Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 7(1): 783, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951619

RESUMEN

Transport of macromolecules through the nuclear envelope (NE) is mediated by nuclear pore complexes (NPCs) consisting of nucleoporins (Nups). Elys/Mel-28 is the Nup that binds and connects the decondensing chromatin with the reassembled NPCs at the end of mitosis. Whether Elys links chromatin with the NE during interphase is unknown. Here, using DamID-seq, we identified Elys binding sites in Drosophila late embryos and divided them into those associated with nucleoplasmic or with NPC-linked Elys. These Elys binding sites are located within active or inactive chromatin, respectively. Strikingly, Elys knockdown in S2 cells results in peripheral chromatin displacement from the NE, in decondensation of NE-attached chromatin, and in derepression of genes within. It also leads to slightly more compact active chromatin regions. Our findings indicate that NPC-linked Elys, together with the nuclear lamina, anchors peripheral chromatin to the NE, whereas nucleoplasmic Elys decompacts active chromatin.


Asunto(s)
Cromatina , Proteínas de Drosophila , Interfase , Proteínas de Complejo Poro Nuclear , Poro Nuclear , Animales , Sitios de Unión , Núcleo Celular/metabolismo , Cromatina/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/embriología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/genética
2.
Development ; 150(12)2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37317951

RESUMEN

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.


Asunto(s)
Drosophila , Retroelementos , Animales , Drosophila/genética , Retroelementos/genética , Telómero/genética , Cromatina , Células Germinativas
3.
Sci Rep ; 12(1): 11191, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35778515

RESUMEN

The nascent polypeptide-associated complex (NAC) consisting of α- and ß-subunits is an essential ribosome-associated protein conserved in eukaryotes. NAC is a ubiquitously expressed co-translational regulator of nascent protein folding and sorting providing for homeostasis of cellular proteins. Here we report on discovering the germline-specific NACαß paralogs (gNACs), whose ß-subunits, non-distinguishable by ordinary immunodetection, are encoded by five highly homologous gene copies, while the α-subunit is encoded by a single αNAC gene. The gNAC expression is detected in the primordial embryonic and adult gonads via immunostaining. The germline-specific α and ß subunits differ from the ubiquitously expressed paralogs by the extended intrinsically disordered regions (IDRs) acquired at the N- and C-termini of the coding regions, predicted to be phosphorylated. The presence of distinct phosphorylated isoforms of gNAC-ß subunits is confirmed by comparing of their profiles by 2D-isoeletrofocusing resolution before and after phosphatase treatment of testis ribosomes. We revealed that the predicted S/T sites of phosphorylation in the individual orthologous IDRs of gNAC-ß sequences of Drosophila species are positionally conserved despite these disordered regions are drastically different. We propose the IDR-dependent molecular crowding and specific coordination of NAC and other proteostasis regulatory factors at the ribosomes of germinal cells. Our findings imply that there may be a functional crosstalk between the germinal and ubiquitous α- and ß-subunits based on assessing their depletion effects on the fly viability and gonad development.


Asunto(s)
Drosophila melanogaster , Proteínas Ribosómicas , Animales , Drosophila , Drosophila melanogaster/genética , Células Germinativas , Masculino , Proteínas Ribosómicas/genética , Ribosomas/genética
4.
Eur J Cell Biol ; 101(3): 151246, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35667338

RESUMEN

The tight interaction between somatic and germline cells is conserved in animal spermatogenesis. The testes of Drosophila melanogaster are the model of choice to identify processes responsible for mature gamete production. However, processes of differentiation and soma-germline interactions occurring in somatic cyst cells are currently understudied. Here we focused on the comparison of transcriptome expression patterns of early and mature somatic cyst cells to find out the developmental changes taking place in them. We employed a FACS-based approach for the isolation of early and mature somatic cyst cells from fly testes, subsequent preparation of RNA-Seq libraries, and analysis of gene differential expression in the sorted cells. We found increased expression of genes involved in cell cycle-related processes in early cyst cells, which is necessary for the proliferation and self-renewal of a crucial population of early cyst cells, cyst stem cells. Genes proposedly required for lamellipodium-like projection organization for proper cyst formation were also detected among the upregulated ones in early cyst cells. Gene Ontology and interactome analyses of upregulated genes in mature cyst cells revealed a striking over-representation of gene categories responsible for metabolic and catabolic cellular processes, as well as genes supporting the energetic state of the cells provided by oxidative phosphorylation that is carried out in mitochondria. Our comparative analyses of differentially expressed genes revealed major peculiarities in early and mature cyst cells and provide novel insight into their regulation, which is important for male fertility.


