piRNA clusters like flam. This scenario supports the hypothesis proposed by Le Rouzic (2005) that successful invasion of a population by TEs should be possible "thanks to an initial transposition burst followed by a strong limitation of their activity" (Zanni, 2013).

    Rounds of high transposition rate can trigger genetic instabilities and disease-associated mutations, but there is no doubt that they also play an essential role in the evolution of species. Actually, the current Drosophila genome witnesses multiple transposition bursts over time for most of the TE families, resulting in ancient and recent copies being present in the genome (examples from this study are Blood, Stalker2, Stalker4, Gypsy1, and Phidippo). The case of Pifo depicted in this study is different and certainly represents a case of a new invasion of D. melanogaster, because no ancient Pifo elements can be found in the genome. Such high dynamicity of piRNA clusters should also remodel heterochromatic regions in other Drosophila species. In D. erecta and D. yakuba, flam loci have been shown to contain a large amount of TEs that are completely different from the D. melanogaster flam elements. These data illustrate the dynamics of piRNA clusters and their coevolution with the rest of the genome regarding TE content. They also highlight the essential role that piRNA clusters might play in speciation by remodeling via TE control of large genomic regions (Zanni, 2013).

    Retrotransposons Down- and Up-Regulation in Aging Somatic Tissues

    The transposon theory of aging hypothesizes the activation of transposable elements (TEs) in somatic tissues with age, leading to a shortening of the lifespan. It is thought that TE activation in aging produces an increase in DNA double-strand breaks, contributing to genome instability and promoting the activation of inflammatory responses. To investigate how TE regulation changes in somatic tissues during aging, this study analyzed the expression of some TEs, as well as a source of small RNAs that specifically silence the analyzed TEs; the Drosophila cluster named flamenco. Significant variations was found in the expression levels of all the analyzed TEs during aging, with a trend toward reduction in middle-aged adults and reactivation in older individuals that suggests dynamic regulation during the lifespan (Giordani, 2021).

    The somatic piRNA pathway controls germline transposition over generations

    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. This study shows, 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 (Barckmann, 2018).

    RNA editing regulates transposon-mediated heterochromatic gene silencing

    Heterochromatin formation drives epigenetic mechanisms associated with silenced gene expression. Repressive heterochromatin is established through the RNA interference pathway, triggered by double-stranded RNAs (dsRNAs) that can be modified via RNA editing. However, the biological consequences of such modifications remain enigmatic. This study shows that RNA editing regulates heterochromatic gene silencing in Drosophila. The binding activity of an RNA-editing enzyme was used to visualize the in vivo production of a long dsRNA trigger mediated by Hoppel transposable elements. Using homologous recombination, this trigger was deleted, dramatically altering heterochromatic gene silencing and chromatin architecture. Furthermore, it was shown that the trigger RNA is edited and that dADAR serves as a key regulator of chromatin state. Additionally, dADAR auto-editing generates a natural suppressor of gene silencing. Lastly, systemic differences in RNA editing activity generates interindividual variation in silencing state within a population. These data reveal a global role for RNA editing in regulating gene expression (Savva, 2013).

    This study pursued an observation of the in vivo localization of the RNA-editing enzyme, dADAR, to the proof of its action on an endogenously expressed inverted repeat of the TE, Hoppel. The results explicitly demonstrate a functional intersection between the processes of RNA editing and RNA silencing. Previous studies in Drosophila implicate Hoppel and the RNAi pathway in determining the global silencing state of chromosome 4, although no dsRNA trigger had been experimentally identified. This study showed that the inverted repeat acts as a genetic element, Hok, and regulates PEV, the global architecture of chromosome 4, and silences the Hoppel transposase. As a general mechanism, ADAR's action on dsRNA should oppose RNAi. It was shown that deficiency for ADAR acts as a global enhancer of silencing state, and dADAR hypomorphism even extends lifespan. In Drosophila, gene silencing decreases with age and has been implicated in the aging process (Wood, 2010). Thus, substantial decreases in ADAR activity may lead to lifespan extension through increased silencing. Interestingly, polymorphisms within a human ADAR gene have been associated with extreme longevity, indicating that interventions involving ADAR activity may be capable of affecting lifespan. Importantly, mutations in human ADAR1 cause Aicardi–Goutières syndrome in which it is hypothesized that ADAR has a role in regulating dsRNA metabolism from repeated elements in the human genome. Thus, the current data are consistent with a conserved role in the regulation of dsRNA levels in animals through RNA editing or RNA binding (Savva, 2013).

    Mechanistically, evidence is provided that dADAR auto-editing has evolved as a natural inhibitor of RNAi, generating dADARG. In dAdar null or dAdarS genetic backgrounds, no dADARG can be produced. Thus, both backgrounds effectively act as enhancers of PEV (E(var)). In the wild-type background, PEV occurs to the extent that each animal expresses dADAR (and the corresponding amount of dADARG). In the extreme, the dAdarG background acts as a strong suppressor of PEV (Su(var)). How can a single amino acid change in dADAR protein affect such a silencing switch? It is speculated that dADARG may interfere indirectly with Dicer activity on dsRNA, simply by blocking access via binding irrespective of editing activity, analogous to the FHV-B2 protein. Alternately, a recent study showed a direct functional interaction between mammalian ADAR and Dicer that is necessary for the processing of small RNAs (Ota, 2013). If dADAR has a similar interaction, it could also mediate all of the effects in the model via dominant-negative interactions of dADARG with Dicer, whereas dADARS (which encodes the conserved amino acid) would function in a similar manner described in mammals to promote small RNA biogenesis. Further biochemical experiments will be necessary to determine whether this phenomenon is conserved across species and the exact molecular mechanisms through which dADARG exerts its effects (Savva, 2013).

    The most engaging aspect of these results lay in their implications for somatic regulation of heterochromatin functioning as a safeguard of transposon activity, especially in the nervous system. The RNA-induced silencing complex isolated from Drosophila tissue-culture cells was shown to be programmed with esiRNAs, largely derived from transposon sequences, a significant portion of which bears the signature of a single dADAR modification. Likewise, in C. elegans, ADAR activity has a profound effect on the abundance and identity of small RNA profiles. Further experiments in this system using deep sequencing technologies will be necessary to shed light on the effects of ADAR on endo-siRNA abundances and functionality. It is envisioned that such RNA-editing-mediated effects may be quite specific to the nature of individual dsRNA triggers. Studies in both mammals and Drosophila have shown that TEs are mobile in the nervous system, revealing an intriguing mechanism for the generation of somatic mutations potentially conferring adaptive value in individuals (Li, 2013; Muotri, 2005; Perrat, 2013). This study demonstrates a mechanistic link between RNA editing and the regulation of transposon silencing, particularly in the nervous system, which may have domesticated uses as diversifiers of neuronal genomes on a neuron-to-neuron and an individual-to-individual basis. The implications of these results, given the universal prevalence of dsRNAs as a component of transcriptomes, are that ADAR activity has an evolved role in determining the fate of RNAs entering silencing pathways, thus globally influencing somatic genomic integrity, gene expression and downstream organismal phenotypes (Savva, 2013).

    Active retrotransposons help maintain pericentromeric heterochromatin required for faithful cell division

    Retrotransposons are populated in vertebrate genomes, and when active, are thought to cause genome instability with potential benefit to genome evolution. Retrotransposon-derived RNAs are also known to give rise to small endo-siRNAs to help maintain heterochromatin at their sites of transcription; however, as not all heterochromatic regions are equally active in transcription, it remains unclear how heterochromatin is maintained across the genome. This study addresses these problems by defining the origins of repeat-derived RNAs and their specific chromatin locations in Drosophila S2 cells. Repeat RNAs are predominantly derived from active gypsy elements and processed by Dcr-2 into small RNAs to help maintain pericentromeric heterochromatin. In cultured S2 cells that synthetic repeat-derived endo-siRNA mimics are sufficient to rescue Dcr-2-deficiency-induced defects in heterochromatin formation in interphase and chromosome segregation during mitosis, demonstrating that active retrotransposons are required for stable genetic inheritance (Hao, 2020).

    The Drosophila fragile X mental retardation protein participates in the piRNA pathway

    RNA metabolism controls multiple biological processes, and a specific class of small RNAs, called piRNAs, act as genome guardians by silencing the expression of transposons and repetitive sequences in the gonads. Defects in the piRNA pathway affect genome integrity and fertility. The possible implications in physiopathological mechanisms of human diseases have made the piRNA pathway the object of intense investigation, and recent work suggests that there is a role for this pathway in somatic processes including synaptic plasticity. The RNA-binding fragile X mental retardation protein (FMRP, also known as FMR1) controls translation and its loss triggers the most frequent syndromic form of mental retardation as well as gonadal defects in humans. This study demonstrates for the first time that germline, as well as somatic expression, of Drosophila Fmr1 (denoted dFmr1), the Drosophila ortholog of FMRP, are necessary in a pathway mediated by piRNAs. Moreover, dFmr1 interacts genetically and biochemically with Aubergine, an Argonaute protein and a key player in this pathway. These data provide novel perspectives for understanding the phenotypes observed in Fragile X patients and support the view that piRNAs might be at work in the nervous system (Bozzetti, 2015).

    dFmr1 is a translational regulator and its role in the miRNA pathway is widely accepted. This study provides several lines of evidence that dFmr1 can be considered as a ‘bona fide’ member of the piRNA pathway that keeps repetitive sequences and transposons silenced. First, dFmr1 mutant testes display crystalline aggregates, as do other mutants of the piRNA pathway. Second, the levels of cry (Suppressor of Stellate)-specific and transposon-specific piRNAs dramatically decrease in dFmr1 mutant testes. Third, as a consequence of this decrease, the Ste RNA is produced and, in addition, transposons are expressed at higher levels than in wt animals. Fourth, dFmr1 mutant animals display fertility defects, a phenotype shown by several mutations affecting the piRNA pathway. The fact that earlier screens did not identify dFmr1 as a member of the somatic piRNA pathway could be due to the heterogeneous phenotypes observed with the somatic transposons (this study) and/or to the material used for those assays. The crySte system thus proves very efficient for identifying new members of this important pathway (Bozzetti, 2015).

    The movement of transposable elements is one of the molecular causes of DNA instability and sterility. Considering that human patients mutant for FMRP also display defects in male and female gonads, it will be interesting to characterize the activity of transposons and repetitive sequences in the gonads of mice or humans that are mutant for the FMRP pathway, although there might be no observable defects in mammals because they express three members of the FMRP family versus the single ortholog in fly. Finally, mutations affecting the piRNA pathway might also induce gonadal defects in humans (Bozzetti, 2015).

    Until now, the members of the piRNA pathway controlling the crySte interaction, including Aub, have been described as being required in the male germline. Surprisingly, the conditional dFmr1 rescue and KD experiments demonstrate that dFmr1 controls the piRNA pathway both in the germline and in the somatic cells of the gonad, which raises questions as to the somatic contribution of other members of the piRNA pathway in the male gonad. The phenotypes induced by somatic Aub expression also suggest that the hub expresses one or more AGO proteins that are involved in the somatic piRNA-mediated Ste silencing and that interact with dFmr1; however, the only other protein of the Piwi clade present in the somatic tissue, Piwi, does not participate in Ste silencing. Based on preliminary data, this study proposes that AGO1 might be one such protein. First, AGO1/+ testes display Ste-made crystals, as do testes expressing UAS-AGO1 RNAi driven by the upd-Gal4 driver. Second, aubsting rescues the AGO1-mediated crystal phenotype. Third, AGO1 and dFmr1 interact biochemically and are known to interact genetically in the ovaries to control germline stem cell maintenance, as well as in the nervous system, where they modulate synaptic plasticity. Taken together, these data suggest that AGO1 contributes to the piRNA pathway that controls the cry–Ste system in the somatic part of the gonad (Bozzetti, 2015).

    The finding that Aub somatic expression affects the NMJ and counteracts the AGO1 loss of function phenotype is also unexpected. Recent work has documented the activation of piRNA pathway in the nervous system in flies, mice, humans and molluscs and it has been proposed that synaptic plasticity, cognitive functions and neurodegeneration might involve the control of genome stability, even though the precise mode of action and impact of this pathway are not completely understood. Because Aub is not required in the larval somatic tissues, its ectopic expression could affect the NMJ by replacing AGO1 in its known role on the miRNA pathway. However, AGO1 might also affect the NMJ through the piRNA pathway, much in the same way as AGO1 loss of function affects a piRNA pathway in the gonad. Even though AGO1 has been previously described as being exclusively involved in the miRNA pathway, some degree of overlapping between different RNAi pathways has been recently described: (1) the double-stranded-RNA-binding protein Loquacious (Loqs) is involved in the miRNA pathway and in the endogenous siRNA pathway, (2) AGO1 and AGO2 can compete for binding with miRNAs, and (3) ectopic expression of Aub in the soma competes for the siRNAs pathway mediated by AGO2. In addition, miRNAs have been demonstrated to have a role on easi-RNA biogenesis in plants. In a similar manner, AGO1 could act on piRNAs through its activity on the miRNA pathway. Although future studies will clarify the connection between AGO1 and the piRNA pathway, the present data provide novel perspectives in the field and could have a broad relevance to diseases affecting cognitive functions (Bozzetti, 2015).

    Expression, genetic and biochemical data indicate that Aub and dFmr1 interact directly. dFmr1 has been proposed to bind specific cargo RNAs and the human FMRP binds small RNA, in addition to mRNAs. Similarly, the Aub–dFmr1 interaction might allow the targeting of piRNAs to the transcripts of repetitive sequences and transposable elements, dFmr1 providing the molecular link between small RNAs and AGO proteins of the RISC (Bozzetti, 2015).

    The Aub and dFmr1 proteins colocalize and likely interact in the piRNA pathway in a specific stage of testis development and also have additional functions that are independent from each other. Typically, dFmr1 accumulates at high levels in more differentiated cells of the testis, where Aub is not detectable, likely accounting for the axoneme phenotype described in dFmr1 testes. In the future, it will be interesting to analyze whether the other genes involved in the piRNA pathway in testis are also required at specific stages, as also recently found in the ovary (Bozzetti, 2015).

    Finally, FMRP proteins work in numerous molecular networks, show complex structural features (TUDOR, KH, NLS, NES RGG domains) and are characterized by widespread expression and subcellular localization (cytoplasm, nucleus, axons, dendrites, P bodies), providing versatile platforms that control mRNA and small RNA metabolism (e.g. translation, degradation and transport). Understanding whether FMRP proteins interact with other members of the piRNA pathway, whether this interaction is modulated physiologically and how does the interaction with this pathway compare with that observed with other AGO proteins will clarify the role and mode of action this family of proteins in small RNA biogenesis and metabolism (Bozzetti, 2015).

    The biogenesis of the piRNAs requires two pathways. The primary pathway involves Piwi and predominantly occurs in the somatic tissues. The ping-pong pathway involves Aub, as well as AGO3, and predominantly occurs in the germline, where Aub is thought to bind an antisense piRNA, to cleave the sense transcript from an active transposon and to produce a sense piRNA that is loaded onto AGO3. The AGO3–piRNA complex binds complementary transcripts from the piRNA cluster, producing the so-called secondary piRNAs by an amplification loop. Although the piRNA pathways have emerged as a very important tool to understand the role of RNA metabolism in physiological and pathological conditions, the relationship and interactions among the involved proteins are not simple to interpret, mostly because not all the players have been characterized. Moreover, recent data support the hypothesis that the somatic and the germline piRNA pathways share components: for example, shutdown (shu), vreteno (vret) and armitage (arm) affect primary as well as ping-pong pathways in ovaries. Results from this study call for a role of dFmr1 in both piRNA pathways at least in testes. Based on the alignment of the human, mouse and fly FMRP family members, dFmr1 might participate in piRNA biogenesis as a Tudor domain (TDRD) containing protein (Bozzetti, 2015).

    TDRDs are regions of about 60 amino acids that were first identified in a Drosophila protein called Tudor. In the recent years, the requirement of TDRD proteins in piRNA biogenesis and metabolism has become evident. Typically, the founding member of the family, Tudor, binds AGO proteins and helps them interact with specific piRNAs. Among the different TDRD proteins, fs(1)Yb works in the primary pathway; Krimper, Tejas, Qin/Kumo, and PAPI work in the ping-pong pathway; and Vret works in both systems. PAPI, the only TDRD protein that has a modular structure closely related to dFmr1 (two KH domains and one TDRD), interacts with the di-methylated arginine residues of AGO3 and controls the ping-pong cycle in the nuage. At least during the early stages of testis development, dFmr1 might interact with Aub in a similar way. Given that TDRDs are involved in the interactions between proteins and in the formation of ribonucleoprotein complexes, future studies will assess whether RNAs mediate the Aub–dFmr1 interaction (Bozzetti, 2015).

    In conclusion, the discovery of dFmr1 as a player in the piRNA pathway highlights the importance of the fly model. Data from this study also adds a new perspective to understanding the role and mode of action of this protein family and the physiopathological mechanisms underlying the Fragile X syndrome (Bozzetti, 2015).

    Unique transposon landscapes are pervasive across Drosophila melanogaster genomes

    To understand how transposon landscapes (TLs) vary across animal genomes, this study describes a new method called the Transposon Insertion and Depletion AnaLyzer (TIDAL) and a database of >300 TLs in Drosophila melanogaster (TIDAL-Fly). This analysis reveals pervasive TL diversity across cell lines and fly strains, even for identically named sub-strains from different laboratories such as the ISO1 strain used for the reference genome sequence. On average, >500 novel insertions exist in every lab strain, inbred strains of the Drosophila Genetic Reference Panel (DGRP), and fly isolates in the Drosophila Genome Nexus (DGN). A minority (<25%) of transposon families comprise the majority (>70%) of TL diversity across fly strains. A sharp contrast between insertion and depletion patterns indicates that many transposons are unique to the ISO1 reference genome sequence. Although TL diversity from fly strains reaches asymptotic limits with increasing sequencing depth, rampant TL diversity causes unsaturated detection of TLs in pools of flies. Finally, novel transposon insertions were shown to negatively correlate with Piwi-interacting RNA (piRNA) levels for most transposon families, except for the highly-abundant roo retrotransposon. This study provides a useful resource for Drosophila geneticists to understand how transposons create extensive genomic diversity in fly cell lines and strains (Rahman, 2015).

    Antisense transcription of retrotransposons in Drosophila: The origin of endogenous small interfering RNA precursors

    To repress transposons and combat genomic instability, eukaryotes have evolved several small RNA mediated defense mechanisms. Specifically, in Drosophila somatic cells, endogenous small interfering (esi)RNAs suppress retrotransposon mobility. EsiRNAs are produced by Dicer-2 processing of double-stranded RNA precursors, yet the origins of these precursors are unknown. This study shows that most transposon families are transcribed in both the sense and antisense direction. LTR retrotransposons are generated from intra element transcription start sites with canonical RNA polymerase II promoters. Retrotransposon antisense transcripts were shown to be less polyadenylated than sense transcripts, which may promote nuclear retention of antisense transcripts and the double-stranded RNAs they form. Dicer-2 RNAi-depletion causes a decrease in the number of esiRNAs mapping to retrotransposons. These data support a model in which double-stranded RNA precursors are derived from convergent transcription and processed by Dicer-2 into esiRNAs that silence both sense and antisense retrotransposon transcripts. Reduction of sense retrotransposon transcripts potentially lowers element specific protein levels to prevent transposition. This mechanism preserves genomic integrity and is especially important for Drosophila fitness because mobile genetic elements are highly active (Russo, 2015).

    piRNAs are associated with diverse transgenerational effects on gene and transposon expression in a hybrid dysgenic syndrome of D. virilis

    Hybrid dysgenic syndromes are a strong form of genomic incompatibility that can arise when transposable element (TE) family abundance differs between two parents. When TEs inherited from the father are absent in the mother's genome, TEs can become activated in the progeny, causing germline damage and sterility. Studies in Drosophila indicate that dysgenesis can occur when TEs inherited paternally are not matched with a pool of corresponding TE silencing PIWI-interacting RNAs (piRNAs) provisioned by the female germline. Using the D. virilis syndrome of hybrid dysgenesis as a model, this study characterize the effects that divergence in TE profile between parents has on offspring. Overall, this study shows that divergence in the TE landscape is associated with persisting differences in germline TE expression when comparing genetically identical females of reciprocal crosses and these differences are transmitted to the next generation. Moreover, chronic and persisting TE expression coincides with increased levels of genic piRNAs associated with reduced gene expression. Gene expression is idiosyncratically influenced by differences in the genic piRNA profile of the parents that arise though polymorphic TE insertions. Overall, these results support a model in which early germline events in dysgenesis establish a chronic, stable state of both TE and gene expression in the germline that is maintained through adulthood and transmitted to the next generation (Erwin, 2015).

    Defective satellite DNA clustering into chromocenters underlies hybrid incompatibility in Drosophila

    Although rapid evolution of pericentromeric satellite DNA repeats is theorized to promote hybrid incompatibility (HI), how divergent repeats affect hybrid cells remains poorly understood. Recently, it was demonstrated that sequence-specific DNA-binding proteins cluster satellite DNA from multiple chromosomes into 'chromocenters', thereby bundling chromosomes to maintain the entire genome in a single nucleus. This study shows that ineffective clustering of divergent satellite DNA in the cells of Drosophila hybrids results in chromocenter disruption, associated micronuclei formation and tissue atrophy. It was further demonstrated that previously identified HI factors trigger chromocenter disruption and micronuclei in hybrids, linking their function to a conserved cellular process. Together, a unifying framework is proposed that explains how the widely observed satellite DNA divergence between closely related species can cause reproductive isolation (Jagannathan, 2021).

    Hybrid incompatibility between Drosophila virilis and D. lummei is stronger in the presence of transposable elements

    Mismatches between parental genomes in selfish elements are frequently hypothesized to underlie hybrid dysfunction and drive speciation. However, because the genetic basis of most hybrid incompatibilities is unknown, testing the contribution of selfish elements to reproductive isolation is difficult. This study evaluated the role of transposable elements (TEs) in hybrid incompatibilities between Drosophila virilis and D. lummei by experimentally comparing hybrid incompatibility in a cross where active TEs are present in D. virilis (TE+) and absent in D. lummei, to a cross where these TEs are absent from both D. virilis (TE-) and D. lummei genotypes. Using genomic data, copy number differences in TEs were confirmed between the D. virilis (TE+) strain and both the D. virilis (TE-) strain and D. lummei. F1 postzygotic reproductive isolation was observed exclusively in the interspecific cross involving TE+ D. virilis but not in crosses involving TE- D. virilis. This mirrors intraspecies dysgenesis where atrophied testes only occur when TE+ D. virilis is the paternal parent. A series of backcross experiments, that accounted for alternative models of hybrid incompatibility, showed that both F1 hybrid incompatibility and intrastrain dysgenesis are consistent with the action of TEs rather than genic interactions. Thus, these data suggest that this TE mechanism manifests as two different incompatibility phenotypes. A further Y-autosome interaction contributes to additional, sex-specific, inviability in one direction of this cross-combination. These experiments demonstrate that TEs that cause intraspecies dysgenesis can increase reproductive isolation between closely related lineages, thereby adding to the processes that consolidate speciation (Castillo, 2022).

    Production of small non-coding RNAs from the flamenco locus is regulated by the gypsy retrotransposon of Drosophila melanogaster

    Protective mechanisms based on RNA silencing directed against the propagation of transposable elements are highly conserved in eukaryotes. The control of transposable elements is mediated by small non-coding RNAs, which derive from transposon-rich heterochromatic regions that function as small RNA-generating loci. These clusters are transcribed and the precursor transcripts are processed to generate piRNAs and endo-siRNAs, which silence transposable elements in gonads and somatic tissues. The flamenco locus is a Drosophila melanogaster small RNA cluster that controls gypsy and other transposable elements, which has played an important role in understanding how small non-coding RNAs repress transposable elements. This study describe a cosuppression mechanism triggered by new euchromatic gypsy insertions in genetic backgrounds carrying flamenco alleles defective in gypsy suppression. The silencing of gypsy was found to be accompanied by the silencing of other transposons regulated by flamenco, and of specific flamenco sequences from which small RNAs against gypsy originate. This cosuppression mechanism seems to depend on a post-transcriptional regulation that involves both endo-siRNA and piRNA pathways and is associated with the occurrence of developmental defects. In conclusion, it is proposed that new gypsy euchromatic insertions trigger a post-transcriptional silencing of gypsy sense and antisense sequences, which modifies the flamenco activity. This cosuppression mechanism interferes with some developmental processes presumably by influencing the expression of specific genes (Guida, 2016).

    Drosophila IRBP bZIP heterodimer binds P-element DNA and affects hybrid dysgenesis

    In Drosophila, P-element transposition causes mutagenesis and genome instability during hybrid dysgenesis. The P-element 31-bp terminal inverted repeats (TIRs) contain sequences essential for transposase cleavage and have been implicated in DNA repair via protein-DNA interactions with cellular proteins. The identity and function of these cellular proteins were unknown. Biochemical characterization of proteins that bind the TIRs identified a heterodimeric basic leucine zipper (bZIP) complex between an uncharacterized protein that is termed 'Inverted Repeat Binding Protein (IRBP) 18' and its partner Xrp1. The reconstituted IRBP18/Xrp1 heterodimer binds sequence-specifically to its dsDNA-binding site within the P-element TIRs. Genetic analyses implicate both proteins as critical for repair of DNA breaks following transposase cleavage in vivo. These results identify a cellular protein complex that binds an active mobile element and plays a more general role in maintaining genome stability (Francis, 2016).

