Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Development ; 150(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37317951

ABSTRACT

Insulators are architectural elements implicated in the organization of higher-order chromatin structures and transcriptional regulation. However, it is still unknown how insulators contribute to Drosophila telomere maintenance. Although the Drosophila telomeric retrotransposons HeT-A and TART occupy a common genomic niche, they are regulated independently. TART elements are believed to provide reverse transcriptase activity, whereas HeT-A transcripts serve as a template for telomere elongation. Here, we report that insulator complexes associate with TART and contribute to its transcriptional regulation in the Drosophila germline. Chromatin immunoprecipitation revealed that the insulator complex containing BEAF32, Chriz, and DREF proteins occupy the TART promoter. BEAF32 depletion causes derepression and chromatin changes at TART in ovaries. Moreover, an expansion of TART copy number was observed in the genome of the BEAF32 mutant strain. BEAF32 localizes between the TART enhancer and promoter, suggesting that it blocks enhancer-promoter interactions. Our study found that TART repression is released in the germ cysts as a result of the normal reduction of BEAF32 expression at this developmental stage. We suggest that coordinated expression of telomeric repeats during development underlies telomere elongation control.


Subject(s)
Drosophila , Retroelements , Animals , Drosophila/genetics , Retroelements/genetics , Telomere/genetics , Chromatin , Germ Cells
2.
Methods Mol Biol ; 2509: 157-169, 2022.
Article in English | MEDLINE | ID: mdl-35796963

ABSTRACT

The RNA fluorescence in situ hybridization (FISH) technique combined with immunostaining is a powerful method to visualize a specific transcript and a protein of interest simultaneously. Although whole-mount RNA FISH is routinely used to determine RNA intracellular localization, a detailed picture of RNA distribution in complex tissues remains a challenge. The main problem is the various permeability of morphologically different cells within a tissue. We overcome this challenge by developing an approach based on differential permeabilization treatment of tissue specimens. We have tested and optimized conditions for RNA FISH combined with immunofluorescent staining (RNA FISH/IF) to detect the maternal telomeric retrotransposon HeT-A RNPs in the Drosophila ovaries and syncytial embryos. Methods described here are applicable to a broad variety of biological tissue specimens.


Subject(s)
Drosophila , RNA , Animals , Drosophila/genetics , Drosophila/metabolism , Fluorescent Antibody Technique , Germ Cells/metabolism , In Situ Hybridization, Fluorescence/methods , RNA/genetics , Ribonucleoproteins/genetics
3.
Biochemistry (Mosc) ; 87(12): 1600-1610, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36717449

ABSTRACT

Chromatin spatial organization in the nucleus is essential for the genome functioning and regulation of gene activity. The nuclear lamina and lamina-associated proteins, lamins, play a key role in this process. Lamin dysfunction leads to the decompaction and transcriptional activation of heterochromatin, which is associated with the premature aging syndrome. In many cell types, telomeres are located at the nuclear periphery, where their replication and stability are ensured by the nuclear lamina. Moreover, diseases associated with defects in lamins and telomeres have similar manifestations and resemble physiological aging. Understanding molecular changes associated with aging at the organismal level is especially important. In this study, we compared the effects caused by the mutation in lamin B and physiological aging in the germline of the model organism Drosophila melanogaster. We have shown that the impaired localization of lamin B leads to the heterochromatin decompaction and transcriptional activation of some transposable elements and telomeric repeats. Both DNA damage and activation of homologous recombination in the telomeres were observed in the germ cells of lamin B mutants. The instability of repeat-enriched heterochromatin can be directly related to the genome destabilization, germ cell death, and sterility observed in lamin B mutants. Similar processes were observed in Drosophila germline in the course of physiological aging, which indicates a close link between the maintenance of the heterochromatin stability at the nuclear periphery and mechanisms of aging.