Asunto(s)
Quistes , Proteínas de Drosophila , Animales , Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Masculino , Espermatogénesis , Testículo/metabolismo
5.
Nucleic Acids Res ; 50(6): 3203-3225, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35166842

RESUMEN

Eukaryotic chromosomes are spatially segregated into topologically associating domains (TADs). Some TADs are attached to the nuclear lamina (NL) through lamina-associated domains (LADs). Here, we identified LADs and TADs at two stages of Drosophila spermatogenesis - in bamΔ86 mutant testes which is the commonly used model of spermatogonia (SpG) and in larval testes mainly filled with spermatocytes (SpCs). We found that initiation of SpC-specific transcription correlates with promoters' detachment from the NL and with local spatial insulation of adjacent regions. However, this insulation does not result in the partitioning of inactive TADs into sub-TADs. We also revealed an increased contact frequency between SpC-specific genes in SpCs implying their de novo gathering into transcription factories. In addition, we uncovered the specific X chromosome organization in the male germline. In SpG and SpCs, a single X chromosome is stronger associated with the NL than autosomes. Nevertheless, active chromatin regions in the X chromosome interact with each other more frequently than in autosomes. Moreover, despite the absence of dosage compensation complex in the male germline, randomly inserted SpG-specific reporter is expressed higher in the X chromosome than in autosomes, thus evidencing that non-canonical dosage compensation operates in SpG.


Asunto(s)
Cromatina , Drosophila , Animales , Diferenciación Celular/genética , Cromatina/genética , Compensación de Dosificación (Genética) , Drosophila/genética , Células Germinativas , Masculino
6.
Cells ; 9(3)2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-32111103

RESUMEN

DDX3 subfamily DEAD-box RNA helicases are essential developmental regulators of RNA metabolism in eukaryotes. belle, the single DDX3 ortholog in Drosophila, is required for fly viability, fertility, and germline stem cell maintenance. Belle is involved both in translational activation and repression of target mRNAs in different tissues; however, direct targets of Belle in the testes are essentially unknown. Here we showed that belle RNAi knockdown in testis cyst cells caused a disruption of adhesion between germ and cyst cells and generation of tumor-like clusters of stem-like germ cells. Ectopic expression of ß-integrin in cyst cells rescued early stages of spermatogenesis in belle knockdown testes, indicating that integrin adhesion complexes are required for the interaction between somatic and germ cells in a cyst. To address Belle functions in spermatogenesis in detail we performed cross-linking immunoprecipitation and sequencing (CLIP-seq) analysis and identified multiple mRNAs that interacted with Belle in the testes. The set of Belle targets includes transcripts of proteins that are essential for preventing the tumor-like clusters of germ cells and for sustaining spermatogenesis. By our hypothesis, failures in the translation of a number of mRNA targets additively contribute to developmental defects observed in the testes with belle knockdowns both in cyst cells and in the germline.


Asunto(s)
Carcinogénesis/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Células Germinativas/metabolismo , ARN Helicasas/metabolismo , Animales , Animales Modificados Genéticamente , Carcinogénesis/patología , Diferenciación Celular , Proliferación Celular , Drosophila melanogaster/citología , Cadenas beta de Integrinas/metabolismo , Masculino , Modelos Biológicos , Fenotipo , Biosíntesis de Proteínas , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Espermatogénesis , Testículo/metabolismo , Testículo/ultraestructura , Transcriptoma/genética , Transgenes
7.
PLoS One ; 13(8): e0201787, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30157274

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/metabolismo , Desarrollo Embrionario , Productos del Gen gag/metabolismo , Oogénesis , Retroelementos , Animales , Animales Modificados Genéticamente , Transporte Biológico , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Drosophila , Femenino , Regulación del Desarrollo de la Expresión Génica , Ovario/citología , Ovario/metabolismo , Óvulo/citología , Óvulo/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ribonucleoproteínas/metabolismo , Telómero/metabolismo
8.
Nucleic Acids Res ; 45(13): 7666-7680, 2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28472469

RESUMEN

Piwi in a complex with Piwi-interacting RNAs (piRNAs) triggers transcriptional silencing of transposable elements (TEs) in Drosophila ovaries, thus ensuring genome stability. To do this, Piwi must scan the nascent transcripts of genes and TEs for complementarity to piRNAs. The mechanism of this scanning is currently unknown. Here we report the DamID-seq mapping of multiple Piwi-interacting chromosomal domains in somatic cells of Drosophila ovaries. These domains significantly overlap with genomic regions tethered to Nuclear Pore Complexes (NPCs). Accordingly, Piwi was coimmunoprecipitated with the component of NPCs Elys and with the Xmas-2 subunit of RNA transcription and export complex, known to interact with NPCs. However, only a small Piwi fraction has transient access to DNA at nuclear pores. Importantly, although 36% of the protein-coding genes overlap with Piwi-interacting domains and RNA-immunoprecipitation results demonstrate promiscuous Piwi binding to numerous genic and TE nuclear transcripts, according to available data Piwi does not silence these genes, likely due to the absence of perfect base-pairing between piRNAs and their transcripts.