    A role for the IRBP complex in the P-element transposition reaction has been postulated since initial identification of its sequence-specific DNA-binding properties (Staveley, 1995; Beall, 1994; Beall, 1996; Rio, 1988). The organizational overlap between IRBP DNA binding and transposase cleavage sites makes the IRBP complex a prime cellular candidate to influence some aspect of the P-element transposition cycle. Several putative IRBP proteins copurified with observed IRBP DNA-binding activity or unambiguously promoted DNA repair posttransposase P-element cleavage (Ku 70 and mus309/DmBLM). None, however, could reconstitute site-specific DNA binding to the P-element 31-bp TIRs (Francis, 2016).

    This report identifies a bZIP heterodimer between a C/EBP family member IRBP18 and a drosophilid-specific protein Xrp1 as the sequence-specific DNA-binding subunits of a larger multiprotein IRBP complex that binds to the P-element TIRs. These proteins work in concert to facilitate efficient DNA repair following P-element transposase-mediated DNA cleavage. Finally, these proteins are required more generally in the cellular DNA damage response and DSB repair in the absence of P elements (Francis, 2016).

    Repair of P-element~induced DNA breaks occurs predominantly through two distinct DSB repair pathways: Non-homologous end joining (NHEJ) or a variant of classical homologous recombinational repair, synthesis-dependent strand annealing (SDSA), The choice between these two pathways is dictated by cell-cycle location, the availability of pathway substrates, and tissue type. Because the IRBP homozygous mutant males are sterile, it is not possible at present to use any of the established DNA repair reporter strains to determine in which DNA repair pathway IRBP18 and Xrp1 participates. Future would should determine if the IRBP18/Xrp1 heterodimer can bind directly to different DNA repair intermediates and thus provide a direct link between the bZIP heterodimer and DNA repair (Francis, 2016).

    A role for bZIP proteins and specifically mammalian C/EBP proteins in DNA repair is well established. In human and mouse keratinocytes, UV-B UV DNA damage induces p53-dependent transcriptional activation of both the C/EBPα and β genes. C/EBPα expressed in prostate cancer cells where it interacts with the DNA repair proteins Ku p70/p80 heterodimer and the poly (ADP ribose) polymerase 1 (PARP-1). Notably, Drosophila p53 up-regulates three bZIP proteins (CG6272/IRBP18, CG17836/Xrp1, and CG15479/Mabiki) upon DNA damage. CG15479/Mabiki is a novel regulator of caspase-independent cell death of excess cells in the expanded head region of 6x-bcd embryos and is thought to work in concert with other caspase-independent cell death mechanisms to ensure proper development. Genetic deletion of the CG17836/Xrp1 gene resulted in a DNA repair phenotype when challenged with ionizing radiation (Akdemir, 2007). Additionally, the Dmp53 DNA damage-induced apoptotic response was unaffected in the Xrp1 mutants, suggesting that Xrp1 functions to preserve genome stability through a pathway independent of apoptosis (Akdemir, 2007). Although this study has demonstrated that IRBP18 and Xrp1 share a similar function in DNA repair, more experiments are needed to understand how these proteins work downstream of p53 transactivation (Francis, 2016).

    D. melanogaster uses multiple endogenous mechanisms to limit P-element transposition. Expression of catalytically active transposase is restricted to the germline by tissue-specific pre-mRNA splicing regulation. The germline piwi-interacting RNA pathway has been demonstrated to repress transposition in trans and plays a critical role in host adaptation to newly invaded P elements. It is proposde that the IRBP18/Xrp1 heterodimer recognizes new P elements and that its native function is to facilitate repair of breaks to maintain genomic stability during a genotoxic event such as ionizing radiation or the massive P-element mobilization that occurs following a hybrid dysgenic cross (Francis, 2016).

    bZIP proteins are well suited to recognize newly invaded foreign DNA due to their inherit ability to form multiple heterodimers. IRBP18 and Xrp1, for example, form heterodimers with other several other bZIP proteins; the net result is expansion of the repertoire of DNA sequences that can be bound. This library of bZIP dimers can be deployed to recognize foreign DNA as part of a survival mechanism against the genome instability created by foreign DNA invasion. In humans, mice and Drosophila p53 transactivate steady-state levels of several bZIP proteins in response to DNA damage. It is unclear how changes in steady-state levels of these DNA repair proteins determine dimer formation or function. What is clear is that bZIP proteins are important players in DNA repair and maintenance of genome stability. In this respect, the IRBP18/Xrp1 heterodimer is a newly identified component of the interconnected pathways to combat the genotoxic effects of mass invasion/mobilization of transposons (Francis, 2016).

    Structure-function analysis of microRNA 3'-end trimming by Nibbler

    Nibbler (Nbr) is a 3'-to-5' exoribonuclease whose catalytic 3'-end trimming activity impacts microRNA (miRNA) and PIWI-interacting RNA (piRNA) biogenesis. This study reports on structural and functional studies to decipher the contributions of Nbr's N-terminal domain (NTD) and exonucleolytic domain (EXO) in miRNA 3'-end trimming. This study has solved the crystal structures of the NTD core and EXO domains of Nbr, both in the apo-state. The NTD-core domain of Aedes aegypti Nbr adopts a HEAT-like repeat scaffold with basic patches constituting an RNA-binding surface exhibiting a preference for binding double-strand RNA (dsRNA) over single-strand RNA (ssRNA). Structure-guided functional assays in Drosophila S2 cells confirmed a principal role of the NTD in exonucleolytic miRNA trimming, which depends on basic surface patches. Gain-of-function experiments revealed a potential role of the NTD in recruiting Nbr to Argonaute-bound small RNA substrates. The EXO domain of A. aegypti and Drosophila melanogaster Nbr adopt a mixed α/β-scaffold with a deep pocket lined by a DEDDy catalytic cleavage motif. Nbr's EXO domain exhibits Mn(2+)-dependent ssRNA-specific 3'-to-5' exoribonuclease activity. Modeling of a 3' terminal Uridine into the catalytic pocket of Nbr EXO indicates that 2'-O-methylation of the 3'-U would result in a steric clash with a tryptophan side chain, suggesting that 2'-O-methylation protects small RNAs from Nbr-mediated trimming. Overall, thee data establish that Nbr requires its NTD as a substrate recruitment platform to execute exonucleolytic miRNA maturation, catalyzed by the ribonuclease EXO domain (Xie, 2020).

    Antisense transcription of retrotransposons in Drosophila: The origin of endogenous small interfering RNA precursors

    To repress transposons and combat genomic instability, eukaryotes have evolved several small RNA mediated defense mechanisms. Specifically, in Drosophila somatic cells, endogenous small interfering (esi)RNAs suppress retrotransposon mobility. EsiRNAs are produced by Dicer-2 processing of double-stranded RNA precursors, yet the origins of these precursors are unknown. This study shows that most transposon families are transcribed in both the sense and antisense direction. LTR retrotransposons are generated from intra element transcription start sites with canonical RNA polymerase II promoters. Retrotransposon antisense transcripts were shown to be less polyadenylated than sense transcripts, which may promote nuclear retention of antisense transcripts and the double-stranded RNAs they form. Dicer-2 RNAi-depletion causes a decrease in the number of esiRNAs mapping to retrotransposons. These data support a model in which double-stranded RNA precursors are derived from convergent transcription and processed by Dicer-2 into esiRNAs that silence both sense and antisense retrotransposon transcripts. Reduction of sense retrotransposon transcripts potentially lowers element specific protein levels to prevent transposition. This mechanism preserves genomic integrity and is especially important for Drosophila fitness because mobile genetic elements are highly active (Russo, 2016).

    Natural variation of piRNA expression affects immunity to transposable elements

    In the Drosophila germline, transposable elements (TEs) are silenced by PIWI-interacting RNA (piRNA) that originate from distinct genomic regions termed piRNA clusters and are processed by PIWI-subfamily Argonaute proteins. This study explores the variation in the ability to restrain an alien TE in different Drosophila strains. The I-element is a retrotransposon involved in the phenomenon of I-R hybrid dysgenesis in Drosophila melanogaster. Genomes of R strains do not contain active I-elements, but harbour remnants of ancestral I-related elements. The permissivity to I-element activity of R females, called reactivity, varies considerably in natural R populations, indicating the existence of a strong natural polymorphism in defense systems targeting transposons. To reveal the nature of such polymorphisms, ovarian small RNAs between R strains with low and high reactivity were compared. It was shown that reactivity negatively correlates with the ancestral I-element-specific piRNA content. Analysis of piRNA clusters containing remnants of I-elements shows increased expression of the piRNA precursors and enrichment by the Heterochromatin Protein 1 homolog, Rhino, in weak R strains, which is in accordance with stronger piRNA expression by these regions. To explore the nature of the differences in piRNA production, weak and strong strains were analyzed and it was shown that the efficiency of maternal inheritance of piRNAs as well as the I-element copy number are very similar in both strains. At the same time, germline and somatic uni-strand piRNA clusters generate more piRNAs in strains with low reactivity, suggesting the relationship between the efficiency of primary piRNA production and variable response to TE invasions. The strength of adaptive genome defense is likely driven by naturally occurring polymorphisms in the rapidly evolving piRNA pathway proteins. The study hypothesizes that hyper-efficient piRNA production is contributing to elimination of a telomeric retrotransposon HeT-A, which was observed in one particular transposon-resistant R strain (Ryazansky, 2017).

    piRNA-mediated regulation of transposon alternative splicing in the soma and germ line

    Transposable elements can drive genome evolution, but their enhanced activity is detrimental to the host and therefore must be tightly regulated. The Piwi-interacting small RNA (piRNA) pathway is vital for the regulation of transposable elements, by inducing transcriptional silencing or post-transcriptional decay of mRNAs. This study shows that piRNAs and piRNA biogenesis components regulate precursor mRNA splicing of P-transposable element transcripts in vivo, leading to the production of the non-transposase-encoding mature mRNA isoform in Drosophila germ cells. Unexpectedly, it was shown that the piRNA pathway components do not act to reduce transcript levels of the P-element transposon during P-M hybrid dysgenesis, a syndrome that affects germline development in Drosophila. Instead, splicing regulation is mechanistically achieved together with piRNA-mediated changes to repressive chromatin states, and relies on the function of the Piwi-piRNA complex proteins Asterix (also known as Gtsf1) and Panoramix (Silencio), as well as Heterochromatin protein 1a [HP1a; encoded by Su(var)205]. Furthermore, this machinery, together with the piRNA Flamenco cluster, not only controls the accumulation of Gypsy retrotransposon transcripts but also regulates the splicing of Gypsy mRNAs in cultured ovarian somatic cells, a process required for the production of infectious particles that can lead to heritable transposition events. These findings identify splicing regulation as a new role and essential function for the Piwi pathway in protecting the genome against transposon mobility, and provide a model system for studying the role of chromatin structure in modulating alternative splicing during development (Teixeira, 2017).

    Hybrid dysgenesis is a syndrome that affects progeny in a non-reciprocal fashion, being normally restricted to the offspring of crosses in which males carry transposable elements but which females lack. In Drosophila, the dysgenic traits triggered by the P-element DNA transposon are restricted to the germ line and include chromosomal rearrangements, high rates of mutation, and sterility. The impairment is most prominent when hybrids are grown at higher temperatures, with adult dysgenic females being completely sterile at 29°C. Despite the severe phenotypes, little is known about the development of germ cells during P-M dysgenesis. To address this, germline development was characterized in the progeny obtained from reciprocal crosses between w1118 (P-element-devoid strain) and Harwich (P-element-containing strain) flies at 29°C. In non-dysgenic progeny, germline development progressed normally throughout embryonic and larval stages, leading to fertile adults. Although the development of dysgenic germline cells was not disturbed during embryogenesis, germ cells decreased in number during early larval stages, leading to animals with no germ cells by late larval stages. These results indicate that the detrimental effects elicited by P-element activity are triggered early on during primordial germ cell (PGC) development in dysgenic progeny, leading to premature germ cell death (Teixeira, 2017).

    Maternally deposited small RNAs cognate to the P-element are thought to provide the 'P-cytotype' by conferring the transgenerationally inherited ability to protect developing germ cells against P-elements. Small RNA-based transposon regulation is typically mediated by either transcriptional silencing or post-transcriptional clearance of mRNAs, both of which result in a decrease in the accumulation of transposon mRNA. To understand how maternally provided small RNAs control P-elements in germ cells, this study focused on embryonic PGCs sorted from 4- to 20-h-old embryos generated from reciprocal crosses between w1118 and Harwich strains. Surprisingly, the accumulation of P-element RNA as measured by quantitative reverse transcription PCR (RT-qPCR) showed no change in dysgenic PGCs when compared to non-dysgenic PGCs. This indicates that P-cytotype small RNAs exert their function by means other than regulating P-element mRNA levels (Teixeira, 2017).

    P-element activity relies on production of a functional P-element transposase protein, the expression of which requires precursor mRNA (pre-mRNA) splicing of three introns. To analyse P-element RNA splicing in germ cells during hybrid dysgenesis, primers were designed that specifically anneal to spliced mRNA transcripts. The accumulation of spliced forms for the first two introns (IVS1 and IVS2) did not show changes in dysgenic PGCs when compared to non-dysgenic PGCs. By contrast, the accumulation of spliced transcripts for the third intron (IVS3) was substantially increased in dysgenic germ cells. Given that the overall accumulation of P-element mRNA showed no changes, the results indicate that the maternally provided P-cytotype can negatively regulate P-element IVS3 splicing and therefore inhibits the production of functional P-transposase in germ cells (Teixeira, 2017).

    Analysis of publically available small RNA sequencing data from 0-2-h-old embryos laid by Harwich females indicated that two classes of small RNAs cognate to the P-element are maternally transmitted: small interfering RNAs (siRNAs, 20-22-nucleotides long) and piRNAs (23-29 nucleotides long). To test the role of distinct small RNA populations on P-element expression, mutants were analyzed uniquely affecting each small RNA biogenesis pathway in the Harwich background. Mutations that disrupt siRNA biogenesis components Dicer-2 (Dcr-2) and Argonaute 2 (AGO2), or mutations ablating components of the piRNA biogenesis pathway, such as the Argonautes piwi, aubergine (aub), and Argonaute 3 (AGO3), as well as the RNA helicase vasa (vas) and spindle E (spn-E), did not affect P-element mRNA accumulation in adult ovaries as measured by RT-qPCR. However, mutations that disrupted piRNA biogenesis, and not the siRNA pathway, led to a strong and specific increase in the accumulation of IVS3-spliced mRNAs. RNA sequencing (RNA-seq) analysis on poly(A)-selected RNAs from aub and piwi mutant adult ovaries confirmed the specific effect on IVS3 splicing. To examine transposon expression in tissue, RNA fluorescent in situ hybridization (FISH) was performed using probes specific for the P-element and for the Burdock retrotransposon, a classic target of the germline piRNA pathway. In mutants affecting piRNA biogenesis, increased abundance of Burdock RNA was readily observed in germline tissues, with most of the signal accumulating close to the oocyte. By contrast, no difference was detected in the P-element RNA FISH signal in piRNA biogenesis mutants compared to control. Nuclear RNA foci observed in nurse cells were of similar intensity and number regardless of the genotype, and cytoplasmic signal showed no detectable difference. Therefore, the results indicate that in germ cells, piRNAs specifically modulate IVS3 splicing. This regulation is reminiscent of the well-documented mechanism that restricts P-element activity to germline tissues, which involves the expression of a host-encoded RNA binding repressor protein that negatively regulates IVS3 splicing in somatic tissues (Teixeira, 2017).

    In somatic tissues, P-element alternative splicing regulation is mediated by the assembly of a splicing repressor complex on an exonic splicing silencer element directly upstream of IVS3. To test whether the P-element IVS3 and flanking exon sequences were sufficient to trigger the piRNA-mediated splicing regulation in germ cells, a transgenic reporter system for IVS3 splicing was used in which a heterologous promoter (Hsp83) drives the expression of an IVS3-lacZ-neo fusion mRNA specifically in the germ line. Using RT-qPCR, the F1 progeny from reciprocal crosses between w1118 and Harwich flies were analyzed in the presence of the hsp83-IVS3-lacZ-neo reporter. The fraction of spliced mRNAs produced from the transgenic reporter was substantially increased in dysgenic compared to non-dysgenic adult ovaries, in agreement with previously reported results. Most importantly, genetic experiments confirmed that the repression of IVS3 splicing in germ cells relies on piRNA biogenesis, as the splicing repression observed with this reporter in non-dysgenic progeny was specifically abolished in adult ovaries of aub and vas mutants (Teixeira, 2017).

    Mechanistically, piRNA-mediated splicing regulation may be achieved through direct action of piRNA complexes on target pre-mRNAs carrying the IVS3 sequence or indirectly by piRNA-mediated changes in chromatin states. Piwi-interacting proteins such as Asterix (Arx) and Panoramix (Panx) are dispensable for piRNA biogenesis but are essential for establishing Piwi-mediated chromatin changes, possibly by acting as a scaffold to recruit histone-modifying enzymes and chromatin-binding proteins to target loci. To test the role of these chromatin regulators on P-element splicing, germline-specific RNA interference (RNAi) knockdown experiments were performed in the Harwich background. Similar to what was observed for the piRNA biogenesis components, germline knockdown of Arx and Panx showed no change in the accumulation of P-element RNA, but a strong and specific effect on IVS3 splicing in adult ovaries. The same pattern on IVS3 splicing was observed in the germline knockdown of HP1a and Maelstrom (Mael), both of which act downstream of Piwi-mediated targeting to modulate chromatin structure. The same genetic requirement for Panx for IVS3 splicing control was also confirmed when using the transgenic IVS3 splicing reporter, further indicating that Piwi-mediated chromatin changes at the target locus are involved in IVS3 splicing regulation. At target loci, Piwi complexes are known to mediate the deposition of the classic heterochromatin mark histone H3 lysine 9 trimethylation (H3K9me3). To assess the effect of piRNA-targeting on P-element chromatin marks directly, H3K9me3 chromatin immunoprecipitation was performed followed by sequencing (ChIP-seq) or quantitative PCR on adult ovaries of progeny from reciprocal crosses between w1118 and Harwich strains (to avoid developmental defects, ChIP was performed on F1 progeny raised at 18°C. This analysis revealed a specific loss of global H3K9me3 levels over P-element insertions in dysgenic progeny when compared to non-dysgenic progeny (Teixeira, 2017).

    To analyse the chromatin structure at individual P-element insertions, DNA sequencing (DNA-seq) data was used to identify all euchromatic insertions in the Harwich strain, and RNA-seq analysis was used to define transcriptionally active insertions. At transcriptionally active P-element euchromatic insertions, the spreading of H3K9me3 into the flanking genomic regions was readily observed in non-dysgenic progeny, but was completely absent in dysgenic offspring. Similarly, a reduction in H3K9me3 modification levels was also observed over the IVS3 transgenic reporter in dysgenic progeny when compared to non-dysgenic progeny. Interestingly, euchromatic insertions with no evidence of transcriptional activity were devoid of an H3K9me3 signal in both non-dysgenic and dysgenic crosses, providing further evidence for a model initially suggested in yeast and more recently proposed for Drosophila and mammals, in which H3K9me3 deposition by piRNA complexes would require transcription of the target loci. Mechanistically different from the well-described somatic repression, the results uncovered the existence of an unexpected piRNA-mediated, chromatin-based mechanism regulating IVS3 alternative splicing in germ cells (Teixeira, 2017).

    To expand the analysis, the literature was searched for other cases of transposon splicing regulation. Drosophila Gypsy elements are retrotransposons that have retrovirus-like, infective capacity owing to their envelope (Env) protein. These elements are expressed in somatic ovarian cells, in which they are regulated by the flamenco locus, a well-known piRNA cluster that is a soma-specific source of antisense piRNAs cognate to Gypsy. Interestingly, it has been shown that mutations in flamenco not only elicited the accumulation of Gypsy RNA, but also modulated pre-mRNA splicing, favouring the production of the env mRNA and therefore germline infection. To test whether the piRNA pathway, in addition to its role in regulating the accumulation of Gypsy RNA, is also responsible for modulating the splicing of Gypsy elements in somatic tissues, publically available RNA-seq data from poly(A)-selected RNAs extracted from in vivo cultures of ovarian somatic cells (OSCs) was analyzed. The analysis indicates that piwi knockdown was sufficient to modulate Gypsy splicing, favouring the accumulation of env-encoding mRNA. In agreement with a chromatin-mediated regulation of alternative splicing, RNAi depletion of Arx, Panx, HP1a and Mael, as well as knockdown of the histone linker H1, was sufficient to favour Gypsy splicing, recapitulating the effect caused by Piwi depletion. Notably, this was also the case for the H3K9 methyltransferase Setdb1, but not for the H3K9 methyltransferases Su(var)3-9 and G9a, indicating specific genetic requirements. Taken together, the results indicate that the piRNA pathway, through its role in mediating changes in chromatin states, regulates the splicing of transposon pre-mRNAs in both somatic and germline tissues (Teixeira, 2017).

    Using P-M hybrid dysgenesis as a model, this study hasa uncovered splicing regulation elicited by chromatin changes as a previously unknown mechanism by which the piRNA pathway protects the genome from the detrimental effects of transposon activity. Splicing control at piRNA-target loci is likely to be mechanistically different from what has been observed for germline piRNA clusters given the low enrichment of the HP1 homologue Rhino (also known as HP1D) protein, which is required for piRNA cluster RNA processing, over the endogenous P-element insertions in the Harwich genome or over the transgenic IVS3 splicing reporter in non-dysgenic and dysgenic progeny (as measured by ChIP-qPCR). Because small RNA-based systems leading to chromatin mark changes at target loci are pervasive in eukaryotes, it is expected that this new type of targeted regulation is of importance in settings far beyond the scope of the piRNA pathway and Drosophila. Indeed, small RNA-guided DNA methylation over the LINE retrotransposon Karma was recently shown to modulate alternative splicing in oil palm, disrupting nearby gene expression and ultimately affecting crop yield. In this context, small RNA-based control of chromatin structure may be crucially important in genomes with a high content of intronic transposon insertions, such as the human genome, by providing a mechanism to suppress exonization of repeat elements. Although the means by which piRNA-mediated changes in chromatin states could regulate alternative splicing remain to be determined, it is tempting to speculate that piRNA pathway components do so by co-transcriptionally modulating interactions between RNA polymerase II and the spliceosome (Teixeira, 2017).

    Spontaneous gain of susceptibility suggests a novel mechanism of resistance to hybrid dysgenesis in Drosophila virilis

    Syndromes of hybrid dysgenesis (HD) have been critical for understanding of the transgenerational maintenance of genome stability by piRNA. HD in D. virilis represents a special case of HD since it includes simultaneous mobilization of a set of TEs that belong to different classes. The standard explanation for HD is that eggs of the responder strains lack an abundant pool of piRNAs corresponding to the asymmetric TE families transmitted solely by sperm. However, there are several strains of D. virilis that lack asymmetric TEs, but exhibit a "neutral" cytotype that confers resistance to HD. To characterize the mechanism of resistance to HD, a comparative analysis of the landscape of ovarian small RNAs was performed in strains that vary in their resistance to HD mediated sterility. It was demonstrated that resistance to HD cannot be solely explained by a maternal piRNA pool that matches the assemblage of TEs that likely cause HD. In support of this, a cytotype shift from neutral (N) to susceptible (M) was observed in a strain devoid of all major TEs implicated in HD. This shift occurred in the absence of significant change in TE copy number and expression of piRNAs homologous to asymmetric TEs. Instead, this shift is associated with a change in the chromatin profile of repeat sequences unlikely to be causative of paternal induction. Overall, these data suggest that resistance to TE-mediated sterility during HD may be achieved by mechanisms that are distinct from the canonical syndromes of HD (Funikov, 2018).

    Conserved noncoding elements influence the transposable element landscape in Drosophila

    Highly conserved noncoding elements (CNEs) constitute a significant proportion of the genomes of multicellular eukaryotes. The function of most CNEs remains elusive, but growing evidence indicates they are under some form of purifying selection. Noncoding regions in many species also harbor large numbers of transposable element (TE) insertions, which are typically lineage specific and depleted in exons because of their deleterious effects on gene function or expression. However, it is currently unknown whether the landscape of TE insertions in noncoding regions is random or influenced by purifying selection on CNEs. This study combine comparative and population genomic data in Drosophila melanogaster to show that abundance of TE insertions in intronic and intergenic CNEs is reduced relative to random expectation, supporting the idea that selective constraints on CNEs eliminate a proportion of TE insertions in noncoding regions. However, no evidence was found for differences in the allele frequency spectra for polymorphic TE insertions in CNEs versus those in unconstrained spacer regions, suggesting that the distribution of fitness effects acting on observable TE insertions is similar across different functional compartments in noncoding DNA. The results provide evidence that selective constraints on CNEs contribute to shaping the landscape of TE insertion in eukaryotic genomes, and provide further evidence that CNEs are indeed functionally constrained and not simply mutational cold spots (Manee, 2018).