Subject(s)
Drosophila , Lamin Type B , Animals , Lamin Type B/genetics , Lamin Type B/metabolism , Drosophila/genetics , Heterochromatin , Drosophila melanogaster/genetics , Aging/genetics , Telomere/genetics , Telomere/metabolism , Germ Cells
4.
PLoS One ; 16(10): e0258156, 2021.
Article in English | MEDLINE | ID: mdl-34624021

ABSTRACT

Telomeres are nucleoprotein complexes that protect the ends of eukaryotic linear chromosomes from degradation and fusions. Telomere dysfunction leads to cell growth arrest, oncogenesis, and premature aging. Telomeric RNAs have been found in all studied species; however, their functions and biogenesis are not clearly understood. We studied the mechanisms of development disorders observed upon overexpression of telomeric repeats in Drosophila. In somatic cells, overexpression of telomeric retrotransposon HeT-A is cytotoxic and leads to the accumulation of HeT-A Gag near centrosomes. We found that RNA and RNA-binding protein Gag encoded by the telomeric retrotransposon HeT-A interact with Polo and Cdk1 mitotic kinases, which are conserved regulators of centrosome biogenesis and cell cycle. The depletion of proteins Spindle E, Ccr4 or Ars2 resulting in HeT-A overexpression in the germline was accompanied by mislocalization of Polo as well as its abnormal stabilization during oogenesis and severe deregulation of centrosome biogenesis leading to maternal-effect embryonic lethality. These data suggest a mechanistic link between telomeric HeT-A ribonucleoproteins and cell cycle regulators that ensures the cell response to telomere dysfunction.


Subject(s)
Centrosome/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Embryonic Development , Oogenesis , Protein Serine-Threonine Kinases/metabolism , Telomere/metabolism , Animals , Cell Death , Centrioles/metabolism , Embryo, Nonmammalian/metabolism , Mitosis , Protein Binding , RNA/metabolism , Retroelements/genetics , Ribonucleoproteins/metabolism , Zygote/metabolism
5.
Nucleic Acids Res ; 48(1): 141-156, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31724732

ABSTRACT

Ccr4-Not is a highly conserved complex involved in cotranscriptional RNA surveillance pathways in yeast. In Drosophila, Ccr4-Not is linked to the translational repression of miRNA targets and the posttranscriptional control of maternal mRNAs during oogenesis and embryonic development. Here, we describe a new role for the Ccr4-Not complex in nuclear RNA metabolism in the Drosophila germline. Ccr4 depletion results in the accumulation of transposable and telomeric repeat transcripts in the fraction of chromatin-associated RNA; however, it does not affect small RNA levels or the heterochromatin state of the target loci. Nuclear targets of Ccr4 mainly comprise active full-length transposable elements (TEs) and telomeric and subtelomeric repeats. Moreover, Ccr4-Not foci localize at telomeres in a Piwi-dependent manner, suggesting a functional relationship between these pathways. Indeed, we detected interactions between the components of the Ccr4-Not complex and piRNA machinery, which indicates that these pathways cooperate in the nucleus to recognize and degrade TE transcripts at transcription sites. These data reveal a new layer of transposon control in the germline, which is critical for the maintenance of genome integrity.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Endopeptidases/genetics , Genome, Insect , Ovum/metabolism , RNA, Messenger/genetics , Ribonucleases/genetics , Animals , Chromatin/chemistry , Chromatin/metabolism , DNA Transposable Elements , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Embryonic Development , Endopeptidases/metabolism , Female , Gene Expression Regulation, Developmental , Oogenesis/genetics , Ovum/cytology , Ovum/growth & development , RNA Stability , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribonucleases/metabolism , Telomere/chemistry , Telomere/metabolism
6.
PLoS One ; 13(8): e0201787, 2018.
Article in English | MEDLINE | ID: mdl-30157274

ABSTRACT

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. We study the biogenesis of transcripts generated by the major Drosophila telomere repeat HeT-A in oogenesis and early development with disrupted telomeric repeat silencing. In wild type ovaries, HeT-A expression is downregulated by the Piwi-interacting RNAs (piRNAs). By repressing piRNA pathway, we show 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, we 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, we suggest that its abnormal localization likely leads to impaired embryonic axis specification typical for piRNA pathway mutants.


Subject(s)
Drosophila Proteins/metabolism , Embryonic Development , Gene Products, gag/metabolism , Oogenesis , Retroelements , Animals , Animals, Genetically Modified , Biological Transport , Cell Nucleus/metabolism , Cytoplasm/metabolism , Drosophila , Female , Gene Expression Regulation, Developmental , Ovary/cytology , Ovary/metabolism , Ovum/cytology , Ovum/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Ribonucleoproteins/metabolism , Telomere/metabolism
7.
Epigenetics Chromatin ; 11(1): 40, 2018 07 12.
Article in English | MEDLINE | ID: mdl-30001204