Asunto(s)
Proteínas Argonautas/metabolismo , Cromatina/metabolismo , Proteínas de Drosophila/metabolismo , Poro Nuclear/metabolismo , Ovario/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Argonautas/química , Proteínas Argonautas/genética , Cromatina/genética , Elementos Transponibles de ADN , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Silenciador del Gen , Genoma de los Insectos , Inestabilidad Genómica , Modelos Biológicos , Poro Nuclear/genética , Ovario/citología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Biosci Trends ; 11(1): 46-53, 2017 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-28190795

RESUMEN

Human DDX3 paralogs are housed on the X chromosome (DDX3X) as well as in the non- recombining region Yq11 of the Y-chromosome (DDX3Y or DBY). A gene encoding RNA helicase DDX3Y is located in the AZoospermia Factor a (AZFa) region of the Y-chromosome and expressed only in male germ cells. Deletions encompassing the DDX3Y gene lead to azoospermia and cause Sertoli Cell-Only Syndrome (SCOS) in humans. SCOS is characterized by a complete germ cell lack with preservation of somatic Sertoli cells. This review summarizes current advances in the study of DDX3Y functions in maintenance and development of early male germ cells. Data obtained from a mouse xenotransplantation model reveals that DDX3Y expression is enough to drive germ cell differentiation of AZFa-deleted human induced pluripotent stem cells (iPSCs) and for activation of the specific set of germline developmental genes. Results achieved using the testes of Drosophila demonstrate that DDX3Y homolog Belle is required cell-autonomously for mitotic progression and survival of germline stem cells and spermatogonia as the upstream regulator of mitotic cyclin expression.


Asunto(s)
ARN Helicasas/metabolismo , Espermatozoides/citología , Espermatozoides/metabolismo , Animales , Humanos , Masculino , Modelos Biológicos , Transcripción Genética
10.
Biochim Biophys Acta ; 1863(6 Pt A): 1093-105, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26876306

RESUMEN

The present study showed that RNA helicase Belle (DDX3) was required intrinsically for mitotic progression and survival of germline stem cells (GSCs) and spermatogonial cells in the Drosophila melanogaster testes. We found that deficiency of Belle in the male germline resulted in a strong germ cell loss phenotype. Early germ cells are lost through cell death, whereas somatic hub and cyst cell populations are maintained. The observed phenotype is related to that of the human Sertoli Cell-Only Syndrome caused by the loss of DBY (DDX3) expression in the human testes and results in a complete lack of germ cells with preservation of somatic Sertoli cells. We found the hallmarks of mitotic G2 delay in early germ cells of the larval testes of bel mutants. Both mitotic cyclins, A and B, are markedly reduced in the gonads of bel mutants. Transcription levels of cycB and cycA decrease significantly in the testes of hypomorph bel mutants. Overexpression of Cyclin B in the germline partially rescues germ cell survival, mitotic progression and fertility in the bel-RNAi knockdown testes. Taken together, these results suggest that a role of Belle in GSC maintenance and regulation of early germ cell divisions is associated with the expression control of mitotic cyclins.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Germinativas/metabolismo , ARN Helicasas/metabolismo , Células Madre/metabolismo , Animales , Animales Modificados Genéticamente , Apoptosis/genética , Western Blotting , División Celular/genética , Ciclina A/genética , Ciclina A/metabolismo , Ciclina B/genética , Ciclina B/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Femenino , Masculino , Microscopía Confocal , Mutación , ARN Helicasas/genética , Interferencia de ARN , Testículo/citología , Testículo/metabolismo
11.
Gene ; 499(1): 143-53, 2012 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-22425977