    Double insertion of transposable elements provides a substrate for the evolution of satellite DNA

    Eukaryotic genomes are replete with repeated sequences in the form of transposable elements (TEs) dispersed across the genome or as satellite arrays, large stretches of tandemly repeated sequences. Many satellites clearly originated as TEs, but it is unclear how mobile genetic parasites can transform into megabase-sized tandem arrays. Comprehensive population genomic sampling is needed to determine the frequency and generative mechanisms of tandem TEs, at all stages from their initial formation to their subsequent expansion and maintenance as satellites. The best available population resources, short-read DNA sequences, are often considered to be of limited utility for analyzing repetitive DNA due to the challenge of mapping individual repeats to unique genomic locations. A new pipeline called ConTExt has been developed that demonstrates that paired-end Illumina data can be successfully leveraged to identify a wide range of structural variation within repetitive sequence, including tandem elements. By analyzing 85 genomes from five populations of Drosophila melanogaster, it was discovered that TEs commonly form tandem dimers. The results further suggest that insertion site preference is the major mechanism by which dimers arise and that, consequently, dimers form rapidly during periods of active transposition. This abundance of TE dimers has the potential to provide source material for future expansion into satellite arrays, and one such copy number expansion of the DNA transposon hobo was found to approximately 16 tandem copies in a single line. The very process that defines TEs-transposition-thus regularly generates sequences from which new satellites can arise (McGurk, 2018).

    Extensive exchange of transposable elements in the Drosophila pseudoobscura group

    As species diverge, so does their transposable element (TE) content. Within a genome, TE families may eventually become dormant due to host-silencing mechanisms, natural selection and the accumulation of inactive copies. The transmission of active copies from a TE families, both vertically and horizontally between species, can allow TEs to escape inactivation if it occurs often enough, as it may allow TEs to temporarily escape silencing in a new host. Thus, the contribution of horizontal exchange to TE persistence has been of increasing interest. This study annotated TEs in five species with sequenced genomes from the D. pseudoobscura species group, and curated a set of TE families found in these species. Compared to host genes, many TE families showed lower neutral divergence between species, consistent with recent transmission of TEs between species. Despite these transfers, there are differences in the TE content between species in the group. It is concluded that the TE content is highly dynamic in the D. pseudoobscura species group, frequently transferring between species, keeping TEs active. This result highlights how frequently transposable elements are transmitted between sympatric species and, despite these transfers, how rapidly species TE content can diverge (Hill, 2018).

    Stress affects the epigenetic marks added by natural transposable element insertions in Drosophila melanogaster

    Transposable elements are emerging as an important source of cis-acting regulatory sequences and epigenetic marks that could influence gene expression. However, few studies have dissected the role of specific transposable element insertions on epigenetic gene regulation. Bari-Jheh is a natural transposon that mediates resistance to oxidative stress by adding cis-regulatory sequences that affect expression of nearby genes. This work, integrated publicly available ChIP-seq and piRNA data with chromatin immunoprecipitation experiments to get a more comprehensive picture of Bari-Jheh molecular effects. Bari-Jheh was shown to be enriched for H3K9me3 in nonstress conditions, and for H3K9me3, H3K4me3 and H3K27me3 in oxidative stress conditions, which is consistent with expression changes in adjacent genes. It was further shown that under oxidative stress conditions, H3K4me3 and H3K9me3 spread to the promoter region of Jheh1 gene. Finally, another insertion of the Bari1 family was associated with increased H3K27me3 in oxidative stress conditions suggesting that Bari1 histone marks are copy-specific. It is concluded that besides adding cis-regulatory sequences, Bari-Jheh influences gene expression by affecting the local chromatin state (Guio, 2018).

    Epigenetic and chromosomal features drive transposon insertion in Drosophila melanogaster

    Transposons are mobile genetic elements prevalent in the genomes of most species. The distribution of transposons within a genome reflects the actions of two opposing processes: initial insertion site selection, and selective pressure from the host. By analyzing whole-genome sequencing data from transposon-activated Drosophila melanogaster, 43,316 de novo and 237 germline insertions were identified from four long-terminal-repeat (LTR) transposons, one LINE transposon (I-element), and one DNA transposon (P-element). All transposon types favored insertion into promoters de novo, but otherwise displayed distinct insertion patterns. De novo and germline P-element insertions preferred replication origins, often landing in a narrow region around transcription start sites and in regions of high chromatin accessibility. De novo LTR transposon insertions preferred regions with high H3K36me3, promoters and exons of active genes; within genes, LTR insertion frequency correlated with gene expression. De novo I-element insertion density increased with distance from the centromere. Germline I-element and LTR transposon insertions were depleted in promoters and exons, suggesting strong selective pressure to remove transposons from functional elements. Transposon movement is associated with genome evolution and disease; therefore, these results can improve understanding of genome and disease biology (Cao, 2023).

    Phenotypic and Transcriptomic Responses to Stress Differ According to Population Geography in an Invasive Species

    Adaptation to rapid environmental changes must occur within a short-time scale. In this context, studies of invasive species may provide insights into the underlying mechanisms of rapid adaptation as these species have repeatedly encountered and adapted to novel environmental conditions. This study investigated how invasive and noninvasive genotypes of Drosophila suzukii deal with oxidative stress at the phenotypic and molecular levels. Also studied was the impact of transposable element (TE) insertions on the gene expression in response to stress. Results show that flies from invasive areas (France and the United States) live longer in natural conditions than the ones from native Japanese areas. As expected, lifespan for all genotypes was significantly reduced following exposure to paraquat, but this reduction varied among genotypes (genotype-by-environment interaction) with invasive genotypes appearing more affected by exposure than noninvasive ones. A transcriptomic analysis of genotypes upon paraquat treatment detected many genes differentially expressed (DE). Although a small core set of genes were DE in all genotypes following paraquat exposure, much of the response of each genotype was unique. Moreover, it was shown that TEs were not activated after oxidative stress and DE genes were significantly depleted of TEs. In conclusion, it is likely that transcriptomic changes are involved in the rapid adaptation to local environments. This study provides new evidence that in the decade since the invasion from Asia, the sampled genotypes in Europe and the United States of D. suzukii diverged from the ones from the native area regarding their phenotypic and genomic response to oxidative stress (Marin, 2021).

    Subcellular localization and Egl-mediated transport of telomeric retrotransposon HeT-A ribonucleoprotein particles in the Drosophila germline and early embryogenesis

    The study of the telomeric complex in oogenesis and early development is important for understanding the mechanisms which maintain genome integrity. Telomeric transcripts are the key components of the telomeric complex and are essential for regulation of telomere function. The biogenesis of transcripts generated by the major Drosophila telomere repeat HeT-A in oogenesis and early development was studied with disrupted telomeric repeat silencing. In wild type ovaries, HeT-A expression is downregulated by the Piwi-interacting RNAs (piRNAs). By repressing piRNA pathway, this study showed that overexpressed HeT-A transcripts interact with their product, RNA-binding protein Gag-HeT-A, forming ribonucleoprotein particles (RNPs) during oogenesis and early embryonic development. Moreover, during early stages of oogenesis, in the nuclei of dividing cystoblasts, HeT-A RNP form spherical structures, which supposedly represent the retrotransposition complexes participating in telomere elongation. During the later stages of oogenesis, abundant HeT-A RNP are detected in the cytoplasm and nuclei of the nurse cells, as well as in the cytoplasm of the oocyte. Further on, it was demonstrate that HeT-A products co-localize with the transporter protein Egalitarian (Egl) both in wild type ovaries and upon piRNA loss. This finding suggests a role of Egl in the transportation of the HeT-A RNP to the oocyte using a dynein motor. Following germline piRNA depletion, abundant maternal HeT-A RNP interacts with Egl resulting in ectopic accumulation of Egl close to the centrosomes during the syncytial stage of embryogenesis. Given the essential role of Egl in the proper localization of numerous patterning mRNAs, it is suggested that its abnormal localization likely leads to impaired embryonic axis specification typical for piRNA pathway mutants (Kordyukova, 2018).

    Telomeric TART elements target the piRNA machinery in Drosophila

    Coevolution between transposable elements (TEs) and their hosts can be antagonistic, where TEs evolve to avoid silencing and the host responds by reestablishing TE suppression, or mutualistic, where TEs are co-opted to benefit their host. The TART-A TE functions as an important component of Drosophila telomeres but has also reportedly inserted into the Drosophila melanogaster nuclear export factor gene nxf2. Rather than inserting into nxf2, TART-A has actually captured a portion of nxf2 sequence. TART-A produces abundant Piwi-interacting small RNAs (piRNAs), some of which are antisense to the nxf2 transcript, and the TART-like region of nxf2 was found to be evolving rapidly. Furthermore, in D. melanogaster, TART-A is present at higher copy numbers, and nxf2 shows reduced expression, compared to the closely related species Drosophila simulans. It is proposed that capturing nxf2 sequence allowed TART-A to target the nxf2 gene for piRNA-mediated repression and that these 2 elements are engaged in antagonistic coevolution despite the fact that TART-A is serving a critical role for its host genome (Ellison, 2020).

    Paramutation-like Epigenetic Conversion by piRNA at the Telomere of Drosophila virilis

    First discovered in maize, paramutation is a phenomenon in which one allele can trigger an epigenetic conversion of an alternate allele. This conversion causes a genetically heterozygous individual to transmit alleles that are functionally the same, in apparent violation of Mendelian segregation. Studies over the past several decades have revealed a strong connection between mechanisms of genome defense against transposable elements by small RNA and the phenomenon of paramutation. For example, a system of paramutation in Drosophila melanogaster has been shown to be mediated by piRNAs, whose primary function is to silence transposable elements in the germline. This paper characterizes a second system of piRNA-mediated paramutation-like behavior at the telomere of Drosophila virilis. In Drosophila, telomeres are maintained by arrays of retrotransposons that are regulated by piRNAs. As a result, the telomere and sub-telomeric regions of the chromosome have unique regulatory and chromatin properties. Previous studies have shown that maternally deposited piRNAs derived from a sub-telomeric piRNA cluster can silence the sub-telomeric center divider gene of Drosophila virilis in trans. This paper shows that this silencing can also be maintained in the absence of the original silencing allele in a subsequent generation. The precise mechanism of this paramutation-like behavior may be explained by either the production of retrotransposon piRNAs that differ across strains or structural differences in the telomere. Altogether, these results show that the capacity for piRNAs to mediate paramutation in trans may depend on the local chromatin environment and proximity to the uniquely structured telomere regulated by piRNAs. This system promises to provide significant insights into the mechanisms of paramutation (Dorador, 2022).

    Drosophila small ovary gene is required for transposon silencing and heterochromatin organisation and ensures germline stem cell maintenance and differentiation

    Self-renewal and differentiation of stem cells is one of the fundamental biological phenomena relying on proper chromatin organisation. This study describes a novel chromatin regulator encoded by the Drosophila small ovary (sov) gene. sov was shown to be required in both the germline stem cells (GSCs) and the surrounding somatic niche cells to ensure GSC survival and differentiation. Sov maintains niche integrity and function by repressing transposon mobility, not only in the germline, but also in the soma. Protein interactome analysis of Sov revealed an interaction between Sov and HP1a. In the germ cell nuclei, Sov co-localises with HP1a, suggesting that Sov affects transposon repression as a component of the heterochromatin. In a position effect variegation assay, a dominant genetic interaction was found between sov and HP1a, indicating their functional cooperation in promoting the spread of heterochromatin. An in vivo tethering assay and FRAP analysis revealed that Sov enhances heterochromatin formation by supporting the recruitment of HP1a to the chromatin. A model is proposed in which sov maintains GSC niche integrity by regulating transposon silencing and heterochromatin formation (Jankovics, 2018).

    A robust transposon-endogenizing response from germline stem cells

    The heavy occupancy of transposons in the genome implies that existing organisms have survived from multiple, independent rounds of transposon invasions. However, how and which host cell types survive the initial wave of transposon invasion has remained unclear. This study shows that the germline stem cells can initiate a robust adaptive response that rapidly endogenizes invading P element transposons by activating the DNA damage checkpoint and piRNA production. Temperature modulates the P element activity in germline stem cells, establishing a powerful tool to trigger transposon hyper-activation. Facing vigorous invasion, Drosophila first shut down oogenesis and induce selective apoptosis. Interestingly, a robust adaptive response occurs in ovarian stem cells through activation of the DNA damage checkpoint. Within 4 days, the hosts amplify P element-silencing piRNAs, repair DNA damage, subdue the transposon, and reinitiate oogenesis. It is proposed that this robust adaptive response can bestow upon organisms the ability to survive recurrent transposon invasions throughout evolution (Moon, 2018).

    Considered as 'selfish DNA sequences,' transposons have heavily accumulated in the genome of nearly all organisms during evolution. Although capable of fueling genomic divergence, the transposon invasion process is disruptive to host cells and often severely impacts host fertility or even survival. Therefore, taming invading transposons is an essential and endless task for the host organism. In this study, by using P element invasion as a model, temperature shifting was established as a powerful tool to adjust the intensity of transposon invasion. By investigating the response from the Drosophila adult ovaries, in which P element activity and germ cell development can be measured in detail, a robust transposon-endogenizing mechanism from the germline stem cells was uncovered. Centered on the key DNA damage checkpoint component, Chk2, this robust adaptive response renders hosts the ability to permanently silence invading transposons within just 4 days (Moon, 2018).

    GFP::Vasa mobilization assay shows that the P element actively hops in germline stem cells. Does the P element also mobilize in other ovarian cells? Since nurse cells are polyploid and the developing oocytes are transcriptionally inactive, the current assay could not faithfully monitor P element mobilization in them. However, previous study shows that nurse cells express the protein P-element somatic inhibitor (PSI), which can block intron removal of P element transcripts and lead to the production of inactive transposases. Therefore, it is unlikely that P elements mobilize within developing egg chambers. As a type of DNA transposon, which employs the cut-and-paste mechanism for transposition, P elements cannot directly increase their copy number through mobilization. Instead, the propagation is likely achieved via homologous repair from the sister DNA strand during S-phase of the cell cycle. Hence, to amplify themselves during Drosophila oogenesis, perhaps P elements evolved to preferentially mobilize in the dividing germline stem cells but not in the developing oocytes, which are under cell cycle arrest (Moon, 2018).

    By investigating adult oogenesis of Drosophila, this study uncovered the Chk2-mediated adaptive response from germline stem cells upon P element transposon invasion (Moon, 2018).

    Interestingly, it appears that arrested germ cells are not equally capable of taming transposons, and Chk2 activation promotes adaptation by eliminating the cells with lower competency. Several lines of evidence support the occurrence of selective cell elimination. First, a significant increase in cell death was detected once P elements became hyperactive after the temperature shift. Second, although GFP-negative egg chambers directly connected to germaria at 25°C were occasionally observed from the GFP::Vasa mobilization assay, no GFP-negative cells were detected in later stage egg chambers at any time points. This suggests that the germ cells that maintained high P element activity, and were presumably less competent to adapt, were eliminated at early stages of oogenesis. Third, the number of new P element insertion events declined to 44% in recovered ovaries after adaptation. This dramatic decline indicates that only the stem cells that had lower transposition rates survived the selection. Therefore, it is tempting to speculate that not all germ cells are created equal and that in addition to germarial arrest, the Chk2-mediated DNA break checkpoint also has a role in selecting the survivors from P element invasion and promoting adaptation (Moon, 2018).

    In the surviving ovarian stem cells, Chk2-mediated oogenesis arrest provides a critical time window to propel piRNA generation from the paternally inherited clusters, initiating the amplification cycles for piRNA biogenesis. With at least two piRNA clusters containing P element sequences in the paternally inherited genome, invaded progeny are capable of generating P element-silencing piRNAs de novo. Although it is still unclear when the clusters become active during pre-adult development, it has been shown that the primordial germ cells in larval ovaries can already initiate de novo piRNA production. Consistently, low levels of piRNAs were detected corresponding to P element before adaptation. However, it appears that the amount of piRNAs produced at this stage is too scarce to silence invading P elements. Their activation results in sterility and triggers the Chk2-dependent acute adaptive response from germline stem cells. Subsequently, the Chk2-mediated arrest blocks differentiation, which would allow the newly produced P element-silencing piRNAs to quickly reach a concentration sufficient for Ping-Pong amplification. Finally, these newly produced piRNAs silence transposons at the post transcriptional level and also initiate transcriptional silencing (Moon, 2018).

    Besides promoting piRNA production, the arrest period also allows germ cells to repair DNA lesions before reinitiating oogenesis, thereby preventing the proliferation of cells with DNA damage and defective differentiation. Having the ability to repair damage and endogenize invading transposons in germline stem cells ensures permanent restoration of robust oogenesis and protection of all daughter cells from transposon activation (Moon, 2018).

    Pathogenic tau-induced piRNA depletion promotes neuronal death through transposable element dysregulation in neurodegenerative tauopathies

    Transposable elements, known colloquially as 'jumping genes', constitute approximately 45% of the human genome. Cells utilize epigenetic defenses to limit transposable element jumping, including formation of silencing heterochromatin and generation of piwi-interacting RNAs (piRNAs), small RNAs that facilitate clearance of transposable element transcripts. This study utilize Drosophila melanogaster and postmortem human brain samples to identify transposable element dysregulation as a key mediator of neuronal death in tauopathies, a group of neurodegenerative disorders that are pathologically characterized by deposits of tau protein in the brain. Mechanistically, it was found that heterochromatin decondensation and reduction of piwi and piRNAs drive transposable element dysregulation in tauopathy. A significant increase is reported in transcripts of the endogenous retrovirus class of transposable elements in human Alzheimer's disease and progressive supranuclear palsy, suggesting that transposable element dysregulation is conserved in human tauopathy. Taken together, these data identify heterochromatin decondensation, piwi and piRNA depletion and consequent transposable element dysregulation as a pharmacologically targetable, mechanistic driver of neurodegeneration in tauopathy (Sun, 2018).

    This study uncovered a therapeutically targetable mechanism whereby pathogenic tau drives neuronal death. Specifically, these studies identified dysregulation of transposable elements as a consequence of pathogenic tau and a driver of aberrant cell cycle activation in neurons and subsequent neuronal death. Genetic, dietary and pharmacological approaches were taken to reduce transposable element dysregulation and suppress tau-induced neurotoxicity in Drosophila. An unbiased transcriptomic approach was taken to extend these findings to postmortem human brains, and differentially expressed transposable elements were identified in Alzheimer's disease and progressive supranuclear palsy (Sun, 2018).

    Because the complexity and repetitive nature of transposable elements present challenges to RNA-seq analysis, which is associated with a greater frequency of false positives and negatives compared to analysis of canonical messenger RNAs, this study performed secondary validation of differentially expressed transposable element transcripts in tau transgenic Drosophila by NanoString. While NanoString data showed a similar expression trend for most of the transposable elements that were identified as differentially expressed in tau transgenic Drosophila by RNA-seq, some of the elements were not confirmed as differentially expressed by NanoString. These data reveal the limitations of each assay when analyzing transposable element transcripts and stress the importance of rigorous secondary validation. Since many members of the copia family are increased at the transcript level in both RNA-seq and NanoString analyses, it is speculated that gypsy-TRAP reporter activation is a result of copia insertion into the <i>ovo locus, rather than gypsy. Attempts to sequence de novo copia insertions within the <i>ovo locus in homogenates prepared from tau transgenic Drosophila heads resulted in a high frequency of mismatches, which is likely a result of the stochastic nature of transposable element insertion (Sun, 2018).

    According to current understanding, cells have two layers of defense against potentially deleterious transposable element activation: transposable element transcription is limited by heterochromatin-mediated silencing, and transposable element transcripts are cleared from the cell by piRNA-mediated degradation. Both mechanisms of transposable element suppression were compromised in tauopathy. It is speculated that tau-induced heterochromatin decondensation facilitates active transcription of transposable elements and that tau-induced piwi and piRNA reduction allows those transcripts to persist. While these results are consistent with the effects of heterochromatin decondensation and piwi reduction on transposable element expression that have been reported in the Drosophila germline, these studies reveal a previously undocumented role for heterochromatin- and piRNA-mediated transposable element silencing in the brain. On the basis of studies in the germline reporting a direct interaction between piwi and HP140 and a requirement for Rhino, a member of the HP1 subfamily, for piRNA production, it is possible that a direct interaction between piwi and HP1 is also required to silence transposable elements in the brain (Sun, 2018).

    Among upregulated transposable elements in human tauopathy, the human endogenous retrovirus (HERV) family, including HERV-K, was significantly over-represented. Elevated HERV-K transcripts are associated with amyotrophic lateral sclerosis (ALS)8 and many human cancers, including melanoma, breast cancer, germ cell tumors, renal cancer and ovarian cancer. A causal association between HERV-K and neuronal dysfunction has previously been established, as ectopic expression of HERV-K or the retroviral envelope protein that it encodes decreases synaptic activity and induces progressive motor dysfunction in mice. Antiretroviral reverse transcriptase inhibitors inhibit HERV-K activation in cultured cells and are now in clinical trials for the treatment of ALS. On the basis of the data presented in this study, reverse transcriptase inhibitors have significant potential as a therapeutic strategy for the treatment of neurodegenerative tauopathies, including Alzheimer's disease (Sun, 2018).

    The ability of flamenco loss-of-function mutations to enhance tau-induced neurotoxicity and the ability of piwi overexpression, dietary restriction and inhibition of reverse transcriptase to reduce transposable element dysregulation and suppress tau-induced neurotoxicity suggest that tau-induced transposable element dysregulation is deleterious to neuronal survival. In addition to the detrimental effects of transposable element jumping, double-stranded RNAs produced by transposable element transcripts, including HERVs, can trigger a type I interferon response through the innate immune system. In light of the HERV increase in human tauopathy and the involvement of the innate immune response as a disease-promoting mechanism in Alzheimer's disease, it is tempting to speculate that expression of endogenous retroviruses in human tauopathy contributes to neuroinflammation, in addition to promoting genomic instability. In future studies, it will be important to investigate a potential effect of transposable element activation on the innate immune response in the context of tauopathy (Sun, 2018).

    Population specific dynamics and selection patterns of transposable element insertions in European natural populations

    Transposable elements (TEs) are ubiquitous sequences in genomes of virtually all species. While TEs have been investigated for several decades, only recently has the opportunity appeared to study their genome-wide population dynamics. Most of the studies so far have been restricted either to the analysis of the insertions annotated in the reference genome or to the analysis of a limited number of populations. Taking advantage of the European Drosophila population genomics consortium (DrosEU) sequencing dataset, this study has identified and measured the dynamics of TEs in a large sample of European Drosophila melanogaster natural populations. The mobilome landscape is population specific and highly diverse depending on the TE family. In contrast with previous studies based on SNP variants, no geographical structure was observed for TE abundance or TE divergence in European populations. De novo individual insertions were also identified using two available programs and, as expected, most of the insertions were present at low frequencies. Nevertheless, a subset of TEs present at high frequencies was identified and located in genomic regions with a high recombination rate. These TEs are candidates for being the target of positive selection, although neutral processes should be discarded before reaching any conclusion on the type of selection acting on them. Finally, parallel patterns of association between the frequency of TE insertions and several geographical and temporal variables were found between European and North American populations, suggesting that TEs can be potentially implicated in the adaptation of populations across continents (Lerat, 2018).

    SQuIRE reveals locus-specific regulation of interspersed repeat expression

    Transposable elements (TEs) are interspersed repeat sequences that make up much of the human genome. Their expression has been implicated in development and disease. However, TE-derived RNA-seq reads are difficult to quantify. Past approaches have excluded these reads or aggregated RNA expression to subfamilies shared by similar TE copies, sacrificing quantitative accuracy or the genomic context necessary to understand the basis of TE transcription. As a result, the effects of TEs on gene expression and associated phenotypes are not well understood. This study presents Software for Quantifying Interspersed Repeat Expression (SQuIRE), the first RNA-seq analysis pipeline that provides a quantitative and locus-specific picture of TE expression. SQuIRE is an accurate and user-friendly tool that can be used for a variety of species. SQuIRE was applied to RNA-seq from normal mouse tissues and a Drosophila model of amyotrophic lateral sclerosis. In both model organisms, previously reported TE subfamily expression levels were recapitulated and locus-specific TE expression was revealed. Differences were identified in TE transcription patterns relating to transcript type, gene expression and RNA splicing that would be lost with other approaches using subfamily-level analyses. Altogether, these findings illustrate the importance of studying TE transcription with locus-level resolution (Yang, 2019).