ABSTRACT

BACKGROUND: Telomeric small RNAs related to PIWI-interacting RNAs (piRNAs) have been described in various eukaryotes; however, their role in germline-specific telomere function remains poorly understood. Using a Drosophila model, we performed an in-depth study of the biogenesis of telomeric piRNAs and their function in telomere homeostasis in the germline. RESULTS: To fully characterize telomeric piRNA clusters, we integrated the data obtained from analysis of endogenous telomeric repeats, as well as transgenes inserted into different telomeric and subtelomeric regions. The small RNA-seq data from strains carrying telomeric transgenes demonstrated that all transgenes belong to a class of dual-strand piRNA clusters; however, their capacity to produce piRNAs varies significantly. Rhino, a paralog of heterochromatic protein 1 (HP1) expressed exclusively in the germline, is associated with all telomeric transgenes, but its enrichment correlates with the abundance of transgenic piRNAs. It is likely that this heterogeneity is determined by the sequence peculiarities of telomeric retrotransposons. In contrast to the heterochromatic non-telomeric germline piRNA clusters, piRNA loss leads to a dramatic decrease in HP1, Rhino, and trimethylated histone H3 lysine 9 in telomeric regions. Therefore, the presence of piRNAs is required for the maintenance of telomere chromatin in the germline. Moreover, piRNA loss causes telomere translocation from the nuclear periphery toward the nuclear interior but does not affect telomere end capping. Analysis of the telomere-associated sequences (TASs) chromatin revealed strong tissue specificity. In the germline, TASs are enriched with HP1 and Rhino, in contrast to somatic tissues, where they are repressed by Polycomb group proteins. CONCLUSIONS: piRNAs play an essential role in the assembly of telomeric chromatin, as well as in nuclear telomere positioning in the germline. Telomeric arrays and TASs belong to a unique type of Rhino-dependent piRNA clusters with transcripts that serve simultaneously as piRNA precursors and as their only targets. Telomeric chromatin is highly sensitive to piRNA loss, implying the existence of a novel developmental checkpoint that depends on telomere integrity in the germline.


Subject(s)
Cell Nucleus/genetics , RNA, Small Interfering/metabolism , Telomere/genetics , Animals , Chromatin/genetics , Chromatin Assembly and Disassembly , Drosophila melanogaster , Germ Cells/chemistry
8.
RNA ; 24(4): 574-584, 2018 04.
Article in English | MEDLINE | ID: mdl-29358235

ABSTRACT

Expression of transposable elements in the germline is controlled by Piwi-interacting (pi) RNAs produced by genomic loci termed piRNA clusters and associated with Rhino, a heterochromatin protein 1 (HP1) homolog. Previously, we have shown that transgenes containing a fragment of the I retrotransposon form de novo piRNA clusters in the Drosophila germline providing suppression of I-element activity. We noted that identical transgenes located in different genomic sites vary considerably in piRNA production and classified them as "strong" and "weak" piRNA clusters. Here, we investigated what chromatin and transcriptional changes occur at the transgene insertion sites after their conversion into piRNA clusters. We found that the formation of a transgenic piRNA cluster is accompanied by activation of transcription from both genomic strands that likely initiates at multiple random sites. The chromatin of all transgene-associated piRNA clusters contain high levels of trimethylated lysine 9 of histone H3 (H3K9me3) and HP1a, whereas Rhino binding is considerably higher at the strong clusters. None of these chromatin marks was revealed at the "empty" sites before transgene insertion. Finally, we have shown that in the nucleus of polyploid nurse cells, the formation of a piRNA cluster at a given transgenic genomic copy works according to an "all-or-nothing" model: either there is high Rhino enrichment or there is no association with Rhino at all. As a result, genomic copies of a weak piRNA transgenic cluster show a mosaic association with Rhino foci, while the majority of strong transgene copies associate with Rhino and are hence involved in piRNA production.