    Dynamic Interactions between the genome and an endogenous retrovirus: Tirant in Drosophila simulans wild-type strains

    All genomes contain repeated sequences that are known as transposable elements (TEs). Among these are endogenous retroviruses (ERVs), which are sequences similar to retroviruses and are transmitted across generations from parent to progeny. These sequences are controlled in genomes through epigenetic mechanisms. At the center of the epigenetic control of TEs are small interfering RNAs of the piRNA class, which trigger heterochromatinization of TE sequences. The tirant ERV of Drosophila simulans displays intra-specific variability in copy numbers, insertion sites, and transcription levels, providing a well-suited model to study the dynamic relationship between a TE family and the host genome through epigenetic mechanisms. tirant transcript amounts and piRNA amounts are positively correlated in ovaries in normal conditions, unlike what was previously described following divergent crosses. In addition, tirant insertion polymorphism in the genomes of three D. simulans wild-type strains is described, revealing a limited number of insertions that may be associated with gene transcript level changes through heterochromatin spreading having phenotypic impacts. Taken together, these results participate in the understanding of the equilibrium between the host genome and its TEs (Fablet, 2019).

    Stress response, behavior, and development are shaped by transposable element-induced mutations in Drosophila

    Most of the current knowledge on the genetic basis of adaptive evolution is based on the analysis of single nucleotide polymorphisms (SNPs). Despite increasing evidence for their causal role, the contribution of structural variants to adaptive evolution remains largely unexplored. This work analyzed the population frequencies of 1,615 Transposable Element (TE) insertions annotated in the reference genome of Drosophila melanogaster, in 91 samples from 60 worldwide natural populations. A set of 300 polymorphic TEs were identified that are present at high population frequencies, and located in genomic regions with high recombination rate, where the efficiency of natural selection is high. The age and the length of these 300 TEs are consistent with relatively young and long insertions reaching high frequencies due to the action of positive selection. Besides, a set of 21 fixed TEs also likely to be adaptive was identified. Indeed evidence has been found of selection for 84 of these reference TE insertions. The analysis of the genes located nearby these 84 candidate adaptive insertions suggested that the functional response to selection is related with the GO categories of response to stimulus, behavior, and development. It was further shown that a subset of the candidate adaptive TEs affects expression of nearby genes, and five of them have already been linked to an ecologically relevant phenotypic effect. These results provide a more complete understanding of the genetic variation and the fitness-related traits relevant for adaptive evolution. Similar studies should help uncover the importance of TE-induced adaptive mutations in other species as well (Rech, 2019).

    Maelstrom represses canonical Polymerase II transcription within bi-directional piRNA clusters in Drosophila melanogaster

    In Drosophila, 23-30 nt long PIWI-interacting RNAs (piRNAs) direct the protein Piwi to silence germline transposon transcription. Most germline piRNAs derive from dual-strand piRNA clusters, heterochromatic transposon graveyards that are transcribed from both genomic strands. These piRNA sources are marked by the heterochromatin protein 1 homolog Rhino (Rhi), which facilitates their promoter-independent transcription, suppresses splicing, and inhibits transcriptional termination. This study reports that the protein Maelstrom (Mael) represses canonical, promoter-dependent transcription in dual-strand clusters, allowing Rhi to initiate piRNA precursor transcription. Mael also represses promoter-dependent transcription at sites outside clusters. At some loci, Mael repression requires the piRNA pathway, while at others, piRNAs play no role. It is proposed that by repressing canonical transcription of individual transposon mRNAs, Mael helps Rhi drive non-canonical transcription of piRNA precursors without generating mRNAs encoding transposon proteins (Chang, 2018).

    Fly piRNA clusters must solve a gene expression paradox. They record the ancient and contemporary exposure of the animal to transposon invasion, and this information must be copied into RNA in order to generate protective, anti-transposon piRNAs. However, recent transposon insertions retain the ability to produce mRNA encoding proteins required for their transposition. In flies, dual-strand piRNA clusters solve this paradox by using Rhi to initiate non-canonical transcription of unspliced RNA from both genomic strands, generating piRNA precursors, while repressing promoter-initiated, canonical transcription. These data suggest that Mael is required for this second process, allowing dual-strand piRNA clusters to safely generate piRNA precursor transcripts without risking production of transposon mRNAs. Within dual-strand clusters, Mael is likely guided to its targets by the piRNA pathway. However, the current analyses also predict that for some loci, Mael functions to repress canonical transcription in heterochromatin separately from the piRNA pathway, probably via one or more proteins that direct Mael to specific genomic sites (Chang, 2018).

    In maelM391/r20 ovaries, piRNAs mapping to dual-strand clusters decrease, despite a concomitant increase in canonical transcription from these same loci. The data suggest that in maelM391 mutants, Rhi-mediated non-canonical transcription, cluster transcript export, and ping-pong amplification become uncoupled. Perhaps, canonical transcription and Rhi-mediated non-canonical transcription compete for Pol II. Instead of fueling piRNA production, the canonical transcripts from dual-strand piRNA clusters produced in the absence of Mael are translated into protein. Mael therefore contributes to dual-strand cluster piRNA production by tipping the balance toward non-canonical transcription (Chang, 2018).

    The data indicate that in addition to relying on the piRNA pathway, Mael can also be guided to its targets by piRNA-independent mechanisms. Moreover, piRNA-dependent repression by Mael may be widespread outside of flies: although Drosophila melanogaster piwi is found only in the gonads, piwi is expressed broadly in the soma of most arthropods. In fact, 12 arthropods with somatic piRNAs also express mael in the soma, while 3 arthropods with no detectable piRNAs outside the gonads have low or undetectable somatic mael mRNA (Chang, 2018).

    In male mice, loss of MAEL also leads to loss of piRNAs, germline transposon derepression, and sterility. As in flies, loss of MAEL in mice does not trigger loss of heterochromatin: DNA methylation of L1 elements is unchanged (Chang, 2018).

    Because Mael is conserved from protists to humans, it is hypothesized that in different organisms Mael may be co-opted by different pathways to repress transcription of various targets. Prior studies suggest a model for how Mael confers repression. In protists, the MAEL domain was predicted to degrade RNA and may directly destroy nascent transcripts. In insects, the MAEL domain interacts with single-stranded RNA; it is speculated that fly Mael may have retained a role in destabilizing RNA. In this view, Mael may promote premature termination or degradation of nascent transcripts. In addition, because fly Mael has a partial HMG domain, it may also directly bind to DNA and repress transcription by preventing canonical core transcription factors from binding to promoters. Another possibility is that fly Mael may play a role in establishing or maintaining chromatin modifications not monitored in this study. Consistent with all of these possible mechanisms, non-canonical transcription mediated by Rhi is expected to be unaffected by Mael because the Rhi allows transcriptional initiation in dual-strand clusters without need for promoters and prevents degradation of unspliced piRNA precursor transcripts (Chang, 2018).

    Transposon silencing in the Drosophila female germline is essential for genome stability in progeny embryos

    The Piwi-interacting RNA pathway functions in transposon control in the germline of metazoans. The conserved RNA helicase Vasa is an essential Piwi-interacting RNA pathway component, but has additional important developmental functions. This study addresses the importance of Vasa-dependent transposon control in the Drosophila female germline and early embryos. Transient loss of vasa expression during early oogenesis leads to transposon up-regulation in supporting nurse cells of the fly egg-chamber. Elevated transposon levels have dramatic consequences, as de-repressed transposons accumulate in the oocyte where they cause DNA damage. Suppression of Chk2-mediated DNA damage signaling in vasa mutant females restores oogenesis and egg production. Damaged DNA and up-regulated transposons are transmitted from the mother to the embryos, which sustain severe nuclear defects and arrest development. These findings reveal that the Vasa-dependent protection against selfish genetic elements in the nuage of nurse cell is essential to prevent DNA damage-induced arrest of embryonic development (Durdevic, 2018).

    This study shows that a transient loss of vas expression during early oogenesis leads to up-regulation of transposon levels and compromised viability of progeny embryos. The observed embryonic lethality is because of DNA DSBs and nuclear damage that arise as a consequence of the elevated levels of transposon mRNAs and proteins, which are transmitted from the mother to the progeny. This study thus demonstrates that transposon silencing in the nurse cells is essential to prevent maternal transmission of transposons and DNA damage, protecting the progeny from harmful transposon-mediated mutagenic effects (Durdevic, 2018).

    The finding that suppression of Chk2-mediated DNA damage signaling in loss-of-function vas mutant flies restores oogenesis, and egg production demonstrates that Chk2 is epistatic to vas. However, hatching is severely impaired, because of the DNA damage sustained by the embryos. The defects displayed by vas, mnk double mutant embryos resembled those of PIWI (piwi, aub, and ago3) single and mnk; PIWI double mutant embryos. Earlier observation that inactivation of DNA damage signaling does not rescue the development of PIWI mutant embryos led to the assumption that PIWI proteins might have an essential role in early somatic development, independent of cell cycle checkpoint signaling. By tracing transposon protein and RNA levels and localization from the mother to the early embryos, it is suggested that, independent of Chk2 signaling, de-repressed transposons are responsible for nuclear damage and embryonic lethality. This study indicates that transposon insertions occur in the maternal genome where they cause DNA DSBs that together with transposon RNAs and proteins are passed on to the progeny embryos. Transposon activity and consequent DNA damage in the early syncytial embryo cause aberrant chromosome segregation, resulting in unequal distribution of the genetic material, nuclear damage and ultimately embryonic lethality. This study shows that early Drosophila embryos are defenseless against transposons and will succumb to their mobilization if the first line of protection against selfish genetic elements in the nuage of nurse cell fails (Durdevic, 2018).

    A recent study showed that in p53 mutants, transposon RNAs are up-regulated and accumulate at the posterior pole of the oocyte, without deleterious effects on oogenesis or embryogenesis. It is possible that the absence of pole plasm in vas mutants results in the release of the transposon products and their ectopic accumulation in the oocyte. Localization of transposons to the germ plasm may restrict their activity to the future germline and protect the embryo soma from transposon activity. Transposon-mediated mutagenesis in the germline would produce genetic variability, a phenomenon thought to play a role in the environmental adaptation and evolution of species. It would therefore be of interest to determine the role of pole plasm in transposon control in the future (Durdevic, 2018).

    Transposon up-regulation in the Drosophila female germline triggers a DNA damage-signaling cascade. In aub mutants, before their oogenesis arrest occurs, Chk2-mediated signaling leads to phosphorylation of Vasa, leading to impaired grk mRNA translation and embryonic axis specification. Considering the genetic interaction of vas and mnk (Chk2) and the fact that Vasa is phosphorylated in Chk2-dependent manner, it is tempting to speculate that phosphorylation of Vasa might stimulate piRNA biogenesis, reinforcing transposon silencing and thus minimizing transposon-induced DNA damage. The arrest of embryonic development as a first, and arrest of oogenesis as an ultimate response to DNA damage, thus, prevents the spreading of detrimental transposon-induced mutations to the next generation (Durdevic, 2018).

    The integrity of piRNA clusters is abolished by insulators in the Drosophila germline

    Piwi-interacting RNAs (piRNAs) control transposable element (TE) activity in the germline. piRNAs are produced from single-stranded precursors transcribed from distinct genomic loci, enriched by TE fragments and termed piRNA clusters. The specific chromatin organization and transcriptional regulation of Drosophila germline-specific piRNA clusters ensure transcription and processing of piRNA precursors. TEs harbour various regulatory elements that could affect piRNA cluster integrity. One of such elements is the suppressor-of-hairy-wing (Su(Hw))-mediated insulator, which is harboured in the retrotransposon gypsy. To understand how insulators contribute to piRNA cluster activity, the effects were studied of transgenes containing gypsy insulators on local organization of endogenous piRNA clusters. Transgene insertions interfere with piRNA precursor transcription, small RNA production and the formation of piRNA cluster-specific chromatin, a hallmark of which is Rhino, the germline homolog of the heterochromatin protein 1 (HP1). The mutations of Su(Hw) restored the integrity of piRNA clusters in transgenic strains. Surprisingly, Su(Hw) depletion enhanced the production of piRNAs by the domesticated telomeric retrotransposon TART, indicating that Su(Hw)-dependent elements protect TART transcripts from piRNA processing machinery in telomeres. A genome-wide analysis revealed that Su(Hw)-binding sites are depleted in endogenous germline piRNA clusters, suggesting that their functional integrity is under strict evolutionary constraints (Radion, 2019).

    Dynamics of transposable element invasions with piRNA clusters

    In mammals and invertebrates the proliferation of an invading transposable element (TE) is thought to be stopped by an insertion into a piRNA cluster. This study explored the dynamics of TE invasions under this trap model using computer simulations. piRNA clusters were found to confer a substantial benefit, effectively preventing extinction of host populations from a proliferation of deleterious TEs. TE invasions consists of three distinct phases: first the TE amplifies within the population, next TE proliferation is stopped by segregating cluster insertions and finally the TE is inactivated by fixation of a cluster insertion. Suppression by segregating cluster insertions is unstable and bursts of TE activity may yet occur. The transposition rate and the population size mostly influence the length of the phases but not the amount of TEs accumulating during an invasion. Solely the size of piRNA clusters was identified as a major factor influencing TE abundance. A single non-recombining cluster is more efficient in stopping invasions than clusters distributed over several chromosomes. Recombination among cluster sites makes it necessary that each diploid carries, on the average, four cluster insertions to stop an invasion. Surprisingly, negative selection in a model with piRNA clusters can lead to a novel equilibrium state, where TE copy numbers remain stable despite only some individuals in a population carrying a cluster insertion. In Drosophila melanogaster the trap model accounts for the abundance of TEs produced in the germline but fails to predict the abundance of TEs produced in the soma (Kofler, 2019).

    Cellular labeling of endogenous retrovirus replication (CLEVR) reveals de novo insertions of the gypsy retrotransposable element in cell culture and in both neurons and glial cells of aging fruit flies

    Evidence is rapidly mounting that transposable element (TE) expression and replication may impact biology more widely than previously thought. Investigation of the biological impact of mobile elements in somatic cells will be greatly facilitated by the use of donor elements that are engineered to report de novo events in vivo. In multicellular organisms, reporter constructs demonstrating engineered long interspersed nuclear element (LINE-1; L1) mobilization have been in use for quite some time, and strategies similar to L1 retrotransposition reporter assays have been developed to report replication of Ty1 elements in yeast and mouse intracisternal A particle (IAP) long terminal repeat (LTR) retrotransposons in cultivated cells. This study describes a novel approach termed cellular labeling of endogenous retrovirus replication (CLEVR), which reports replication of the gypsy element within specific cells in vivo in Drosophila. The gypsy-CLEVR reporter reveals gypsy replication both in cell culture and in individual neurons and glial cells of the aging adult fly.The gypsy-CLEVR replication rate is increased when the short interfering RNA (siRNA) silencing system is genetically disrupted. This CLEVR strategy makes use of universally conserved features of retroviruses and should be widely applicable to other LTR retrotransposons, endogenous retroviruses (ERVs), and exogenous retroviruses (Chang, 2019).

    Transposable element landscape in Drosophila populations selected for longevity

    Transposable elements (TEs) inflict numerous negative effects on health and fitness as they replicate by integrating into new regions of the host genome. Even though organisms employ powerful mechanisms to demobilize TEs, transposons gradually lose repression during aging. The rising TE activity causes genomic instability and was implicated in age-dependent neurodegenerative diseases, inflammation and the determination of lifespan. It is therefore conceivable that long-lived individuals have improved TE silencing mechanisms resulting in reduced TE expression relative to their shorter-lived counterparts and fewer genomic insertions. This study tested this hypothesis by performing the first genome-wide analysis of TE insertions and expression in populations of Drosophila melanogaster selected for longevity through late-life reproduction for 50-170 generations from four independent studies. Contrary to expectation, TE families were generally more abundant in long-lived populations compared to non-selected controls. Although simulations showed that this was not expected under neutrality, little evidence was found for selection driving TE abundance differences. Additional RNA-seq analysis revealed a tendency for reducing TE expression in selected populations, which might be more important for lifespan than regulating genomic insertions. Limited evidence was found of parallel selection on genes related to TE regulation and transposition. However, telomeric TEs were genomically and transcriptionally more abundant in long-lived flies, suggesting improved telomere maintenance as a promising TE-mediated mechanism for prolonging lifespan. The results provide a novel viewpoint indicating that reproduction at old age increases the opportunity of TEs to be passed on to the next generation with little impact on longevity (Fabian, 2021).

    Diverse families of transposable elements affect the transcriptional regulation of stress-response genes in Drosophila melanogaster

    Although transposable elements are an important source of regulatory variation, their genome-wide contribution to the transcriptional regulation of stress-response genes has not been studied yet. Stress is a major aspect of natural selection in the wild, leading to changes in the transcriptional regulation of a variety of genes that are often triggered by one or a few transcription factors. In this work, advantage was taken of the wealth of information available for Drosophila melanogaster and humans to analyze the role of transposable elements in six stress regulatory networks: immune, hypoxia, oxidative, xenobiotic, heat shock, and heavy metal. Transposable elements were found to be enriched for caudal, dorsal, HSF, and tango binding sites in D. melanogaster and for NFE2L2 binding sites in humans. Taking into account the D. melanogaster population frequencies of transposable elements with predicted binding motifs and/or binding sites, this study has shown that those containing three or more binding motifs/sites are more likely to be functional. For a representative subset of these TEs, in vivo transgenic reporter assays were performed in different stress conditions. Overall, the results showed that TEs are relevant contributors to the transcriptional regulation of stress-response genes (Villanueva-Canas, 2019).

    Trapping a somatic endogenous retrovirus into a germline piRNA cluster immunizes the germline against further invasion

    For species survival, the germline must faithfully transmit genetic information to the progeny. Transposable elements (TEs) constitute a significant threat to genome stability due to their mobility. In the metazoan germline, their mobilization is limited by a class of small RNAs called PIWI-interacting RNAs (piRNAs) produced by dedicated genomic loci called piRNA clusters. Although the piRNA pathway is an adaptive genomic immunity system, it remains unclear how the germline gains protection from a new transposon invasion. To address this question, Drosophila lines harboring a deletion within flamenco, a major piRNA cluster specifically expressed in somatic follicular cells. This deletion leads to derepression of the retrotransposon ZAM in the somatic follicular cells and subsequent germline genome invasion. In this mutant line, de novo production of sense and antisense ZAM-derived piRNAs were identified that display a germinal molecular signature. These piRNAs originated from a new ZAM insertion into a germline dual-strand piRNA cluster and silence ZAM expression specifically in germ cells. Finally, it was found that ZAM trapping in a germinal piRNA cluster is a frequent event that occurs early during the isolation of the mutant line. Transposons can hijack the host developmental process to propagate whenever their silencing is lost. This study shows that the germline can protect itself by trapping invading somatic-specific TEs into germline piRNA clusters. This is the first demonstration of "auto-immunization" of a germline endangered by mobilization of a surrounding somatic TE (Duc, 2019).

    Specialization of the Drosophila nuclear export family protein Nxf3 for piRNA precursor export

    The PIWI-interacting RNA (piRNA) pathway is a conserved small RNA-based immune system that protects animal germ cell genomes from the harmful effects of transposon mobilization. In Drosophila ovaries, most piRNAs originate from dual-strand clusters, which generate piRNAs from both genomic strands. Dual-strand clusters use noncanonical transcription mechanisms. Although transcribed by RNA polymerase II, cluster transcripts lack splicing signatures and poly(A) tails. mRNA processing is important for general mRNA export mediated by nuclear export factor 1 (Nxf1). Although UAP56, a component of the transcription and export complex, has been implicated in piRNA precursor export, it remains unknown how dual-strand cluster transcripts are specifically targeted for piRNA biogenesis by export from the nucleus to cytoplasmic processing centers. This study reports that dual-strand cluster transcript export requires CG13741/Bootlegger and the Drosophila nuclear export factor family protein Nxf3. Bootlegger is specifically recruited to piRNA clusters and in turn brings Nxf3. Nxf3 specifically binds to piRNA precursors and is essential for their export to piRNA biogenesis sites, a process that is critical for germline transposon silencing. These data shed light on how dual-strand clusters compensate for a lack of canonical features of mature mRNAs to be specifically exported via Nxf3, ensuring proper piRNA production (Kneuss, 2019).

    The evolutionary arms race between transposable elements and piRNAs in Drosophila melanogaster

    The piwi-interacting RNAs (piRNAs) are small non-coding RNAs that specifically repress transposable elements (TEs) in the germline of Drosophila. Despite expanding understanding of TE:piRNA interaction, whether there is an evolutionary arms race between TEs and piRNAs was unclear. This study examined the population genomics of TEs and piRNAs in the worldwide strains of D. melanogaster. By conducting a correlation analysis between TE contents and the abundance of piRNAs from ovaries of representative strains of D. melanogaster, positive correlations were found between TEs and piRNAs in six TE families. Simulations further highlight that TE activities and the strength of purifying selection against TEs are important factors shaping the interactions between TEs and piRNAs. These studies also suggest that the de novo generation of piRNAs is an important mechanism to repress the newly invaded TEs. These results revealed the existence of an evolutionary arms race between the copy numbers of TEs and the abundance of antisense piRNAs at the population level. Although the interactions between TEs and piRNAs are complex and many factors should be considered to impact their interaction dynamics, the results suggest the emergence, repression specificity and strength of piRNAs on TEs should be considered in studying the landscapes of TE insertions in Drosophila. These results deepen understanding of the interactions between piRNAs and TEs, and also provide novel insights into the nature of genomic conflicts of other forms (Luo, 2020).

    Viral infection impacts transposable element transcript amounts in Drosophila

    Transposable elements (TEs) are genomic parasites that are found in all genomes, some of which display sequence similarity to certain viruses. In insects, TEs are controlled by the Piwi-interacting small interfering RNA (piRNA) pathway in gonads, while the small interfering RNA (siRNA) pathway is dedicated to TE somatic control and defense against viruses. So far, these two small interfering RNA pathways are considered to involve distinct molecular effectors and are described as independent. Using Sindbis virus (SINV) in Drosophila, this study shows that viral infections affect TE transcript amounts via modulations of the piRNA and siRNA repertoires, with the clearest effects in somatic tissues. These results suggest that viral acute or chronic infections may impact TE activity and, thus, the tempo of genetic diversification. In addition, these results deserve further evolutionary considerations regarding potential benefits to the host, the virus, or the TEs (Roy, 2020).

    The Hsp70 chaperone is a major player in stress-induced transposable element activation

    Previous studies have shown that heat shock stress may activate transposable elements (TEs) in Drosophila and other organisms. Such an effect depends on the disruption of a chaperone complex that is normally involved in biogenesis of Piwi-interacting RNAs (piRNAs), the largest class of germline-enriched small noncoding RNAs implicated in the epigenetic silencing of TEs. However, a satisfying picture of how chaperones could be involved in repressing TEs in germ cells is still unknown. This study shows that, in Drosophila, heat shock stress increases the expression of TEs at a posttranscriptional level by affecting piRNA biogenesis through the action of the inducible chaperone Hsp70. Stress-induced TE activation is triggered by an interaction of Hsp70 with the Hsc70-Hsp90 complex and other factors all involved in piRNA biogenesis in both ovaries and testes. Such interaction induces a displacement of all such factors to the lysosomes, resulting in a functional collapse of piRNA biogenesis. This mechanism has clear evolutionary implications. In the presence of drastic environmental changes, Hsp70 plays a key dual role in increasing both the survival probability of individuals and the genetic variability in their germ cells. The consequent increase of genetic variation in a population potentiates evolutionary plasticity and evolvability (Cappucci, 2019).

    Evolutionary history and classification of Micropia retroelements in Drosophilidae species

    Transposable elements (TEs) have the main role in shaping the evolution of genomes and host species, contributing to the creation of new genes and promoting rearrangements frequently associated with new regulatory networks. Micropia belongs to the Ty3/Gypsy group of long terminal repeats (LTR) retroelements and comprises one of the least studied Drosophila transposable elements. This study assessed the evolutionary history of Micropia within Drosophilidae, while trying to assist in the classification of this TE. At first, searches were performed of Micropia presence in the genome of natural populations from several species. Then, based on searches within online genomic databases, Micropia-like sequences were retrieved from the genomes of distinct Drosophilidae species. The knowledge of Micropia distribution within Drosophila species. The Micropia retroelements consist of an array of divergent sequences, which was subdivided into 20 subfamilies. Even so, a patchy distribution of Micropia sequences within the Drosophilidae phylogeny could be identified, with incongruences between the species phylogeny and the Micropia phylogeny. Comparing the pairwise synonymous distance (dS) values between Micropia and three host nuclear sequences, several cases were found of unexpectedly high levels of similarity between Micropia sequences in divergent species. All these findings provide a hypothesis to the evolution of Micropia within Drosophilidae, which include several events of vertical and horizontal transposon transmission, associated with ancestral polymorphisms and recurrent Micropia sequences diversification (Cordeiro, 2019).

    Patterns of piRNA Regulation in Drosophila Revealed through Transposable Element Clade Inference

    Transposable elements (TEs) are self-replicating "genetic parasites" ubiquitous to eukaryotic genomes. In addition to conflict between TEs and their host genomes, TEs of the same family are in competition with each other. They compete for the same genomic niches while experiencing the same regime of copy-number selection. This suggests that competition among TEs may favor the emergence of new variants that can outcompete their ancestral forms. To investigate the sequence evolution of TEs, a method was developed to infer clades: collections of TEs that share SNP variants and represent distinct TE family lineages. This method was applied to a panel of 85 Drosophila melanogaster genomes and found that the genetic variation of several TE families shows significant population structure that arises from the population-specific expansions of single clades. Population genetic theory was used to classify these clades into younger versus older clades, and it was found that younger clades are associated with a greater abundance was found of sense and antisense piRNAs per copy than older ones. Further, the abundance of younger, but not older clades, is positively correlated with antisense piRNA production, suggesting a general pattern where hosts preferentially produce antisense piRNAs from recently active TE variants. Together these findings suggest a pattern whereby new TE variants arise by mutation and then increase in copy number, followed by the host producing antisense piRNAs that may be used to silence these emerging variants (Said, 2022).

    Requirements for multivalent Yb body assembly in transposon silencing in Drosophila

    Female sterile (1) Yb (Yb) is a primary component of Yb bodies, perinuclear foci considered to be the site of PIWI-interacting RNA (piRNA) biogenesis in Drosophila ovarian somatic cells (OSCs). Yb consists of three domains: Helicase C-terminal (Hel-C), RNA helicase, and extended Tudor (eTud) domains. Previous work has shown that the RNA helicase domain is necessary for Yb-RNA interaction, Yb body formation, and piRNA biogenesis. This study investigated the functions of Hel-C and eTud and revealed that Hel-C is dedicated to Yb-Yb homotypic interaction, while eTud is necessary for Yb-RNA association, as is the RNA helicase domain. All of these domains are indispensable for Yb body formation and transposon-repressing piRNA production. Strikingly, however, genic piRNAs unrelated to transposon silencing are produced in OSCs where Yb bodies are disassembled. It was also revealed that Yb bodies are liquid-like multivalent condensates whose assembly depends on Yb-Yb homotypic interaction and Yb binding particularly with flamenco RNA transcripts, the source of transposon-repressing piRNAs. New insights into Yb body assembly and biological relevance of Yb bodies in transposon silencing have emerged (Hirakata, 2019).

    PIWI-interacting RNAs (piRNAs) are engaged in an arms race with mobile transposons to maintain the genetic integrity of the germline. In Drosophila ovarian somatic cells (OSCs), transposon-repressing piRNAs arise nearly exclusively from the uni-strand piRNA cluster flamenco (flam), located at the pericentromeric region of the X chromosome. Upon transcription, the flam RNA transcripts undergo partial splicing and are exported to the cytoplasm, where they accumulate into the perinuclear structures Flam bodies/Dot COM for further processing. The flam-piRNA precursors may also be stored at nuclear Dot COM prior to nuclear export (Hirakata, 2019).

    piRNA processing in OSCs occurs in a manner dependent on a number of piRNA factors. Upon processing, mature piRNAs form piRNA-induced silencing complexes (piRISCs) with Piwi, a member of the PIWI protein family in Drosophila. The other two PIWI members, Aubergine and Ago3, are not expressed in OSCs. Piwi-piRISCs are then imported to the nucleus by Importin &alpha, where they repress transposons cotranscriptionally with multiple cofactors such as Gtsf1/Asterix, Maelstrom, Histone H1, HP1a, Eggless/dSetDB1, and Panoramix/Silencio (Hirakata, 2019).

    Yb bodies are gonadal soma-specific membraneless organelles to which piRNA biogenesis factors, female sterile (1) Yb (Yb), Armitage (Armi), Sister of Yb (SoYb), Vreteno (Vret), and Shutdown (Shu) are localized. Loss of these factors abrogates piRNA biogenesis; thus, Yb bodies are considered to be the site of piRNA production. Other processing factors, Zucchini (Zuc), Gasz, and Minotaur (Mino), are not localized to Yb bodies but are anchored on the surface of mitochondria through their own transmembrane signals. Yb bodies tend to be surrounded by mitochondria and adjacent to Flam bodies. This spatial arrangement of the organelles locally concentrates piRNA-processing factors and precursor RNAs, facilitating piRNA biogenesis and Piwi-piRISC formation. The hierarchy of Yb body assembly has previously been examined. However, a comprehensive and systematic analysis including SoYb has not been performed (Hirakata, 2019).

    Yb contains three functional domains: Helicase C-terminal (Hel-C), RNA helicase, and extended Tud (eTud) domains. The RNA helicase domain consists of P-loop NTPase and Hel-C domains. Previous work has shown that alteration of Gln399 or Asp537 in the RNA helicase domain to alanine severely reduced the RNA-binding activity of Yb (Murota, 2014). Both mutants Q399A and D537A failed to form Yb bodies and barely restored the piRNA biogenesis and transposon silencing abrogated by loss of endogenous Yb in OSCs. This suggested that the association of Yb with piRNA precursors via the RNA helicase domain is essential for Yb body formation and piRNA biogenesis (Hirakata, 2019).

    The cis-elements that drive piRNA biogenesis were identified in flam transcripts and genic piRNA sources such as traffic jam (tj) mRNAs. Enforced tagging of the cis-element to the 5' end, but not the 3' end, of arbitrary RNAs induced artificial piRNA production from the downstream regions, which repressed genes highly complementary to the piRNAs. Yb acts as the trans-acting factor, binding to the cis-element and triggering piRNA biogenesis. RNA binding of Yb also determines the regions from which piRNAs are produced. Yb is not involved in the transcription of flam and possibly genic piRNA sources (Hirakata, 2019).

    Both the Hel-C and the eTud domains of Yb are necessary for piwi and hedgehog expression in ovarian cap cells and for germarium development. However, their molecular functions in piRNA biogenesis remain elusive. This study produced two Yb deletion mutants, ΔHel-C and ΔeTud, and analyzed them biochemically, revealing that Hel-C is necessary for self-association of Yb while eTud is essential for Yb to interact with piRNA precursors and Armi. Armi is required for localizing the SoYb-Vret heterodimer to Yb bodies, and all three domains of Yb are necessary for Yb body assembly and transposon silencing. The production of flam-piRNAs was severely impaired by the loss of Hel-C and eTud, leading to transposon derepression. Strikingly, however, non-transposon-repressing genic piRNAs, whose functions remain rather unclear, were expressed in OSCs lacking Hel-C, but not those lacking eTud. Similar phenotypes were observed in flam mutants where the transcription of flam was impaired. It was also found that Yb bodies had properties compatible with liquid-like phase separation. In summary, Yb bodies are multivalent RNA-protein condensates whose assembly depends both on homotypic interaction of Yb through the Hel-C domain, and on Yb binding flam RNAs. Yb body formation is not absolutely required for piRNA biogenesis. However, without Yb bodies, transposon-repressing piRNAs are scarcely produced, resulting in the failure of transposon repression. The biological relevance of Yb/flam-driven Yb body assembly in the somatic piRNA pathway has now emerged (Hirakata, 2019).

    This study has determined the hierarchy of protein components in Yb body assembly and the distinct functions of the Hel-C and eTud domains of Yb. It was also determined how all three domains of Yb, the Hel-C, RNA helicase, and eTud domains, cooperatively function in Yb body formation, piRNA biogenesis, and transposon silencing. A previous genetic study claimed that all of the regions of Yb are essential for fertility (Szakmary, 2009), but the link between the different regions and fertility remained unclear. This study successfully uncovered the molecular bases of this link (Hirakata, 2019).

    Surprisingly, piRNA biogenesis in OSCs took place even without Yb body assembly. However, the products are mostly genic piRNAs, which are considered to be 'junk' because they cannot target transposons. Yb bodies were originally thought to be the site of piRNA biogenesis. However, the current study provides new insight; that is, Yb bodies are the elaborative system specialized for producing piRNAs functional in transposon silencing (Hirakata, 2019).

    It was noticed that recombinant Yb in vitro oligomerizes without RNAs. Thus, the idea is favored that 'minimum size Yb body units' assembling under the detection levels of regular microscopy play a role in genic piRNA production in normal OSCs and also in flamKG ovaries. In normal OSCs, genic piRNA precursors may also be sequestered into Yb bodies together with flam RNAs. Determination of the sequences of RNAs residing in Yb bodies in normal OSCs or in the ovaries should elucidate this (Hirakata, 2019).

    Stress granules are generated through liquid-liquid phase separation (LLPS), where longer transcripts are more strongly enriched in the granules. This was attributed to the fact that longer RNAs contain higher numbers of RNA-protein binding sites. In fact, the flam RNAs contained a great number of Yb binding sites throughout their entirety, while genic piRNA sources contained far fewer Yb binding sites, which were normally limited in the 3' UTRs. This difference may be the cause of the enrichment of flam, but no other, RNAs in Yb bodies. However, it was difficult to confirm this in this study because the lengths of flam RNA isoforms per se have not been determined yet. It is imagined that sequestering RNAs to Yb bodies in a manner largely dependent on the number of Yb binding sites would be beneficial for cells, as this would avoid the background accumulation of incorrect RNAs to Yb bodies because the incorrect RNAs would be unlikely to have many Yb binding sites (Hirakata, 2019).

    The data suggested that the eTud domain of Yb is involved in both Armi binding and RNA binding. Even though the eTud domain generally recognizes symmetrically dimethylated arginine (sDMA) residue of the target protein, Armi lacks motifs for such modification (GRG, ARG, GRA). In addition, the eTud domain of Yb lacks aromatic residues that are responsible for the recognition of symmetrically dimethylated arginine (sDMA). Therefore, Yb-Armi interaction appears to be mediated by sDMA-independent eTud-protein interaction, as is the case for interaction between BmTDRD12 and Siwi. eTud domain is composed of a Tudor domain and a nuclease-like structure. Because of the nuclease-like folding and the computational prediction that the surface of the eTud domain of Yb is rather basic, it is speculated that the eTud domain itself may have the RNA-binding activity (Hirakata, 2019).

    It is clear that the Hel-C domain is important in Yb body formation. However, the Hel-C domain was predicted to be neither an IDR nor a prion-like domain. Nonetheless, IDRs also confer flexibility on whole protein structures. Therefore, in the case of Yb, having an IDR between the N-terminal Hel-C and the RNA helicase domain may be beneficial for the protein to form multivalent condensates with flam RNAs (Hirakata, 2019).

    Yb bodies are often observed to be surrounded by mitochondria, and piRNA biogenesis most likely occurs at the interface of the two organelles where the piRNA factors and RNA substrates concentrate. Providing 'liquid-like condensate' properties to Yb bodies would be beneficial for this, allowing increased areas of attachment between the two organelles. The mechanism by which the two organelles are attached is not yet known. Gene screening combined with measuring the distance between Yb bodies and mitochondria may resolve this issue (Hirakata, 2019).

    Yb bodies are detected not only in the ovaries but also in the testes, where Yb seems to play a role in regulating germline stem cell self-renewal, as it does in the ovaries. However, yb null mutant males are fertile; hence, Yb may not be absolutely required in spermatogenesis. This suggests that piRNA biogenesis in males is capable of bypassing Yb. Yb is a member of the TDRD12 family of proteins, in which SoYb is involved. Other species express a counterpart of SoYb but lack one of Yb. The current study clarified that the functions of Yb in Drosophila OSCs are as follows: (1) the selection of piRNA precursors, (2) the induction of piRNA biogenesis, and (3) the facilitation of production of transposon-repressing piRNAs by assembling Yb bodies. Without these functions, the piRNA pathway would be defective. Thus, identification of Yb counterparts in fly testes and in the gonads of other species is anticipated (Hirakata, 2019).

    High frequency of horizontal transfer in Jockey families (LINE order) of drosophilids

    The use of large-scale genomic analyses has resulted in an improvement of transposable element sampling and a significant increase in the number of reported HTT (horizontal transfer of transposable elements) events by expanding the sampling of transposable element sequences in general and of specific families of these elements in particular, which were previously poorly sampled. This study investigated the occurrence of HTT events in a group of elements that, until recently, were uncommon among the HTT records in Drosophila - the Jockey elements, members of the LINE (long interspersed nuclear element) order of non-LTR (long terminal repeat) retrotransposons. The sequences of 111 Jockey families deposited in Repbase that met the criteria of the analysis were used to identify Jockey sequences in 48 genomes of Drosophilidae (genus Drosophila, subgenus Sophophora: melanogaster, obscura and willistoni groups; subgenus Drosophila: immigrans, melanica, repleta, robusta, virilis and grimshawi groups; subgenus Dorsilopha: busckii group; genus/subgenus Zaprionus and genus Scaptodrosophila). Phylogenetic analyses revealed 72 Jockey families in 41 genomes. Combined analyses revealed 15 potential HTT events between species belonging to different genera and species groups of Drosophilidae, providing evidence for the flow of genetic material favoured by the spatio-temporal sharing of these species present in the Palaeartic or Afrotropical region. These results provide phylogenetic, biogeographic and temporal evidence of horizontal transfers of the Jockey elements, increase the number of rare records of HTT in specific families of LINE elements, increase the number of known occurrences of these events, and enable a broad understanding of the evolutionary dynamics of these elements and the host species (Tambones, 2019).

    Nanopore sequencing and Hi-C scaffolding provide insight into the evolutionary dynamics of transposable elements and piRNA production in wild strains of Drosophila melanogaster

    Illumina sequencing has allowed for population-level surveys of transposable element (TE) polymorphism via split alignment approaches, which has provided important insight into the population dynamics of TEs. However, such approaches are not able to identify insertions of uncharacterized TEs, nor can they assemble the full sequence of inserted elements. This study used nanopore sequencing and Hi-C scaffolding to produce de novo genome assemblies for two wild strains of Drosophila melanogaster from the Drosophila Genetic Reference Panel (DGRP). Ovarian piRNA populations and Illumina split-read TE insertion profiles have been previously produced for both strains. Nanopore sequencing with Hi-C scaffolding produces highly contiguous, chromosome-length scaffolds, and this study identified hundreds of TE insertions that were missed by Illumina-based methods, including a novel micropia-like element that has recently invaded the DGRP population. Hundreds of piRNA-producing loci were found that are specific to each strain. Some of these loci are created by strain-specific TE insertions, while others appear to be epigenetically controlled. These results suggest that Illumina approaches reveal only a portion of the repetitive sequence landscape of eukaryotic genomes and that population-level resequencing using long reads is likely to provide novel insight into the evolutionary dynamics of repetitive elements (Ellison, 2019).

    Transposon activity, local duplications and propagation of structural variants across haplotypes drive the evolution of the Drosophila S2 cell line

    Immortalized cell lines are widely used model systems whose genomes are often highly rearranged and polyploid. However, their genome structure is seldom deciphered and is thus not accounted for during analyses. This study therefore used linked short- and long-read sequencing to perform haplotype-level reconstruction of the genome of a Drosophila melanogaster cell line (S2-DRSC) with a complex genome structure. Using a custom implementation (that is designed to use ultra-long reads in complex genomes with nested rearrangements) to call structural variants (SVs), it was found that the most common SV was repetitive sequence insertion or deletion (> 80% of SVs), with Gypsy retrotransposon insertions dominating. The second most common SV was local sequence duplication. SNPs and other SVs were rarer, but several large chromosomal translocations and mitochondrial genome insertions were observed. Haplotypes were highly similar at the nucleotide level but structurally very different. Insertion SVs existed at various haplotype frequencies and were unlinked on chromosomes, demonstrating that haplotypes have different structures and suggesting the existence of a mechanism that allows SVs to propagate across haplotypes. Finally, using public short-read data, it was found that transposable element insertions and local duplications are common in other D. melanogaster cell lines. It is concluded that The S2-DRSC cell line evolved through retrotransposon activity and vast local sequence duplications, that are hypothesized to be the products of DNA re-replication events. Additionally, mutations can propagate across haplotypes (possibly explained by mitotic recombination), which enables fine-tuning of mutational impact and prevents accumulation of deleterious events, an inherent problem of clonal reproduction. It is concluded that traditional linear homozygous genome representation conceals the complexity when dealing with rearranged and heterozygous clonal cells (Lewerentz, 2022).

    Dynamics and Impacts of Transposable Element Proliferation in the Drosophila nasuta Species Group Radiation

    Transposable element (TE) mobilization is a constant threat to genome integrity. Eukaryotic organisms have evolved robust defensive mechanisms to suppress their activity, yet TEs can escape suppression and proliferate, creating strong selective pressure for host defense to adapt. This genomic conflict fuels a never-ending arms race that drives the rapid evolution of TEs and recurrent positive selection of genes involved in host defense; the latter has been shown to contribute to postzygotic hybrid incompatibility. However, how TE proliferation impacts genome and regulatory divergence remains poorly understood. This paper reports the highly complete and contiguous genome assemblies of seven closely related Drosophila species that belong to the nasuta species group-a poorly studied group of flies that radiated in the last 2 My. A high-quality de novo TE library was constructed and germline RNA-seq data was gathered, allowing a comprehensive annotation, and TE insertion patterns were compared between the species, and the evolutionary forces controlling their spread were inferred. A strong negative association was found between TE insertion frequency and expression of genes nearby; this likely reflects survivor bias from reduced fitness impact of TEs inserting near lowly expressed, nonessential genes, with limited TE-induced epigenetic silencing. Phylogenetic analyses of insertions of 147 TE families reveal that 53% of them show recent amplification in at least one species. The most highly amplified TE is a nonautonomous DNA element (Drosophila INterspersed Element; DINE) which has gone through multiple bouts of expansions with thousands of full-length copies littered throughout each genome. Across all TEs, this study found that TEs expansions are significantly associated with high expression in the expanded species consistent with suppression escape. Thus, whereas horizontal transfer followed by the invasion of a naive genome has been highlighted to explain the long-term survival of TEs, this analysis suggests that evasion of host suppression of resident TEs is a major strategy to persist over evolutionary times. Altogether, these results shed light on the heterogenous and context-dependent nature in which TEs affect gene regulation and the dynamics of rampant TE proliferation amidst a recently radiated species group (Wei, 2022).

    Transposable element accumulation drives size differences among polymorphic Y Chromosomes in Drosophila

    Y Chromosomes of many species are gene poor and show low levels of nucleotide variation, yet they often display high amounts of structural diversity. Dobzhansky cataloged several morphologically distinct Y Chromosomes in Drosophila pseudoobscura that differ in size and shape, but the molecular causes of their large size differences are unclear. This study used cytogenetics and long-read sequencing to study the sequence content of polymorphic Y Chromosomes in D. pseudoobscura. The Y Chromosomes were shown to differ almost twofold in size, ranging from 30 to 60 Mb. Most of this size difference is caused by a handful of active transposable elements (TEs) that have recently expanded on the largest Y Chromosome, with different elements being responsible for Y expansion on differently sized D. pseudoobscura Y's. Y Chromosomes differ in their heterochromatin enrichment and expression of Y-enriched TEs, and also influence expression of dozens of autosomal and X-linked genes. The same helitron element that showed the most drastic amplification on the largest Y in D. pseudoobscura independently amplified on a polymorphic large Y Chromosome in Drosophila affinis, suggesting that some TEs are inherently more prone to become deregulated on Y Chromosomes (Nguyen, 2022).

    Special vulnerability of somatic niche cells to transposable element activation in Drosophila larval ovaries

    In the Drosophila ovary, somatic escort cells (ECs) form a niche that promotes differentiation of germline stem cell (GSC) progeny. The piRNA (Piwi-interacting RNA) pathway, which represses transposable elements (TEs), is required in ECs to prevent the accumulation of undifferentiated germ cells (germline tumor phenotype). The soma-specific piRNA cluster flamenco (flam) produces a substantial part of somatic piRNAs. This study characterized the biological effects of somatic TE activation on germ cell differentiation in flam mutants. The choice between normal and tumorous phenotypes of flam mutant ovaries depends on the number of persisting ECs, which is determined at the larval stage. Accordingly, much more frequent DNA breaks were found in somatic cells of flam larval ovaries than in adult ECs. The absence of Chk2 or ATM checkpoint kinases dramatically enhanced oogenesis defects of flam mutants, in contrast to the germline TE-induced defects that are known to be mostly suppressed by chk2 mutation. These results demonstrate a crucial role of checkpoint kinases in protecting niche cells against deleterious TE activation and suggest substantial differences between DNA damage responses in ovarian somatic and germ cells (Sokolova, 2020).

    Unraveling the features of somatic transposition in the Drosophila intestine

    Siudeja, K., van den Beek, M., Riddiford, N., Boumard, B., Wurmser, A., Stefanutti, M., Lameiras, S. and Bardin, A. J. (2021). Unraveling the features of somatic transposition in the Drosophila intestine. Embo j: e106388. PubMed ID: 33634906

    Unraveling the features of somatic transposition in the Drosophila intestine

    Transposable elements (TEs) play a significant role in evolution, contributing to genetic variation. However, TE mobilization in somatic cells is not well understood. This study addressed the prevalence of transposition in a somatic tissue, exploiting the Drosophila midgut as a model. Using whole-genome sequencing of in vivo clonally expanded gut tissue, hundreds of high-confidence somatic TE integration sites were mapped genome-wide. Somatic retrotransposon insertions were shown to be associated with inactivation of the tumor suppressor Notch, likely contributing to neoplasia formation. Moreover, applying Oxford Nanopore long-read sequencing technology, evidence is provided for tissue-specific differences in retrotransposition. Comparing somatic TE insertional activity with transcriptomic and small RNA sequencing data, this study demonstrates that transposon mobility cannot be simply predicted by whole tissue TE expression levels or by small RNA pathway activity. Finally, it was revealed that somatic TE insertions in the adult fly intestine are enriched in genic regions and in transcriptionally active chromatin. Together, these findings provide clear evidence of ongoing somatic transposition in Drosophila and delineate previously unknown features underlying somatic TE mobility in vivo (Siudeja, 2021).

    Different structural variants of roo retrotransposon are active in Drosophila melanogaster

    Retrotransposon roo is one of the most active elements in Drosophila melanogaster. The level of nucleotide diversity between copies of roo is very low but structural variation in the 5'-UTR is considerable. Transposition of roo at high frequency (around 5 x 10(-2) per generation) has been shown previously in the set of mutation accumulation lines named Oviedo. This study isolated thirteen individual insertions by inverse PCR and sequenced the 5' end of the elements (between 1663 and 2039 nt) including the LTR, the 5'-UTR and a fragment of 661 nucleotides from the ORF, to study whether the new transposed copies come from a unique variant (the master copy model) or different elements are able to move (the transposon model). The elements in the Oviedo lines presented the same structural variants as the reference genome. Different structural variants were active, a behaviour compatible with the "transposon model" in which the copies localized in multiple sites in the genome are able to transpose. At the level of sequence, the copies of roo in these lines are highly similar to the elements in the reference genome. The phylogenetic tree shows a shallow diversification with unsupported nodes denoting that all the elements currently active are very young. This observation together with the great polymorphism in insertion sites implies a rapid turnover of the elements (Diaz-Gonzalez, 2020).

    Interrogating the 5'UTR tandem repeats of retrotransposon roo of Drosophila about horizontal transfer

    Horizontal transfer in Drosophila has been inferred for several families of transposable elements. Specifically, the retroelement roo has been suggested to have been horizontally transferred between the species D. melanogaster, D. simulans, D. sechellia and D. yakuba. The inferences were based on the observation that divergence between transposable elements in different species was lower than the divergence found in typical nuclear genes and in the incongruence of phylogenies of the species and their TEs. This study addressed the question of the possible horizontal transfer of roo between species of the Drosophila genus by studying the presence absence of a duplication of 99 bp in the 5'UTR of the transposon, as well as comparing the sequences of the paralogous and orthologous duplicated repeats within and between species. First, the repeats were only found in five species of the melanogaster subgroup. Second, the date of occurrence of the duplication event originating the repeats was posterior to the split of the subgroup. The duplication date suggests an origin previous to the split of D. simulans and D. sechellia and close to the divergence of D. melanogaster from the D. simulans complex. These data point to horizontal transfer to the afrotropical species D. yakuba and D. erecta from one of the cosmopolitan species D. melanogaster or D. simulans. It is proposed that the parasitoid wasp Leptopilina could have been the vector of horizontal transfer after the observation that a sequence of 845 bp with high homology to a fragment of roo was isolated from this wasp (Dominguez, 2021).

    Chromatin remodelling and retrotransposons activities during regeneration in Drosophila

    Regeneration is a response mechanism aiming to reconstruct lost or damaged structures. To achieve this, the cells repopulating the lost tissue often have to change their original identity, a process that involves chromatin remodelling. This study analysed the issue of chromatin remodelling during regeneration in the wing disc of Drosophila. In this disc the ablation of the central region (the pouch) induces the regenerative response of the cells from the lateral region (the hinge), which reconstitute the wing pouch. Euchromatin and heterochromatin histone marks were examined during the process, and it was found that heterochromatin marks disappear but are recovered when regeneration is complete. Euchromatin marks are not modified. The transcription of two retrotransposons, Roo and F-element in the regenerating cells is described. A temporal correlation was established between the alterations of heterochromatin marks and the levels of transcription of two retrotransposons, Roo and F-element, both during embryonic development and in the regeneration process (Azpiazu, 2022).