Subject(s)
Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , RNA, Small Interfering/genetics , Transcription, Genetic/genetics , Animals , Animals, Genetically Modified , Chromobox Protein Homolog 5 , Female , Histones/metabolism , Methylation , Protein Binding , Retroelements/genetics , Transcriptional Activation/genetics , Transgenes/genetics
9.
PLoS Genet ; 13(4): e1006731, 2017 04.
Article in English | MEDLINE | ID: mdl-28448516

ABSTRACT

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. Here, we explore 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, we compared ovarian small RNAs between R strains with low and high reactivity and show 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, we focused on two R strains, weak and strong, and showed 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. We hypothesize that hyper-efficient piRNA production is contributing to elimination of a telomeric retrotransposon HeT-A, which we have observed in one particular transposon-resistant R strain.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , DNA Transposable Elements/genetics , Drosophila Proteins/genetics , RNA, Small Interfering/genetics , Telomere/genetics , Animals , Argonaute Proteins/genetics , Argonaute Proteins/immunology , Chromosomal Proteins, Non-Histone/metabolism , DNA Transposable Elements/immunology , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/immunology , Female , Gene Expression Regulation/immunology , Gene Silencing , Genome, Insect , Germ Cells , Heterochromatin/genetics , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/immunology , Telomere/immunology
10.
J Mol Biol ; 429(21): 3280-3289, 2017 10 27.
Article in English | MEDLINE | ID: mdl-27939293

ABSTRACT

PIWI-interacting RNAs (piRNAs) provide the silencing of transposable elements in the germline. Drosophila telomeres are maintained by transpositions of specialized telomeric retroelements. piRNAs generated from sense and antisense transcripts of telomeric elements provide telomere length control in the germline. Previously, we have found that antisense transcription of the major telomeric retroelement HeT-A is initiated upstream of the HeT-A sense transcription start site. Here, we performed a deletion analysis of the HeT-A promoter and show that common regulatory elements are shared by sense and antisense promoters of HeT-A. Therefore, the HeT-A promoter is a bidirectional promoter capable of processive sense and antisense transcription. Ovarian small RNA data show that a solo HeT-A promoter within an euchromatic transgene initiates the divergent transcription of transgenic reporter genes and subsequent processing of these transcripts into piRNAs. These events lead to the formation of a divergent unistrand piRNA cluster at solo HeT-A promoters, in contrast to endogenous telomeres that represent strong dual-strand piRNA clusters. Solo HeT-A promoters are not immunoprecipitated with heterochromatin protein 1 (HP1) homolog Rhino, a marker of the dual-strand piRNA clusters, but are associated with HP1 itself, which provides piRNA-mediated transcriptional repression of the reporter genes. Unlike endogenous dual-strand piRNA clusters, the solo HeT-A promoter does not produce overlapping transcripts. In a telomeric context, however, bidirectional promoters of tandem HeT-A repeats provide a read-through transcription of both genomic strands, followed by Rhi binding. These data indicate that Drosophila telomeres share properties of unistrand and dual-strand piRNA clusters.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Promoter Regions, Genetic/genetics , RNA Precursors/genetics , RNA, Small Interfering/genetics , Retroelements/genetics , Telomere/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Germ Cells , Telomere/metabolism , Transcription, Genetic
12.
Nucleic Acids Res ; 43(18): 8762-73, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26240377

ABSTRACT

The germline-specific role of telomeres consists of chromosome end elongation and proper chromosome segregation during early developmental stages. Despite the crucial role of telomeres in germ cells, little is known about telomere biology in the germline. We analyzed telomere homeostasis in the Drosophila female germline and early embryos. A novel germline-specific function of deadenylase complex Ccr4-Not in the telomeric transcript surveillance mechanism is reported. Depletion of Ccr4-Not complex components causes strong derepression of the telomeric retroelement HeT-A in the germ cells, accompanied by elongation of the HeT-A poly(A) tail. Dysfunction of transcription factors Woc and Trf2, as well as RNA-binding protein Ars2, also results in the accumulation of excessively polyadenylated HeT-A transcripts in ovaries. Germline knockdowns of Ccr4-Not components, Woc, Trf2 and Ars2, lead to abnormal mitosis in early embryos, characterized by chromosome missegregation, centrosome dysfunction and spindle multipolarity. Moreover, the observed phenotype is accompanied by the accumulation of HeT-A transcripts around the centrosomes in early embryos, suggesting the putative relationship between overexpression of telomeric transcripts and mitotic defects. Our data demonstrate that Ccr4-Not, Woc, Trf2 and Ars2, components of different regulatory pathways, are required for telomere protection in the germline in order to guarantee normal development.


Subject(s)
Drosophila/genetics , Gene Expression Regulation, Developmental , Gene Silencing , Retroelements , Telomere , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/embryology , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryonic Development/genetics , Female , Mitosis/genetics , Ovary/metabolism , Ovum/metabolism , Polyadenylation , RNA-Binding Proteins , Ribonucleases/genetics , Ribonucleases/metabolism , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...