    The transposition rate has little influence on the plateauing level of the P-element

    The popular trap model assumes that the invasion of transposable elements (TEs) in mammals and invertebrates are stopped by piRNAs that emerge after insertion of the TE into a piRNA cluster. It remains, however, still unclear which factors influence the dynamics of TE invasions. The activity of the TE (i.e. transposition rate) is one frequently discussed key factor. This study took advantage of the temperature dependent activity of the P-element, a widely studied eukaryotic TE, to test how TE activity affects the dynamics of a TE invasion. P-element invasion dynamics were monitored in experimental Drosophila simulans populations at hot and cold culture conditions. Despite marked differences in transposition rates, the P-element reached very similar copy numbers at both temperatures. The reduction of the insertion rate upon approaching the copy number plateau was accompanied by similar amounts of piRNAs against the P-element at both temperatures. Nevertheless, fewer numbers of P-element insertions were observed in piRNA clusters than expected, which is not compatible with a simple trap model. The ping-pong cycle, which degrades TE transcripts, becomes typically active after the copy number plateaued. A model, with few parameters, was generated that largely captures the observed invasion dynamics. It is concluded that the transposition rate has at the most only a minor influence on TE abundance, but other factors, such as paramutations or selection against TE insertions are shaping the TE composition (Kofler, 2022).

    Hybrid dysgenesis in Drosophila virilis results in clusters of mitotic recombination and loss-of-heterozygosity but leaves meiotic recombination unaltered

    Transposable elements (TEs) are endogenous mutagens and their harmful effects are especially evident in syndromes of hybrid dysgenesis. In Drosophila virilis, hybrid dysgenesis is a syndrome of incomplete gonadal atrophy that occurs when males with multiple active TE families fertilize females that lack active copies of the same families. This study sought to determine how the landscape of germline recombination is affected by parental TE asymmetry. Recombination rate and TE density were shown to be negatively correlated in this species. Then recombination events in the germline of dysgenic versus non-dysgenic F1 females were contrasted to show that the landscape of meiotic recombination is hardly perturbed during hybrid dysgenesis. In contrast, hybrid dysgenesis in the female germline increases transmission of chromosomes with mitotic recombination. Clusters of mitotic recombination events in dysgenic females were shown to be associated with genomic regions with transposons implicated in hybrid dysgenesis. Overall, it is concluded that increased mitotic recombination is likely the result of early TE activation in dysgenic progeny, but a stable landscape of meiotic recombination indicates that either transposition is ameliorated in the adult female germline or that regulation of meiotic recombination is robust to ongoing transposition. These results indicate that the effects of parental TE asymmetry on recombination are likely sensitive to the timing of transposition (Hemmer, 2020)

    Transposable elements contribute to the genomic response to insecticides in Drosophila melanogaster

    Most of the genotype-phenotype analyses to date have largely centered attention on single nucleotide polymorphisms. However, transposable element (TE) insertions have arisen as a plausible addition to the study of the genotypic-phenotypic link because of to their role in genome function and evolution. This work investigated the contribution of TE insertions to the regulation of gene expression in response to insecticides. Four Drosophila melanogaster strains were exposed to malathion, a commonly used organophosphate insecticide. By combining information from different approaches, including RNA-seq and ATAC-seq, it was found that TEs can contribute to the regulation of gene expression under insecticide exposure by rewiring cis-regulatory networks. This article is part of a discussion meeting issue 'Crossroads between transposons and gene regulation' (Salces-Ortiz, 2020).

    Paternally inherited P-element copy number affects the magnitude of hybrid dysgenesis in Drosophila simulans and D. melanogaster

    Transposable elements (TEs) are repetitive regions of DNA that are able to self-replicate and reinsert themselves throughout host genomes. Since the discovery of TEs, a prevalent question has been whether increasing TE copy number has an effect on the fitness of their hosts. P-elements (PEs) in Drosophila are a well-studied TE that has strong phenotypic effects. When a female without PEs (M) is crossed to a male with them (P), the resulting females are often sterile, a phenomenon called hybrid dysgenesis (HD). This study used short and long-read sequencing to infer the number of PEs in the genomes of dozens of isofemale lines from two Drosophila species and measured whether the magnitude of HD was correlated with the number of PEs in the paternal genome. Consistent with previous reports, evidence was found for a positive correlation between the paternal PE copy number and the magnitude of HD in progeny from ♀M x ♂ P crosses for both species. Other crosses are not affected by the number of PE copies. This study also found that the correlation between the strength of HD and PE copy number differs between species which suggest there are genetic differences that might make some genomes more resilient to the potentially deleterious effects of TEs. These results suggest that PE copy number interacts with other factors in the genome and the environment to cause HD and that the importance of these interactions is species-specific (Serrato-Capuchina, 2020).

    A Transposon Story: From TE Content to TE Dynamic Invasion of Drosophila Genomes Using the Single-Molecule Sequencing Technology from Oxford Nanopore

    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. This paper describes 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, the landscapes of the most recent TEIs could be precisely describe 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, the pipeline was used to analyze ONT sequencing data from a D. melanogaster unstable line in which LTR transposition was derepressed for 73 successive generations. It was possible to 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 (Mohamed, 2020).

    A Forward Genetic Approach to Mapping a P-Element Second Site Mutation Identifies DCP2 as a Novel Tumour Suppressor in Drosophila melanogaster

    The use of transposons to create mutations has been the cornerstone of Drosophila genetics in the past few decades. Second-site mutations caused by transpositions are often devoid of transposons and thereby affect subsequent analyses. In a P-element mutagenesis screen, a second site mutation was identified on chromosome 3, wherein the homozygous mutants exhibit classic hallmarks of tumour suppressor mutants, including brain tumour and lethality; hence the mutant line was initially named as lethal (3) tumorous brain [l(3)tb]. Classical genetic approaches relying on meiotic recombination and subsequent complementation with chromosomal deletions and gene mutations mapped the mutation to CG6169, the mRNA decapping protein 2 (DCP2), on the left arm of the third chromosome (3L). Thus the mutation was renamed as DCP2(l(3)tb) Fine mapping of the mutation further identified the presence of a Gypsy--LTR like sequence in the 5'UTR coding region of DCP2, along with the expansion of the adjacent upstream intergenic AT-rich sequence. The mutant phenotypes are rescued by the introduction of a functional copy of DCP2 in the mutant background, thereby establishing the causal role of the mutation and providing a genetic validation of the allelism. With the increasing repertoire of genes being associated with tumour biology, this is the first instance of mRNA decapping protein being implicated in Drosophila tumourigenesis. These findings, therefore, imply a plausible role for the mRNA degradation pathway in tumorigenesis and identify DCP2 as a potential candidate for future explorations of cell cycle regulatory mechanisms (Mishra, 2020).

    Epigenetic conflict on a degenerating Y chromosome increases mutational burden in Drosophila males

    Large portions of eukaryotic genomes consist of transposable elements (TEs), and the establishment of transcription-repressing heterochromatin during early development safeguards genome integrity in Drosophila. Repeat-rich Y chromosomes can act as reservoirs for TEs ('toxic' Y effect), and incomplete epigenomic defenses during early development can lead to deleterious TE mobilization. This study contrasted the dynamics of early TE activation in two Drosophila species with vastly different Y chromosomes of different ages. Zygotic TE expression is elevated in male embryos relative to females in both species, mostly due to expression of Y-linked TEs. Interestingly, male-biased TE expression diminishes across development in D. pseudoobscura, but remains elevated in D. miranda, the species with the younger and larger Y chromosome. The repeat-rich Y of D. miranda still contains many actively transcribed genes, which compromise the formation of silencing heterochromatin. Elevated TE expression results in more de novo insertions of repeats in males compared to females. This lends support to the idea that the 'toxic' Y chromosome can create a mutational burden in males when genome-wide defense mechanisms are compromised, and suggests a previously unappreciated epigenetic conflict on evolving Y chromosomes between transcription of essential genes and silencing of selfish DNA (Wei, 2020).

    Toxic Y chromosome: Increased repeat expression and age-associated heterochromatin loss in male Drosophila with a young Y chromosome

    Sex-specific differences in lifespan are prevalent across the tree of life and influenced by heteromorphic sex chromosomes. In species with XY sex chromosomes, females often outlive males. Males and females can differ in their overall repeat content due to the repetitive Y chromosome, and repeats on the Y might lower survival of the heterogametic sex (toxic Y effect). This study took advantage of the well-assembled young Y chromosome of Drosophila miranda to study the sex-specific dynamics of chromatin structure and repeat expression during aging in male and female flies. Male D. miranda have about twice as much repetitive DNA compared to females, and live shorter than females. Heterochromatin is crucial for silencing of repetitive elements, yet old D. miranda flies lose H3K9me3 modifications in their pericentromere, with heterochromatin loss being more severe during aging in males than females. Satellite DNA becomes de-repressed more rapidly in old vs. young male flies relative to females. In contrast to what is observed in D. melanogaster, it was found that transposable elements (TEs) are expressed at higher levels in male D. miranda throughout their life. Epigenetic silencing via heterochromatin formation is ineffective on the TE-rich neo-Y chromosome, presumably due to active transcription of a large number of neo-Y linked genes, resulting in up-regulation of Y-linked TEs already in young males. This is consistent with an interaction between the evolutionary age of the Y chromosome and the genomic effects of aging. These data support growing evidence that 'toxic Y chromosomes' can diminish male fitness and a reduction in heterochromatin can contribute to sex-specific aging (Nguyen, 2021).

    The prolyl-isomerase PIN1 is essential for nuclear Lamin-B structure and function and protects heterochromatin under mechanical stress

    Chromatin organization plays a crucial role in tissue homeostasis. Heterochromatin relaxation and consequent unscheduled mobilization of transposable elements (TEs) are emerging as key contributors of aging and aging-related pathologies, including Alzheimer's disease (AD) and cancer. However, the mechanisms governing heterochromatin maintenance or its relaxation in pathological conditions remain poorly understood. This study shows that PIN1, the only phosphorylation-specific cis/trans prolyl isomerase, whose loss is associated with premature aging and AD, is essential to preserve heterochromatin. This study demonstrated that the PIN1 function is conserved from Drosophila to humans and prevents TE mobilization-dependent neurodegeneration and cognitive defects. Mechanistically, PIN1 maintains nuclear type-B Lamin structure and anchoring function for heterochromatin protein 1α (HP1α). This mechanism prevents nuclear envelope alterations and heterochromatin relaxation under mechanical stress, which is a key contributor to aging-related pathologies (Napoletano, 2021).

    Evidence of the Physical Interaction between Rpl22 and the Transposable Element Doc5, a Heterochromatic Transposon of Drosophila melanogaster

    Chromatin is a highly dynamic biological entity that allows for both the control of gene expression and the stabilization of chromosomal domains. Given the high degree of plasticity observed in model and non-model organisms, it is not surprising that new chromatin components are frequently described. This work tested the hypothesis that the remnants of the Doc5 transposable element, which retains a heterochromatin insertion pattern in the melanogaster species complex, can be bound by chromatin proteins, and thus be involved in the organization of heterochromatic domains. Using the Yeast One Hybrid approach, Rpl22 was found to be a potential interacting protein of Doc5. The observed interaction was further tested through an Electrophoretic Mobility Shift Assay, uncovering that the N-terminal portion of the protein is sufficient to interact with Doc5. However, in situ localization of the native protein failed to detect Rpl22 association with chromatin. The results obtained are discussed in the light of the current knowledge on the extra-ribosomal role of ribosomal protein in eukaryotes, which suggests a possible role of Rpl22 in the determination of the heterochromatin in Drosophila (Berloco, 2021).

    Involvement of DNA Repair Genes and System of Radiation-Induced Activation of Transposons in Formation of Transgenerational Effects
    The study of the genetic basis of the manifestation of radiation-induced effects and their transgenerational inheritance makes it possible to identify the mechanisms of adaptation and possible effective strategies for the survival of organisms in response to chronic radioactive stress. One persistent hypothesis is that the activation of certain genes involved in cellular defense is a specific response of the cell to irradiation. There is also data indicating the important role of transposable elements in the formation of radiosensitivity/radioresistance of biological systems. This work studied the interaction of the systems of hobo transposon activity and DNA repair in the cell under conditions of chronic low-dose irradiation and its participation in the inheritance of radiation-induced transgenerational instability in Drosophila. The results showed a significant increase of sterility and locus-specific mutability, a decrease of survival, fertility and genome stability (an increase the frequency of dominant lethal mutations and DNA damage) in non-irradiated F(1)/F(2) offspring of irradiated parents with dysfunction of the mus304 gene which is responsible for excision and post-replicative recombination repair and repair of double-stranded DNA breaks. The combined action of dysfunction of the mus309 (Bloom syndrome helicase) gene and transpositional activity of hobo elements also led to the transgenerational effects of irradiation but only in the F(1) offspring. Dysfunction of the genes of other DNA repair systems (mus101 and mus210) showed no visible effects inherited from irradiated parents subjected to hobo transpositions. The mei-41 gene showed specificity in this type of interaction, which consists in its higher efficiency in sensing events induced by transpositional activity rather than irradiation (Yushkova, 2020).

    Contribution of transposable elements to transgenerational effects of chronic radioactive exposure of natural populations of Drosophila melanogaster living for a long time in the zone of the Chernobyl nuclear disaster

    The accident at the Chernobyl Nuclear Power Plant (ChNPP) led to the negative impact of chronic radioactive contamination on populations of organisms associated with the transgenerational transmission of genome instability. In this study, for the first time, the features of the influence of transposable elements (TEs) on the long-term biological consequences of the ChNPP are considered. In this work, specimens of D. melanogaster obtained from natural populations in 2007 in the areas of the ChNPP with heterogeneous radioactive contamination were studied. The descendants from these populations were maintained in laboratory (inbred) conditions for 160 generations. A stable transgenerational transmission of dominant lethal mutations (DLMs) to the offspring of all studied populations was shown. The DLM frequencies strongly were correlated with the level of survival of offspring. The mean frequencies of recessive sex-linked lethal mutations varied at the level of spontaneous point mutations. The simultaneous presence of P, hobo and I elements indicates that the studied populations do not have a definite cytotype, their phenotypic status is unstable. The behavior of TEs in the genomes of offspring depends not only on parental exposure, but also on origin of population, distance to the ChNPP, and inbred conditions. The obtained results confirm the hypothesis that TEs are involved in transgenerational transmission and accumulation of mutations by the offspring of irradiated parents. The TEs pattern present in the Chernobyl genomes of D. melanogaster is a peculiar of epigenetic mechanism for the regulation of plasticity and adaptation of populations living for many generations under conditions of a technogenically caused radiation background (Yushkova, 2022a).

    Expression of retrotransposons contributes to aging in Drosophila Genetics

    Retrotransposons are a class of transposable elements capable of self-replication and insertion into new genomic locations. Across species, the mobilization of retrotransposons in somatic cells has been suggested to contribute to the cell and tissue functional decline that occurs during aging. Retrotransposons are broadly expressed across cell types, and de novo insertions have been observed to correlate with tumorigenesis. However, the extent to which new retrotransposon insertions occur during normal aging and their effect on cellular and animal function remains understudied. This study used a single nucleus whole genome sequencing approach in Drosophila to directly test whether transposon insertions increase with age in somatic cells. Analyses of nuclei from thoraces and indirect flight muscles using a newly developed pipeline, Retrofind, revealed no significant increase in the number of transposon insertions with age. Despite this, reducing the expression of two different retrotransposons, 412 and Roo, extended lifespan, but did not alter indicators of health such as stress resistance. This suggests a key role for transposon expression and not insertion in regulating longevity. Transcriptomic analyses revealed similar changes to gene expression in 412 and Roo knockdown flies and highlighted changes to genes involved in proteolysis and immune function as potential contributors to the observed changes in longevity. Combined, these data show a clear link between retrotransposon expression and aging (Schneider, 2023).

    Transposon expression in the Drosophila brain is driven by neighboring genes and diversifies the neural transcriptome

    Somatic transposon expression in neural tissue is commonly considered as a measure of mobilization and has therefore been linked to neuropathology and organismal individuality. This study combined genome sequencing data with single-cell mRNA sequencing of the same, inbred fly strain to map transposon expression in the Drosophila midbrain and found that transposon expression patterns are highly stereotyped. Every detected transposon is resident in at least one cellular gene with a matching expression pattern. Bulk RNA sequencing from fly heads of the same strain revealed that coexpression is a physical link in the form of abundant chimeric transposon-gene mRNAs. 264 genes were identified where transposons introduce cryptic splice sites into the nascent transcript and thereby significantly expand the neural transcript repertoire. Some genes exclusively produce chimeric mRNAs with transposon sequence and on average 11.6% of the mRNAs produced from a given gene are chimeric. Conversely, most transposon-containing transcripts are chimeric, which suggests that somatic expression of these transposons is largely driven by cellular genes. It is proposed that chimeric mRNAs produced by alternative splicing into polymorphic transposons, rather than transposon mobilization, may contribute to functional differences between individual cells and animals (Treiber, 2020).

    piRNA-mediated gene regulation and adaptation to sex-specific transposon expression in D. melanogaster male germline

    Small noncoding piRNAs act as sequence-specific guides to repress complementary targets in Metazoa. Prior studies in Drosophila ovaries have demonstrated the function of the piRNA pathway in transposon silencing and therefore genome defense. However, the ability of the piRNA program to respond to different transposon landscapes and the role of piRNAs in regulating host gene expression remain poorly understood. This study comprehensively analyzed piRNA expression and defined the repertoire of their targets in Drosophila melanogaster testes. Comparison of piRNA programs between sexes revealed sexual dimorphism in piRNA programs that parallel sex-specific transposon expression. Using a novel bioinformatic pipeline, new piRNA clusters were identified, and complex satellites were identified as dual-strand piRNA clusters. While sharing most piRNA clusters, the two sexes employ them differentially to combat the sex-specific transposon landscape. Two piRNA clusters were identified that produce piRNAs antisense to four host genes in testis, including CG12717/pirate, a SUMO protease gene. piRNAs encoded on the Y chromosome silence pirate, but not its paralog, to exert sex- and paralog-specific gene regulation. Interestingly, pirate is targeted by endogenous siRNAs in a sibling species, Drosophila mauritiana, suggesting distinct but related silencing strategies invented in recent evolution to regulate a conserved protein-coding gene (Chen, 2021).

    Infections by Transovarially Transmitted DMelSV in Drosophila Have No Impact on Ovarian Transposable Element Transcripts but Increase Their Amounts in the Soma

    Transposable elements (TEs) are genomic parasites, which activity is tightly controlled in germline cells. Using Sindbis virus, it was recently demonstrated that viral infections affect TE transcript amounts in somatic tissues. However, the strongest evolutionary impacts are expected in gonads, because that is where the genomes of the next generations lie. This aspect was investigated using the Drosophila melanogaster Sigma virus. It is particularly relevant in the genome/TE interaction given its tropism to ovaries, which is the organ displaying the more sophisticated TE control pathways. Results in Drosophila simulans flies allowed confirmation of the existence of a strong homeostasis of the TE transcriptome in ovaries upon infection, which, however, rely on TE-derived small RNA modulations. In addition, this study performed a meta-analysis of RNA-seq data and propose that the immune pathway that is triggered upon viral infection determines the direction of TE transcript modulation in somatic tissues (Roy, 2021).

    Regulatory regions in natural transposable element insertions drive interindividual differences in response to immune challenges in Drosophila

    Variation in gene expression underlies interindividual variability in relevant traits including immune response. However, the genetic variation responsible for these gene expression changes remains largely unknown. Among the non-coding variants that could be relevant, transposable element insertions are promising candidates as they have been shown to be a rich and diverse source of cis-regulatory elements. This work used a population genetics approach to identify transposable element insertions likely to increase the tolerance of Drosophila melanogaster to bacterial infection by affecting the expression of immune-related genes. This study identified 12 insertions associated with allele-specific expression changes in immune-related genes. three of these insertions were experimentally validate including one likely to be acting as a silencer, one as an enhancer, and one with a dual role as enhancer and promoter. The direction in the change of gene expression associated with the presence of several of these insertions is consistent with an increased survival to infection. Indeed, for one of the insertions, it was shown that this is the case by analyzing both natural populations and CRISPR/Cas9 mutants in which the insertion is deleted from its native genomic context. It was shown that transposable elements contribute to gene expression variation in response to infection in D. melanogaster and that this variation is likely to affect their survival capacity. Because the role of transposable elements as regulatory elements is not restricted to Drosophila, transposable elements are likely to play a role in immune response in other organisms as well (Ullastres, 2021).

    Retrotransposon activation during Drosophila metamorphosis conditions adult antiviral responses

    Retrotransposons are one type of mobile genetic element that abundantly reside in the genomes of nearly all animals. Their uncontrolled activation is linked to sterility, cancer and other pathologies, thereby being largely considered detrimental. This study reports that, within a specific time window of development, retrotransposon activation can license the host's immune system for future antiviral responses. mdg4 (also known as Gypsy) retrotransposon selectively becomes active during metamorphosis at the Drosophila pupal stage. At this stage, mdg4 activation educates the host's innate immune system by inducing the systemic antiviral function of the nuclear factor-κB protein Relish in a dSTING-dependent manner. Consequently, adult flies with mdg4, Relish or dSTING silenced at the pupal stage are unable to clear exogenous viruses and succumb to viral infection. Altogether, these data reveal that hosts can establish a protective antiviral response that endows a long-term benefit in pathogen warfare due to the developmental activation of mobile genetic elements (Wang, 2022).

    Impaired function of rDNA transcription initiation machinery leads to derepression of ribosomal genes with insertions of R2 retrotransposon

    Eukaryotic genomes harbor hundreds of rRNA genes, many of which are transcriptionally silent. In Drosophila, some rDNA repeats contain insertions of the R2 retrotransposon, which is capable to be transcribed only as part of pre-rRNA molecules. rDNA units with R2 insertions are usually inactivated, although R2 expression may be beneficial in cells with decreased rDNA copy number. This study found that R2-inserted rDNA units are enriched with HP1a and H3K9me3 repressive mark, whereas disruption of the heterochromatin components slightly affects their silencing in ovarian germ cells. Surprisingly, a dramatic upregulation of R2-inserted rRNA genes was observed in ovaries lacking Udd (Under-developed) or other subunits (TAF1b and TAF1c-like) of the SL1-like complex, which is homologues to mammalian Selective factor 1 (SL1) involved in rDNA transcription initiation. Derepression of rRNA genes with R2 insertions was accompanied by a reduction of H3K9me3 and HP1a enrichment. It is suggested that the impairment of the SL1-like complex affects a mechanism of selective activation of intact rDNA units which competes with heterochromatin formation. It is also proposed that R2 derepression may serve as an adaptive response to compromised rRNA synthesis (Fefelova, 2022).

    Histone H3.3 K27M and K36M mutations de-repress transposable elements through perturbation of antagonistic chromatin marks
    Histone H3.3 lysine-to-methionine substitutions K27M and K36M impair the deposition of opposing chromatin marks, H3K27me3/me2 and H3K36me3/me2. This study shows that these mutations induce hypotrophic and disorganized eyes in Drosophila eye primordia. Restriction of H3K27me3 spread in H3.3K27M and its redistribution in H3.3K36M result in transcriptional deregulation of PRC2-targeted eye development and of piRNA biogenesis genes, including krimp. Notably, both mutants promote redistribution of H3K36me2 away from repetitive regions into active genes, which associate with retrotransposon derepression in eye discs. Aberrant expression of krimp represses LINE retrotransposons but does not contribute to the eye phenotype. Depletion of H3K36me2 methyltransferase ash1 in H3.3K27M, and of PRC2 component E(z) in H3.3K36M, restores the expression of eye developmental genes and normal eye growth, showing that redistribution of antagonistic marks contributes to K-to-M pathogenesis. These results implicate a novel function for H3K36me2 and showcase convergent downstream effects of oncohistones that target opposing epigenetic marks (Chaouch, 2021).

    Transposable element profiles reveal cell line identity and loss of heterozygosity in Drosophila cell culture

    Cell culture systems allow key insights into biological mechanisms yet suffer from irreproducible outcomes in part because of cross-contamination or mislabeling of cell lines. Cell line misidentification can be mitigated by the use of genotyping protocols, which have been developed for human cell lines but are lacking for many important model species. This study leveraged the classical observation that transposable elements (TEs) proliferate in cultured Drosophila cells to demonstrate that genome-wide TE insertion profiles can reveal the identity and provenance of Drosophila cell lines. Multiple cases were identified where TE profiles clarify the origin of Drosophila cell lines (Sg4, mbn2, and OSS_E) relative to published reports, and also provide evidence that insertions from only a subset of long-terminal repeat retrotransposon families are necessary to mark Drosophila cell line identity. A new bioinformatics approach was developed to detect TE insertions and estimate intra-sample allele frequencies in legacy whole-genome sequencing data (called ngs_te_mapper2), which revealed loss of heterozygosity as a mechanism shaping the unique TE profiles that identify Drosophila cell lines. This work contributes to the general understanding of the forces impacting metazoan genomes as they evolve in cell culture and paves the way for high-throughput protocols that use TE insertions to authenticate cell lines in Drosophila and other organisms (Han, 2021).

    Transposable element landscapes in aging Drosophila

    Genetic mechanisms that repress transposable elements (TEs) in young animals decline during aging, as reflected by increased TE expression in aged animals. Does increased TE expression during aging lead to more genomic TE copies in older animals? To address this question, TE Landscapes (TLs) were quantified via whole genome sequencing of young and aged Drosophila strains of wild-type and mutant backgrounds. TLs were quantified in whole flies and dissected brains, and the feasibility of this approach was validated in detecting new TE insertions in aging Drosophila genomes when small RNA and RNA interference (RNAi) pathways are compromised. This study also describes improved sequencing methods to quantify extra-chromosomal DNA circles (eccDNAs) in Drosophila as an additional source of TE copies that accumulate during aging. Lastly, to combat the natural progression of aging-associated TE expression, it was shown that knocking down PAF1, a conserved transcription elongation factor that antagonizes RNAi pathways, may bolster suppression of TEs during aging and extend lifespan. This study suggests that in addition to a possible influence by different genetic backgrounds, small RNA and RNAi mechanisms may mitigate genomic TL expansion despite the increase in TE transcripts during aging (Yang, 2022).

    The effects of transpositions of functional I retrotransposons depend on the conditions and dose of parental exposure

    Transposable elements (TEs) cause destabilization of animal genomes. I retrotransposons of Drosophila melanogaster, as well as human LINE1 retrotransposons, are sources of intra- and interindividual diversity and responses to the action of internal and external factors. The aim of this study was to investigate the response to irradiation for the offspring of Drosophila melanogaster with the increased activity of inherited functional I elements. The material used was dysgenic Drosophila females with active I retrotransposons obtained as a result of crossing irradiated/non-irradiated parents of a certain genotype. Non-dysgenic females (without functional I elements) were used as controls. The effects of different conditions (irradiation of both parents simultaneously or separately) and doses (1-100 Gy) of parental irradiation have been assessed by analyzing SF-sterility, DNA damage and lifespan. The presence of full-size I retrotransposons was determined by PCR analysis. The maternal exposure and exposure of both parents are efficient in contrast with paternal exposure. Irradiation of mothers reduces the reproductive potential and viability of their female offspring which undergo high activity of functional I retrotransposons. Though I retrotranspositions negatively affect the female gonads, irradiation of the paternal line can increase the lifespan of SF-sterile females. Radiation stress in the range of 1-100 Gy increases DNA fragmentation in both somatic and germ cells of the ovaries with high I-retrotransposition. These results allow for the specificity of the radiation-induced behavior of I retrotransposons and their role in survival under conditions of strong radiation stress (Yushkova, 2022a).

    Intercellular viral spread and intracellular transposition of Drosophila gypsy

    It has become increasingly clear that retrotransposons (RTEs) are more widely expressed in somatic tissues than previously appreciated. RTE expression has been implicated in a myriad of biological processes ranging from normal development and aging, to age related diseases such as cancer and neurodegeneration. Long Terminal Repeat (LTR)-RTEs are evolutionary ancestors to, and share many features with, exogenous retroviruses. In fact, many organisms contain endogenous retroviruses (ERVs) derived from exogenous retroviruses that integrated into the germ line. These ERVs are inherited in Mendelian fashion like RTEs, and some retain the ability to transmit between cells like viruses, while others develop the ability to act as RTEs. The process of evolutionary transition between LTR-RTE and retroviruses is thought to involve multiple steps by which the element loses or gains the ability to transmit copies between cells versus the ability to replicate intracellularly. But, typically, these two modes of transmission are incompatible because they require assembly in different sub-cellular compartments. Like murine IAP/IAP-E elements, the gypsy family of retroelements in arthropods appear to sit along this evolutionary transition. Indeed, there is some evidence that gypsy may exhibit retroviral properties. Given that gypsy elements have been found to actively mobilize in neurons and glial cells during normal aging and in models of neurodegeneration, this raises the question of whether gypsy replication in somatic cells occurs via intracellular retrotransposition, intercellular viral spread, or some combination of the two. These modes of replication in somatic tissues would have quite different biological implications. This study demonstrates that Drosophila gypsy is capable of both cell-associated and cell-free viral transmission between cultured S2 cells of somatic origin. Further, the ability of gypsy to move between cells was demonstrated to be dependent upon a functional copy of its viral envelope protein. This argues that the gypsy element has transitioned from an RTE into a functional endogenous retrovirus with the acquisition of its envelope gene. On the other hand, it was also found that intracellular retrotransposition of the same genomic copy of gypsy can occur in the absence of the Env protein. Thus, gypsy exhibits both intracellular retrotransposition and intercellular viral transmission as modes of replicating its genome (Keegan, 2021).

    Channel Nuclear Pore Complex subunits are required for transposon silencing in Drosophila

    The Nuclear Pore Complex (NPC) is the principal gateway between nucleus and cytoplasm that enables exchange of macromolecular cargo. Composed of multiple copies of ~30 different nucleoporins (Nups), the NPC acts as a selective portal, interacting with factors which individually license passage of specific cargo classes. This study shows that two Nups of the inner channel, Nup54 and Nup58, are essential for transposon silencing via the PIWI-interacting RNA (piRNA) pathway in the Drosophila ovary. In ovarian follicle cells, loss of Nup54 and Nup58 results in compromised piRNA biogenesis exclusively from the flamenco locus, whereas knockdowns of other NPC subunits have widespread consequences. This provides evidence that some nucleoporins can acquire specialised roles in tissue-specific contexts. These findings consolidate the idea that the NPC has functions beyond simply constituting a barrier to nuclear/cytoplasmic exchange, as genomic loci subjected to strong selective pressure can exploit NPC subunits to facilitate their expression (Munafo, 2021).

    This study has found that Nup54 and Nup58 are specifically required for TE regulation in the follicle cells of Drosophila ovaries by enabling export and subsequent piRNA production from flam transcripts. This functional requirement is distinct from the general role of the NPC, since other Nups, even those most proximal to Nup54 and Nup58 within the pore, are broadly required for gene expression and cell survival. These findings consolidate the view of Nups as dynamic players in various cellular processes and expand the variety of roles ascribed to the Nups (Munafo, 2021).

    Though flam bears canonical features common to other mRNAs, it is nonetheless specifically recognised and processed into piRNAs. A dedicated export-coupled licensing, involving Nup54 and Nup58, may allow this long transcript to be escorted directly from its genomic origin to the Yb-bodies to facilitate piRNA production. If this machinery is disrupted, for example, by loss of Nup54 and Nup58, flam transcripts are confined to the nucleus and destabilised, thus underscoring the need for a unified process from transcription to licensing. It is interesting to note that siYb caused a slight decrease of PRO-seq signal across the entire piRNA cluster locus, possibly indicating that disruption of flam export-coupled licensing negatively affects its transcription via a yet-unknown feedback mechanism. Nup54 and Nup58 interact with both the nuclear (Nxf1/Nxt1) and cytosolic (Yb) components of flam expression and thus suggest that these factors bridge nuclear export to cytosolic fate specification. Although several aspects of the molecular mechanism remain to be elucidated (i.e., how this links to upstream transcription and whether additional factors are involved in the nucleus or in the cytosol), a tentative model is proposed whereby this Nxf1-NPC-Yb axis coordinates initiation, processivity, and directionality of flam export, directly feeding the transcript into the piRNA biogenesis route via Yb-bodies. Rescue experiments and protein-protein interaction studies argue for this TE silencing function of Nup54 and Nup58 to be carried out from within the pore, especially since neither Nup is among the most dynamic components of the NPC. However, a function for Nup54 and Nup58 in the cytosol, possibly as components of Yb-bodies, cannot be completely excluded. Nup54 and Nup58 interact directly with both Yb and Nxf1; however, it is presently unclear how the transcript is released from the Nxf1/Nxt1 export complexes and handed over to Yb, especially since physical interactions between Yb and Nxf1 was not detected. It cannot be excluded that such interaction exists and remained undetected in over-expression experiments or that additional adaptor proteins take part in this process. Alternatively, Nup54/58 may form independent complexes with Yb and Nxf1; however, lack of enrichment for flam transcripts associated with Nup54/58 makes this seem unlikely. Super-resolution imaging of the NPC will be required to clarify the relative position of each component of this export route and to understand whether Nup54 and Nup58 function also outside the NPC. Of note, cytosolic Nup358 has been shown to be required for piRNA production from the dual-strand cluster 42AB in nurse cells. Its depletion leads to prominent de-localisation of Piwi, which was not observed upon loss of Nup54/Nup58, thus suggesting a different mode of action. Nonetheless, this further underscores that different NPC subunits can be co-opted for TE silencing in tissue-specific contexts (Munafo, 2021).

    The proposed mechanism for flam export-coupled licensing relies on a tissue-specific effector (Yb) and could represent a broader paradigm for co-option of phenylalanine and glycine rich (FG) Nups for specific transcript trafficking routes. It is envisioned that in principle any transcript subject to strong selective pressure could evolve a dedicated export machinery via adapting FG-Nup functions in a cell-type-specific manner. Such mechanisms may have gone unnoticed previously because of the general role of the NPC. Interestingly, several Nup genes in Drosophila show signs of rapid adaptive evolution that result in hybrid incompatibilities, which are often hallmarks of genes involved in genetic conflicts, such as transposon control. It is further speculated that a tissue-specific control of export routes might contribute to explain the molecular mechanisms underlying so-called 'nucleoporopathies', human syndromes caused by mutated Nups. In these diseases, mutation or expression changes of a Nup present in all cells of the organism leads to tissue-specific phenotypes. It is hypothesised that this might stem from specific roles of those Nups in regulating genes that are essential for the functionality of that particular tissue, which would in turn make the said tissue especially susceptible to the loss of the Nup. Future investigation will shed light on how widespread these mechanisms might be (Munafo, 2021).

    The evolution of small-RNA-mediated silencing of an invading transposable element

    Transposable elements (TEs) are genomic parasites that impose fitness costs on their hosts by producing deleterious mutations and disrupting gametogenesis. Host genomes avoid these costs by regulating TE activity, particularly in germline cells where new insertions are heritable and TEs are exceptionally active. However, the capacity of different TE-associated fitness costs to select for repression in the host, and the role of selection in the evolution of TE regulation more generally, remain controversial. This study used forward, individual-based simulations to examine the evolution of small-RNA-mediated TE regulation, a conserved mechanism for TE repression that is employed by both prokaryotes and eukaryotes. To design and parameterize a biologically realistic model, this study drew on an extensive survey of empirical studies of the transposition and regulation of P-element DNA transposons in Drosophila melanogaster. Even under conservative assumptions, where small-RNA-mediated regulation reduces transposition only, repression evolves rapidly and adaptively after the genome is invaded by a new TE in simulated populations. It was further shown that the spread of repressor alleles through simulated populations is greatly enhanced by two additional TE-imposed fitness costs: dysgenic sterility and ectopic recombination. Finally, it was demonstrated that the adaptive mutation rate to repression is a critical parameter that influences both the evolutionary trajectory of host repression and the associated proliferation of TEs after invasion in simulated populations. These findings suggest that adaptive evolution of TE regulation may be stronger and more prevalent than previously appreciated, and provide a framework for interpreting empirical data (Kelleher, 2018).

    Maternally inherited piRNAs direct transient heterochromatin formation at active transposons during early Drosophila embryogenesis

    The PIWI-interacting RNA (piRNA) pathway controls transposon expression in animal germ cells, thereby ensuring genome stability over generations. In Drosophila, piRNAs are intergenerationally inherited through the maternal lineage, and this has demonstrated importance in the specification of piRNA source loci and in silencing of I- and P-elements in the germ cells of daughters. Maternally inherited Piwi protein enters somatic nuclei in early embryos prior to zygotic genome activation and persists therein for roughly half of the time required to complete embryonic development. To investigate the role of the piRNA pathway in the embryonic soma, a conditionally unstable Piwi protein was created. This enabled maternally deposited Piwi to be cleared from newly laid embryos within 30 minutes and well ahead of the activation of zygotic transcription. Examination of RNA and protein profiles over time, and correlation with patterns of H3K9me3 deposition, suggests a role for maternally deposited Piwi in attenuating zygotic transposon expression in somatic cells of the developing embryo. In particular, robust deposition of piRNAs targeting roo, an element whose expression is mainly restricted to embryonic development, results in the deposition of transient heterochromatic marks at active roo insertions. It is hypothesized that roo, an extremely successful mobile element, may have adopted a lifestyle of expression in the embryonic soma to evade silencing in germ cells (Fabry, 2021).

    Establishment of H3K9me3-dependent heterochromatin during embryogenesis in Drosophila miranda

    Heterochromatin is a key architectural feature of eukaryotic genomes crucial for silencing of repetitive elements. During Drosophila embryonic cellularization, heterochromatin rapidly appears over repetitive sequences, but the molecular details of how heterochromatin is established are poorly understood. This study mapped the genome-wide distribution of H3K9me3-dependent heterochromatin in individual embryos of Drosophila miranda at precisely staged developmental time points. Canonical H3K9me3 enrichment was found to be established prior to cellularization and matures into stable and broad heterochromatin domains through development. Intriguingly, initial nucleation sites of H3K9me3 enrichment appear as early as embryonic stage 3 over transposable elements (TEs) and progressively broaden, consistent with spreading to neighboring nucleosomes. The earliest nucleation sites are limited to specific regions of a small number of recently active retrotransposon families and often appear over promoter and 5' regions of LTR retrotransposons, while late nucleation sites develop broadly across the entirety of most TEs. Interestingly, early nucleating TEs are strongly associated with abundant maternal piRNAs and show early zygotic transcription. These results support a model of piRNA-associated co-transcriptional silencing while also suggesting additional mechanisms for site-restricted H3K9me3 nucleation at TEs in pre-cellular Drosophila embryos (Wei, 2021).

    Piwi-piRNA complexes induce stepwise changes in nuclear architecture at target loci. EMBO J: e108345

    PIWI-interacting RNAs (piRNAs) are germline-specific small RNAs that form effector complexes with PIWI proteins (Piwi-piRNA complexes) and play critical roles for preserving genomic integrity by repressing transposable elements (TEs). Drosophila Piwi transcriptionally silences specific targets through heterochromatin formation and increases histone H3K9 methylation (H3K9me3) and histone H1 deposition at these loci, with nuclear RNA export factor variant Nxf2 serving as a co-factor. Using ChEP and DamID-seq, this study has uncovered a Piwi/Nxf2-dependent target association with nuclear lamins. Hi-C analysis of Piwi or Nxf2-depleted cells reveals decreased intra-TAD and increased inter-TAD interactions in regions harboring Piwi-piRNA target TEs. Using a forced tethering system, the functional effects were analyzed of Piwi-piRNA/Nxf2-mediated recruitment of piRNA target regions to the nuclear periphery. Removal of active histone marks is followed by transcriptional silencing, chromatin conformational changes, and H3K9me3 and H1 association. These data show that the Piwi-piRNA pathway can induce stepwise changes in nuclear architecture and chromatin state at target loci for transcriptional silencing (Iwasaki, 2021).

    A model is proposed where Piwi-piRNA-mediated TE regulation of targets is linked to changes in nuclear localization, chromatin conformation, and histone modifications. Interestingly, this occurs in a stepwise manner, where localization and removal of active histone marks is followed by repressive histone marks and chromatin conformational change. These results suggest that nuclear small RNA-mediated regulation modifies nuclear architecture at the genomic level (Iwasaki, 2021).

    A general conundrum in this field is whether nuclear positioning controls gene expression or vice versa. There is some evidence that transcription can counteract lamina interactions and modulate Hi-C contacts. The relocation of piRNA target TEs after Piwi- or Nxf2-KD could be a secondary effect of transcriptional activation. Meanwhile, slight de-repression of piRNA target TEs upon the depletion of lamin proteins may suggest a possible defect in transcription upon disruption of lamina association. Defects in transcriptional regulation and nuclear localization upon Piwi-KD occur prior to changes in H3K9me3 histone marks and chromatin conformation. In C. elegans, heterochromatin regions with H3K9me3 histone marks are anchored to perinuclear regions. This suggests that in the Piwi-piRNA pathway, deposition of H3K9me3 at a later step reinforces the association of chromatin regions with perinuclear regions. Indeed, Piwi or Nxf2 knockdown, which decreases the Lamin DamID-seq signal at Piwi-piRNA target regions, also results in a major decrease in H3K9me3 levels. These results suggest that lamina localization may both be the cause and result of TE repression. Nuclear peripheral localization may affect gene expression to some extent, and the regulation of gene expression may be enforced due to its localization to the nuclear periphery (Iwasaki, 2021).

    It is proposed that the Piwi-piRNA pathway triggers spatial regulation of genomic regions in order to repress transposable elements. In other words, Piwi-piRNA system may be able to send target regions to specific positions within the nucleus in order to efficiently regulate their expression. In OSCs, piRNAs are generated and loaded onto Piwi protein at the cytoplasmic Yb body. Therefore, target gene localization to the nuclear periphery may facilitate regulation by Piwi-piRNAs imported from the cytoplasm. Within the nucleus, Piwi-piRNA functions together with Panx-Nxf2-p15 at the nascent transcript, in order to silence target TEs. Recent studies have suggested that Panx-Nxf2-p15 complex forms condensates upon Piwi-piRNA-mediated silencing (Eastwood, 2021; Schnabl, 2021). Condensate formation is coupled to dimerization of Panx-Nxf2-p15 complex, and piRNA target regions may therefore be spatially regulated upon silencing. Moreover, it has been proposed that condensate formation is stimulated by nascent RNA produced during early steps in transcription initiation. By analogy with these observations, nuclear localization and chromatin conformational changes observed in this study may also be linked to condensate formation and possibly form a spatial structure for Piwi-piRNA regulation (Iwasaki, 2021).

    Although Piwi-piRNA regulation affects nuclear localization of target chromatin regions, the effect on TAD boundaries was limited, and the effect mainly observed as changes in intra- and inter-TAD interactions. In line with this observation, it has been reported that even depletion of nuclear lamins does not result in modulation of TAD boundaries in Drosophila or mouse ES cells. This suggests that changes in nuclear localization of target chromatin regions are not directly linked to modulation of TAD boundaries, but Piwi-piRNA regulation affects expression levels of genes that are localized within the same TAD as piRNA target TEs. This is consistent with the fact that Piwi-piRNA regulation regulates not only expression, histone modifications, and chromatin accessibility of its target TEs, but also their neighboring regions (Iwasaki, 2021).

    Recent studies have revealed that heterochromatin structure is dynamic and relatively large molecules can move between both heterochromatin and euchromatin regions. In addition, some essential genes have been reported to be encoded and transcribed in heterochromatin. The Piwi-piRNA system, which is reversible and can be reversed upon loss of Piwi proteins or specific piRNAs, may contribute to this dynamic regulation of heterochromatic regions. Since Piwi proteins and piRNAs are specifically expressed in the germline, it may be the case that the Piwi-piRNA system has come to regulate nuclear architecture in these inheritable populations. It is tempting to speculate that gene expression and nuclear localization of chromatin is designed to account for TE regulation by Piwi-piRNAs. Inversely, TEs may even function as 'functional motifs' in order to rewire nuclear architecture. The dynamics of heterochromatin regulation by the Piwi-piRNA pathway therefore provides insight into complex gene regulation controlled by TEs and even impacts the genome at the level of nuclear architecture (Iwasaki, 2021).

    piRNA-independent transposon silencing by the Drosophila THO complex

    piRNAs guide Piwi/Panoramix-dependent H3K9me3 chromatin modification and transposon silencing during Drosophila germline development. The THO RNA export complex is composed of Hpr1, Tho2, and Thoc5-7. Null thoc7 mutations, which displace Thoc5 and Thoc6 from a Tho2-Hpr1 subcomplex, reduce expression of a subset of germline piRNAs and increase transposon expression, suggesting that THO silences transposons by promoting piRNA biogenesis. This study shows that the thoc7-null mutant combination increases transposon transcription but does not reduce anti-sense piRNAs targeting half of the transcriptionally activated transposon families. These mutations also fail to reduce piRNA-guided H3K9me3 chromatin modification or block Panoramix-dependent silencing of a reporter transgene, and unspliced transposon transcripts co-precipitate with THO through a Piwi- and Panoramix-independent mechanism. Mutations in piwi also dominantly enhance germline defects associated with thoc7-null alleles. THO thus functions in a piRNA-independent transposon-silencing pathway, which acts cooperatively with Piwi to support germline development (Zhang, 2021). <

    Vasa sequesters transposon mRNAs in nuage via phase separation requiring RNA binding and self-association

    Bombyx Vasa (BmVasa) assembles non-membranous organelle, nuage or Vasa bodies, in germ cells, known as the center for Siwi-dependent transposon silencing and concomitant Ago3-piRISC biogenesis. However, details of the body assembly remain unclear. This study shows that the N-terminal intrinsically disordered region (N-IDR) and RNA helicase domain of BmVasa are responsible for self-association and RNA binding, respectively, but N-IDR is also required for full RNA-binding activity. Both domains are essential for Vasa body assembly in vivo and droplet formation in vitro via phase separation. FAST-iCLIP reveals that BmVasa preferentially binds transposon mRNAs. Loss of Siwi function derepresses transposons but has marginal effects on BmVasa-RNA binding. This study shows that BmVasa assembles nuage by phase separation via its ability to self-associate and bind newly exported transposon mRNAs. This unique property of BmVasa allows transposon mRNAs to be sequestered and enriched in nuage, resulting in effective Siwi-dependent transposon repression and Ago3-piRISC biogenesis (Yamazaki, 2023).


    REFERENCES

    Azpiazu, N. and Morata, G. (2022). Chromatin remodelling and retrotransposons activities during regeneration in Drosophila. Dev Biol 482: 7-16. PubMed ID: 34822846

    Barckmann, B., El-Barouk, M., Pelisson, A., Mugat, B., Li, B., Franckhauser, C., Fiston Lavier, A. S., Mirouze, M., Fablet, M. and Chambeyron, S. (2018). The somatic piRNA pathway controls germline transposition over generations. Nucleic Acids Res 46(18): 9524-9536. PubMed ID: 30312469

    Berloco, M. F., Minervini, C. F., Moschetti, R., Palazzo, A., Viggiano, L. and Marsano, R. M. (2021). Evidence of the Physical Interaction between Rpl22 and the Transposable Element Doc5, a Heterochromatic Transposon of Drosophila melanogaster. Genes (Basel) 12(12). PubMed ID: 34946947

    Bozzetti, M.P., Specchia, V., Cattenoz, P.B., Laneve, P., Geusa, A., Sahin, H.B., Di Tommaso, S., Friscini, A., Massari, S., Diebold, C. and Giangrande, A. (2015). The Drosophila fragile X mental retardation protein participates in the piRNA pathway. J Cell Sci 128: 2070-2084. PubMed ID: 25908854

    Cao, J., Yu, T., Xu, B., Hu, Z., Zhang, X. O., Theurkauf, W. E. and Weng, Z. (2023). Epigenetic and chromosomal features drive transposon insertion in Drosophila melanogaster. Nucleic Acids Res 51(5): 2066-2086. PubMed ID: 36762470

    Cappucci, U., Noro, F., Casale, A. M., Fanti, L., Berloco, M., Alagia, A. A., Grassi, L., Le Pera, L., Piacentini, L. and Pimpinelli, S. (2019). The Hsp70 chaperone is a major player in stress-induced transposable element activation. Proc Natl Acad Sci U S A 116(36): 17943-17950. PubMed ID: 31399546

    Castillo, D. M. and Moyle, L. C. (2022). Hybrid incompatibility between Drosophila virilis and D. lummei is stronger in the presence of transposable elements. J Evol Biol 35(10): 1319-1334. PubMed ID: 35988129

    Chang, T. H., Mattei, E., Gainetdinov, I., Colpan, C., Weng, Z. and Zamore, P. D. (2018). Maelstrom represses canonical Polymerase II transcription within bi-directional piRNA clusters in Drosophila melanogaster. Mol Cell. PubMed ID: 30527661

    Chang, Y. H., Keegan, R. M., Prazak, L. and Dubnau, J. (2019). Cellular labeling of endogenous retrovirus replication (CLEVR) reveals de novo insertions of the gypsy retrotransposable element in cell culture and in both neurons and glial cells of aging fruit flies. PLoS Biol 17(5): e3000278. PubMed ID: 31095565

    Chaouch, A., Berlandi, J., Chen, C. C. L., Frey, F., Badini, S., Harutyunyan, A. S., Chen, X., Krug, B., Hebert, S., Jeibmann, A., Lu, C., Kleinman, C. L., Hasselblatt, M., Lasko, P., Shirinian, M. and Jabado, N. (2021). Histone H3.3 K27M and K36M mutations de-repress transposable elements through perturbation of antagonistic chromatin marks. Mol Cell. PubMed ID: 34739871

    Chen, P., Kotov, A. A., Godneeva, B. K., Bazylev, S. S., Olenina, L. V. and Aravin, A. A. (2021). piRNA-mediated gene regulation and adaptation to sex-specific transposon expression in D. melanogaster male germline. Genes Dev. PubMed ID: 33985970

    Cordeiro, J., Carvalho, T. L., Valente, V. and Robe, L. J. (2019). Evolutionary history and classification of Micropia retroelements in Drosophilidae species. PLoS One 14(10): e0220539. PubMed ID: 31622354

    Diaz-Gonzalez, J. and Dominguez, A. (2020). Different structural variants of roo retrotransposon are active in Drosophila melanogaster. Gene 741: 144546. PubMed ID: 32165306

    Dominguez, A. (2021). Interrogating the 5'UTR tandem repeats of retrotransposon roo of Drosophila about horizontal transfer. Genetica. PubMed ID: 33900494

    Dorador, A. P., Dalikova, M., Cerbin, S., Stillman, C. M., Zych, M. G., Hawley, R. S., Miller, D. E., Ray, D. A., Funikov, S. Y., Evgen'ev, M. B. and Blumenstiel, J. P. (2022). Paramutation-like Epigenetic Conversion by piRNA at the Telomere of Drosophila virilis. Biology (Basel) 11(10). PubMed ID: 36290385

    Duc, C., Yoth, M., Jensen, S., Mouniee, N., Bergman, C. M., Vaury, C. and Brasset, E. (2019). Trapping a somatic endogenous retrovirus into a germline piRNA cluster immunizes the germline against further invasion. Genome Biol 20(1): 127. PubMed ID: 31227013

    Durdevic, Z., Pillai, R. S. and Ephrussi, A. (2018). Transposon silencing in the Drosophila female germline is essential for genome stability in progeny embryos. Life Sci Alliance 1(5): e201800179. PubMed ID: 30456388

    Eastwood, E. L., Jara, K. A., Bornelov, S., Munafo, M., Frantzis, V., Kneuss, E., Barbar, E. J., Czech, B. and Hannon, G. J. (2021). Dimerisation of the PICTS complex via LC8/Cut-up drives co-transcriptional transposon silencing in Drosophila. Elife 10. PubMed ID: 33538693

    Ellison, C. E. and Cao, W. (2019). Nanopore sequencing and Hi-C scaffolding provide insight into the evolutionary dynamics of transposable elements and piRNA production in wild strains of Drosophila melanogaster. Nucleic Acids Res 117(1):464-471. PubMed ID: 31754714

    Ellison, C. E., Kagda, M. S. and Cao, W. (2020). Telomeric TART elements target the piRNA machinery in Drosophila. PLoS Biol 18(12): e3000689. PubMed ID: 33347429

    Erwin, A. A., Galdos, M. A., Wickersheim, M. L., Harrison, C. C., Marr, K. D., Colicchio, J. M. and Blumenstiel, J. P. (2015). piRNAs are associated with diverse transgenerational effects on gene and transposon expression in a hybrid dysgenic syndrome of D. virilis. PLoS Genet 11: e1005332. PubMed ID: 26241928

    Fabian, D. K., Melike Donertaş, H., Fuentealba, M., Partridge, L. and Thornton, J. M. (2021). Transposable element landscape in Drosophila populations selected for longevity. Genome Biol Evol. PubMed ID: 33595657

    Fablet, M., Jacquet, A., Rebollo, R., Haudry, A., Rey, C., Salces-Ortiz, J., Bajad, P., Burlet, N., Jantsch, M. F., Garcia Guerreiro, M. P. and Vieira, C. (2019). Dynamic Interactions between the genome and an endogenous retrovirus: Tirant in Drosophila simulans wild-type strains. G3 (Bethesda). PubMed ID: 30658967

    Fabry, M. H., Falconio, F. A., Joud, F., Lythgoe, E. K., Czech, B. and Hannon, G. J. (2021). Maternally inherited piRNAs direct transient heterochromatin formation at active transposons during early Drosophila embryogenesis. Elife 10. PubMed ID: 34236313

    Fefelova, E. A., Pleshakova, I. M., Mikhaleva, E. A., Pirogov, S. A., Poltorachenko, V. A., Abramov, Y. A., Romashin, D. D., Shatskikh, A. S., Blokh, R. S., Gvozdev, V. A. and Klenov, M. S. (2022). Impaired function of rDNA transcription initiation machinery leads to derepression of ribosomal genes with insertions of R2 retrotransposon. Nucleic Acids Res 50(2): 867-884. PubMed ID: 35037046

    Francis, M. J., Roche, S., Cho, M. J., Beall, E., Min, B., Panganiban, R. P. and Rio, D. C. (2016). Drosophila IRBP bZIP heterodimer binds P-element DNA and affects hybrid dysgenesis. Proc Natl Acad Sci U S A 113(46): 13003-13008. PubMed ID: 27799520

    Funikov, S. Y., Kulikova, D. A., Krasnov, G. S., Rezvykh, A. P., Chuvakova, L. N., Shostak, N. G., Zelentsova, E. S., Blumenstiel, J. P. and Evgen'ev, M. B. (2018). Spontaneous gain of susceptibility suggests a novel mechanism of resistance to hybrid dysgenesis in Drosophila virilis. PLoS Genet 14(5): e1007400. PubMed ID: 29813067

    Giordani, G., Cavaliere, V., Gargiulo, G., Lattanzi, G. and Andrenacci, D. (2021). Retrotransposons Down- and Up-Regulation in Aging Somatic Tissues. Cells 11(1). PubMed ID: 35011640

    Guio, L., Vieira, C. and Gonzalez, J. (2018). Stress affects the epigenetic marks added by natural transposable element insertions in Drosophila melanogaster. Sci Rep 8(1): 12197. PubMed ID: 30111890

    Han, S., Basting, P. J., Dias, G. B., Luhur, A., Zelhof, A. C. and Bergman, C. M. (2021). Transposable element profiles reveal cell line identity and loss of heterozygosity in Drosophila cell culture. Genetics 219(2). PubMed ID: 34849875

    Hao, Y., Wang, D., Wu, S., Li, X., Shao, C., Zhang, P., Chen, J. Y., Lim, D. H., Fu, X. D., Chen, R. and He, S. (2020). Active retrotransposons help maintain pericentromeric heterochromatin required for faithful cell division. Genome Res 30(11): 1570-1582. PubMed ID: 33060173

    Hemmer, L. W., Dias, G. B., Smith, B., Van Vaerenberghe, K., Howard, A., Bergman, C. M. and Blumenstiel, J. P. (2020). Hybrid dysgenesis in Drosophila virilis results in clusters of mitotic recombination and loss-of-heterozygosity but leaves meiotic recombination unaltered. Mob DNA 11: 10. PubMed ID: 32082426

    Hill, T. and Betancourt, A. J. (2018). Extensive exchange of transposable elements in the Drosophila pseudoobscura group. Mob DNA 9: 20. PubMed ID: 29946370

    Hirakata, S., Ishizu, H., Fujita, A., Tomoe, Y. and Siomi, M. C. (2019). Requirements for multivalent Yb body assembly in transposon silencing in Drosophila. EMBO Rep 20(7): e47708. PubMed ID: 31267711

    Iwasaki, Y. W., Sriswasdi, S., Kinugasa, Y., Adachi, J., Horikoshi, Y., Shibuya, A., Iwasaki, W., Tashiro, S., Tomonaga, T. and Siomi, H. (2021). Piwi-piRNA complexes induce stepwise changes in nuclear architecture at target loci. EMBO J: e108345. PubMed ID: 34337769

    Jagannathan, M. and Yamashita, Y. M. (2021). Defective satellite DNA clustering into chromocenters underlies hybrid incompatibility in Drosophila. Mol Biol Evol. PubMed ID: 34302471

    Jankovics, F., Bence, M., Sinka, R., Farago, A., Bodai, L., Pettko-Szandtner, A., Ibrahim, K., Takacs, Z., Szarka-Kovacs, A. B. and Erdelyi, M. (2018). Drosophila small ovary gene is required for transposon silencing and heterochromatin organisation and ensures germline stem cell maintenance and differentiation. Development. PubMed ID: 30389853

    Keegan, R. M., Talbot, L. R., Chang, Y. H., Metzger, M. J. and Dubnau, J. (2021). Intercellular viral spread and intracellular transposition of Drosophila gypsy. PLoS Genet 17(4): e1009535. PubMed ID: 33886543

    Kelleher, E. S., Azevedo, R. B. R. and Zheng, Y. (2018). The evolution of small-RNA-mediated silencing of an invading transposable element. Genome Biol Evol. PubMed ID: 30252073

    Kneuss, E., Munafo, M., Eastwood, E. L., Deumer, U. S., Preall, J. B., Hannon, G. J. and Czech, B. (2019). Specialization of the Drosophila nuclear export family protein Nxf3 for piRNA precursor export. Genes Dev 33(17-18): 1208-1220. PubMed ID: 31416967

    Kofler, R. (2019). Dynamics of transposable element invasions with piRNA clusters. Mol Biol Evol. PubMed ID: 30968135

    Kofler, R., Nolte, V. and Schlotterer, C. (2022). The transposition rate has little influence on the plateauing level of the P-element. Mol Biol Evol. PubMed ID: 35731857

    Kordyukova, M., Morgunova, V., Olovnikov, I., Komarov, P. A., Mironova, A., Olenkina, O. M. and Kalmykova, A. (2018). Subcellular localization and Egl-mediated transport of telomeric retrotransposon HeT-A ribonucleoprotein particles in the Drosophila germline and early embryogenesis. PLoS One 13(8): e0201787. PubMed ID: 30157274

    Lerat, E., Goubert, C., Guirao-Rico, S., Merenciano, M., Dufour, A. B., Vieira, C. and Gonzalez, J. (2018). Population specific dynamics and selection patterns of transposable element insertions in European natural populations. Mol Ecol. PubMed ID: 30506554

    Le Rouzic, A. and Deceliere, G. (2005). Models of the population genetics of transposable elements. Genet Res 85(3): 171-181. PubMed ID: 16174335

    Lewerentz, J., Johansson, A. M., Larsson, J. and Stenberg, P. (2022). Transposon activity, local duplications and propagation of structural variants across haplotypes drive the evolution of the Drosophila S2 cell line. BMC Genomics 23(1): 276. PubMed ID: 35392795

    Li, W., Prazak, L., Chatterjee, N., Gruninger, S., Krug, L., Theodorou, D. and Dubnau, J. (2013). Activation of transposable elements during aging and neuronal decline in Drosophila. Nat Neurosci 16: 529-531. PubMed ID: 23563579

    Luo, S., Zhang, H., Duan, Y., Yao, X., Clark, A. G. and Lu, J. (2020). The evolutionary arms race between transposable elements and piRNAs in Drosophila melanogaster. BMC Evol Biol 20(1): 14. PubMed ID: 31992188

    Marin, P., Jaquet, A., Picarle, J., Fablet, M., Merel, V., Delignette-Muller, M. L., Ferrarini, M. G., Gibert, P. and Vieira, C. (2021). Phenotypic and Transcriptomic Responses to Stress Differ According to Population Geography in an Invasive Species. Genome Biol Evol 13(9). PubMed ID: 34505904

    McGurk, M. P. and Barbash, D. A. (2018). Double insertion of transposable elements provides a substrate for the evolution of satellite DNA. Genome Res 28(5): 714-725. PubMed ID: 29588362

    Mishra, R., Kunar, R., Mandal, L., Alone, D. P., Chandrasekharan, S., Tiwari, A. K., Tapadia, M. G., Mukherjee, A. and Roy, J. K. (2020). A Forward Genetic Approach to Mapping a P-Element Second Site Mutation Identifies DCP2 as a Novel Tumour Suppressor in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 32591349

    Mohamed, M., Dang, N. T., Ogyama, Y., Burlet, N., Mugat, B., Boulesteix, M., Merel, V., Veber, P., Salces-Ortiz, J., Severac, D., Pelisson, A., Vieira, C., Sabot, F., Fablet, M. and Chambeyron, S. (2020). A Transposon Story: From TE Content to TE Dynamic Invasion of Drosophila Genomes Using the Single-Molecule Sequencing Technology from Oxford Nanopore. Cells 9(8). PubMed ID: 32722451

    Moon, S., Cassani, M., Lin, Y. A., Wang, L., Dou, K. and Zhang, Z. Z. (2018). A robust transposon-endogenizing response from germline stem cells. Dev Cell. PubMed ID: 30393075

    Muotri, A. R., Chu, V. T., Marchetto, M. C., Deng, W., Moran, J. V. and Gage, F. H. (2005). Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition. Nature 435: 903-910. PubMed ID: 15959507

    Murota, Y., Ishizu, H., Nakagawa, S., Iwasaki, Y. W., Shibata, S., Kamatani, M. K., Saito, K., Okano, H., Siomi, H. and Siomi, M. C. (2014) Yb integrates piRNA intermediates and processing factors into perinuclear bodies to enhance piRISC assembly. Cell Rep 8: 103-113. PubMed ID: 24953657

    Napoletano, F., Ferrari Bravo, G., Voto, I. A. P., Santin, A., Celora, L., Campaner, E., Dezi, C., Bertossi, A., Valentino, E., Santorsola, M., Rustighi, A., Fajner, V., Maspero, E., Ansaloni, F., Cancila, V., Valenti, C. F., Santo, M., Artimagnella, O. B., Finaurini, S., Gioia, U., Polo, S., Sanges, R., Tripodo, C., Mallamaci, A., Gustincich, S., d'Adda di Fagagna, F., Mantovani, F., Specchia, V. and Del Sal, G. (2021). The prolyl-isomerase PIN1 is essential for nuclear Lamin-B structure and function and protects heterochromatin under mechanical stress. Cell Rep 36(11): 109694. PubMed ID: 34525372

    Nguyen, A. H. and Bachtrog, D. (2021). Toxic Y chromosome: Increased repeat expression and age-associated heterochromatin loss in male Drosophila with a young Y chromosome. PLoS Genet 17(4): e1009438. PubMed ID: 33886541

    Nguyen, A. H., Wang, W., Chong, E., Chatla, K. and Bachtrog, D. (2022). Transposable element accumulation drives size differences among polymorphic Y Chromosomes in Drosophila. Genome Res 32(6): 1074-1088. PubMed ID: 35501131

    Pai, C. Y., Lei, E. P., Ghosh, D. and Corces, V. G. (2004). The centrosomal protein CP190 is a component of the gypsy chromatin insulator. Mol Cell 16(5): 737-748. PubMed ID: 15574329

    Perrat, P. N., DasGupta, S., Wang, J., Theurkauf, W., Weng, Z., Rosbash, M. and Waddell, S. (2013). Transposition-driven genomic heterogeneity in the Drosophila brain. Science 340: 91-95. PubMed ID: 23559253

    Roy, M., Viginier, B., Saint-Michel, E., Arnaud, F., Ratinier, M. and Fablet, M. (2020). Viral infection impacts transposable element transcript amounts in Drosophila. Proc Natl Acad Sci U S A 117(22): 12249-12257. PubMed ID: 32434916

    Roy, M., Viginier, B., Mayeux, C. A., Ratinier, M. and Fablet, M. (2021). Infections by Transovarially Transmitted DMelSV in Drosophila Have No Impact on Ovarian Transposable Element Transcripts but Increase Their Amounts in the Soma. Genome Biol Evol 13(9). PubMed ID: 34498066

    Russo, J., Harrington, A. W. and Steiniger, M. (2015). Antisense transcription of retrotransposons in Drosophila: The origin of endogenous small interfering RNA precursors. Genetics [Epub ahead of print]. PubMed ID: 26534950

    Guida, V., Cernilogar, F. M., Filograna, A., De Gregorio, R., Ishizu, H., Siomi, M. C., Schotta, G., Bellenchi, G. C. and Andrenacci, D. (2016). Production of small non-coding RNAs from the flamenco locus is regulated by the gypsy retrotransposon of Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 27558137

    Manee, M. M., Jackson, J. and Bergman, C. M. (2018). Conserved noncoding elements influence the transposable element landscape in Drosophila. Genome Biol Evol. PubMed ID: 29850787

    Munafo, M., Lawless, V. R., Passera, A., MacMillan, S., Bornelov, S., Haussmann, I. U., Soller, M., Hannon, G. J. and Czech, B. (2021). Channel Nuclear Pore Complex subunits are required for transposon silencing in Drosophila. Elife 10. PubMed ID: 33856346

    Radion, E., Sokolova, O., Ryazansky, S., Komarov, P. A., Abramov, Y. and Kalmykova, A. (2019). The integrity of piRNA clusters is abolished by insulators in the Drosophila germline. Genes (Basel) 10(3). PubMed ID: 30862119

    Rahman, R., Chirn, G. W., Kanodia, A., Sytnikova, Y. A., Brembs, B., Bergman, C. M. and Lau, N. C. (2015). Unique transposon landscapes are pervasive across Drosophila melanogaster genomes. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 26578579

    Rech, G. E., Bogaerts-Marquez, M., Barron, M. G., Merenciano, M., Villanueva-Canas, J. L., Horvath, V., Fiston-Lavier, A. S., Luyten, I., Venkataram, S., Quesneville, H., Petrov, D. A. and Gonzalez, J. (2019). Stress response, behavior, and development are shaped by transposable element-induced mutations in Drosophila. PLoS Genet 15(2): e1007900. PubMed ID: 30753202

    Russo, J., Harrington, A. W. and Steiniger, M. (2015). Antisense transcription of retrotransposons in Drosophila: The origin of endogenous small interfering RNA precursors. Genetics 202(1):107-21. PubMed ID: 26534950

    Ryazansky, S., Radion, E., Mironova, A., Akulenko, N., Abramov, Y., Morgunova, V., Kordyukova, M.Y., Olovnikov, I. and Kalmykova, A. (2017). Natural variation of piRNA expression affects immunity to transposable elements. PLoS Genet 13: e1006731. PubMed ID: 28448516

    Said, I., McGurk, M. P., Clark, A. G. and Barbash, D. A. (2022). Patterns of piRNA Regulation in Drosophila Revealed through Transposable Element Clade Inference. Mol Biol Evol 39(1). PubMed ID: 34921315

    Salces-Ortiz, J., Vargas-Chavez, C., Guio, L., Rech, G. E. and Gonzalez, J. (2020). Transposable elements contribute to the genomic response to insecticides in Drosophila melanogaster. Philos Trans R Soc Lond B Biol Sci 375(1795): 20190341. PubMed ID: 32075557

    Savva, Y. A., Jepson, J. E., Chang, Y. J., Whitaker, R., Jones, B. C., St Laurent, G., Tackett, M. R., Kapranov, P., Jiang, N., Du, G., Helfand, S. L. and Reenan, R. A. (2013). RNA editing regulates transposon-mediated heterochromatic gene silencing. Nat Commun 4: 2745. PubMed ID: 24201902

    Schnabl, J., Wang, J., Hohmann, U., Gehre, M., Batki, J., Andreev, V. I., Purkhauser, K., Fasching, N., Duchek, P., Novatchkova, M., Mechtler, K., Plaschka, C., Patel, D. J. and Brennecke, J. (2021). Molecular principles of Piwi-mediated cotranscriptional silencing through the dimeric SFiNX complex. Genes Dev 35(5-6): 392-409. PubMed ID: 33574069

    Schneider, B. K., Sun, S., Lee, M., Li, W., Skvir, N., Neretti, N., Vijg, J. and Secombe, J. (2023). Expression of retrotransposons contributes to aging in Drosophila. Genetics. PubMed ID: 37084379

    Serrato-Capuchina, A., Wang, J., Earley, E., Peede, D., Isbell, K. and Matute, D. R. (2020). Paternally inherited P-element copy number affects the magnitude of hybrid dysgenesis in Drosophila simulans and D. melanogaster. Genome Biol Evol. PubMed ID: 32339225

    Sokolova, O. A., Mikhaleva, E. A., Kharitonov, S. L., Abramov, Y. A., Gvozdev, V. A. and Klenov, M. S. (2020). Special vulnerability of somatic niche cells to transposable element activation in Drosophila larval ovaries. Sci Rep 10(1): 1076. PubMed ID: 31974416

    Sun, W, Samimi, H., Gamez, M., Zare, H. and Frost, B. (2018). Pathogenic tau-induced piRNA depletion promotes neuronal death through transposable element dysregulation in neurodegenerative tauopathies. Nat Neurosci 21(8): 1038-1048. PubMed ID: 30038280

    Szakmary, A., Reedy, M., Qi, H. and Lin, H. (2009). The Yb protein defines a novel organelle and regulates male germline stem cell self-renewal in Drosophila melanogaster. J Cell Biol 185: 613-627. PubMed ID: 19433453

    Ullastres, A., Merenciano, M. and Gonzalez, J. (2021). Regulatory regions in natural transposable element insertions drive interindividual differences in response to immune challenges in Drosophila. Genome Biol 22(1): 265. PubMed ID: 34521452

    Tambones, I. L., Haudry, A., Simao, M. C. and Carareto, C. M. A. (2019). High frequency of horizontal transfer in Jockey families (LINE order) of drosophilids. Mob DNA 10: 43. PubMed ID: 31709017

    Teixeira, F. K., Okuniewska, M., Malone, C. D., Coux, R. X., Rio, D. C. and Lehmann, R. (2017). piRNA-mediated regulation of transposon alternative splicing in the soma and germ line. Nature 552(7684): 268-272. PubMed ID: 29211718

    Treiber, C. D. and Waddell, S. (2020). Transposon expression in the Drosophila brain is driven by neighboring genes and diversifies the neural transcriptome. Genome Res. PubMed ID: 32973040

    Villanueva-Canas, J. L., Horvath, V., Aguilera, L. and Gonzalez, J. (2019). Diverse families of transposable elements affect the transcriptional regulation of stress-response genes in Drosophila melanogaster. Nucleic Acids Res. PubMed ID: 31175824

    Wang, L., Tracy, L., Su, W., Yang, F., Feng, Y., Silverman, N. and Zhang, Z. Z. Z. (2022). Retrotransposon activation during Drosophila metamorphosis conditions adult antiviral responses. Nat Genet. PubMed ID: 36396707

    Wei, K. H., Gibilisco, L. and Bachtrog, D. (2020). Epigenetic conflict on a degenerating Y chromosome increases mutational burden in Drosophila males. Nat Commun 11(1): 5537. PubMed ID: 33139741

    Wei, K. H., Chan, C. and Bachtrog, D. (2021). Establishment of H3K9me3-dependent heterochromatin during embryogenesis in Drosophila miranda. Elife 10. PubMed ID: 34128466

    Wei, K. H., Mai, D., Chatla, K. and Bachtrog, D. (2022). Dynamics and Impacts of Transposable Element Proliferation in the Drosophila nasuta Species Group Radiation. Mol Biol Evol 39(5). PubMed ID: 35485457

    Xie, W., Sowemimo, I., Hayashi, R., Wang, J., Burkard, T. R., Brennecke, J., Ameres, S. L. and Patel, D. J. (2020). Structure-function analysis of microRNA 3'-end trimming by Nibbler. Proc Natl Acad Sci U S A. PubMed ID: 33199607

    Yamazaki, H., Namba, Y., Kuriyama, S., Nishida, K. M., Kajiya, A. and Siomi, M. C. (2023). Bombyx Vasa sequesters transposon mRNAs in nuage via phase separation requiring RNA binding and self-association. Nat Commun 14(1): 1942. PubMed ID: 37029111

    Yang, W. R., Ardeljan, D., Pacyna, C. N., Payer, L. M. and Burns, K. H. (2019). SQuIRE reveals locus-specific regulation of interspersed repeat expression. Nucleic Acids Res. PubMed ID: 30624635

    Yang, N., Srivastav, S. P., Rahman, R., Ma, Q., Dayama, G., Li, S., Chinen, M., Lei, E. P., Rosbash, M. and Lau, N. C. (2022). Transposable element landscapes in aging Drosophila. PLoS Genet 18(3): e1010024. PubMed ID: 35239675

    Yushkova, E. (2020). Involvement of DNA Repair Genes and System of Radiation-Induced Activation of Transposons in Formation of Transgenerational Effects. Front Genet 11: 596947. PubMed ID: 33329741

    Yushkova, E. (2022a). Contribution of transposable elements to transgenerational effects of chronic radioactive exposure of natural populations of Drosophila melanogaster living for a long time in the zone of the Chernobyl nuclear disaster. J Environ Radioact 251-252: 106945. PubMed ID: 35696883

    Yushkova, E. A. (2022b). The effects of transpositions of functional I retrotransposons depend on the conditions and dose of parental exposure. Int J Radiat Biol: 1-13. PubMed ID: 36318749

    Zanni, V., Eymery, A., Coiffet, M., Zytnicki, M., Luyten, I., Quesneville, H., Vaury, C. and Jensen, S. (2013). Distribution, evolution, and diversity of retrotransposons at the flamenco locus reflect the regulatory properties of piRNA clusters. Proc Natl Acad Sci U S A 110: 19842-19847. PubMed ID: 24248389

    Zhang, G., Yu, T., Parhad, S. S., Ho, S., Weng, Z. and Theurkauf, W. E. (2021). piRNA-independent transposon silencing by the Drosophila THO complex. Dev Cell 56(18): 2623-2635. PubMed ID: 34547226


    Home page: The Interactive Fly © 2018 Thomas B. Brody, Ph.D.

    The Interactive Fly resides on the
    Society for Developmental Biology's Web server.