RESUMEN

Testis-specific tandemly repeated Stellate genes are part of the Ste-Su(Ste) genetic system required for male fertility in Drosophila melanogaster. Stellate genes encode a functional homolog of the ß-subunit of protein kinase CK2. Derepression of Stellate results in their over-expression, meiotic disturbances and male sterility. Stellate genes are represented by clustered copies in the X chromosome and carry promoters shared with another X-chromosome cluster, ßNACtes genes, encoding putative ß-subunits of the nascent polypeptide-associated complex. Using Electrophoretic Mobility Shift Assay, we revealed in the Stellate promoter three cis-acting elements, E-boxes, the loss of which greatly diminished the reporter gene expression in Drosophila testes. We identified that these E-boxes were recognized by helix-loop-helix protein, dUSF (Drosophila ortholog of mammalian USF) in testis nuclear extract. All three E-boxes were preserved in the promoters of both euchromatic and heterochromatic Stellate clusters. Two analogous E-boxes were detected in the promoters of 5'-copies of the duplicated ßNACtes gene pairs, whereas the 3'-copies lacked these sites but possessed a new binding site for a testis protein distinct from dUSF. Here we characterized a new type of testis-specific core promoter and identified dUSF as its interacting transcription factor.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regiones Promotoras Genéticas/fisiología , Proteínas Quinasas/genética , Testículo/metabolismo , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Masculino , Modelos Biológicos , Datos de Secuencia Molecular , Familia de Multigenes/genética , Especificidad de Órganos/genética , Regiones Promotoras Genéticas/genética , Proteínas Quinasas/metabolismo , Factores Estimuladores hacia 5'/metabolismo
12.
Mol Biol Cell ; 22(18): 3410-9, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21775629

RESUMEN

Proteins of the PIWI subfamily Aub and AGO3 associated with the germline-specific perinuclear granules (nuage) are involved in the silencing of retrotransposons and other selfish repetitive elements in the Drosophila genome. PIWI proteins and their 25- to 30-nt PIWI-interacting RNA (piRNAs) are considered as key participants of the piRNA pathway. Using immunostaining, we found a large, nuage-associated organelle in the testes, the piNG-body (piRNA nuage giant body), which was significantly more massive than an ordinary nuage granule. This body contains known ovarian nuage proteins, including Vasa, Aub, AGO3, Tud, Spn-E, Bel, Squ, and Cuff, as well as AGO1, the key component of the microRNA pathway. piNG-bodies emerge at the primary spermatocyte stage of spermatogenesis during the period of active transcription. Aub, Vasa, and Tud are located at the periphery of the piNG-body, whereas AGO3 is found in its core. Mutational analysis revealed that Vasa, Aub, and AGO3 were crucial for both the maintenance of the piNG-body structure and the silencing of selfish Stellate repeats. The piNG-body destruction caused by csul mutations that abolish specific posttranslational symmetrical arginine methylation of PIWI proteins is accompanied by strong derepression of Stellate genes known to be silenced via the piRNA pathway.


Asunto(s)
Drosophila melanogaster/genética , Células Germinativas/metabolismo , Orgánulos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Sustitución de Aminoácidos , Animales , Arginina/metabolismo , Proteínas Argonautas/metabolismo , ARN Helicasas DEAD-box/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Genes de Insecto , Masculino , Profase Meiótica I , Microscopía Fluorescente , Mutagénesis Sitio-Dirigida , Tamaño de los Orgánulos , Factores de Iniciación de Péptidos/metabolismo , Proteína Metiltransferasas/genética , Proteína Metiltransferasas/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteína-Arginina N-Metiltransferasas , ARN Interferente Pequeño/genética , Testículo/citología , Testículo/metabolismo
13.
J Mol Biol ; 389(5): 895-906, 2009 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-19422836

RESUMEN

SUMMARY: The X-chromosome-linked clusters of the tandemly repeated testis-specific Stellate genes of Drosophila melanogaster, encoding proteins homologous to the regulatory beta-subunit of the protein kinase casein kinase 2 (CK2), are repressed in wild-type males. Derepression of Stellate genes in the absence of the Y chromosome or Y-linked crystal locus (crystal line) causes accumulation of abundant protein crystals in testes and different meiotic abnormalities, which lead to partial or complete male sterility. To understand the cause of abnormalities in chromosome behavior owing to Stellate overexpression, we studied subcellular localization of Stellate proteins by biochemical fractionation and immunostaining of whole testes. We showed that, apart from the known accumulation of Stellate in crystalline form, soluble Stellate was located exclusively in the nucleoplasm, whereas Stellate crystals were located mainly in the cytoplasm. Coimmunoprecipitation experiments revealed that the alpha-subunit of the protein kinase CK2 (CK2alpha) was associated with soluble Stellate. Interaction between soluble Stellate and CK2alpha in the nucleus could lead to modulations in the phosphorylation of nuclear targets of CK2 and abnormalities in the meiotic segregation of chromosomes. We also observed that Stellate underwent lysine methylation and mimicked trimethyl-H3K9 epigenetic modification of histone H3 tail.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Proteínas de Insectos/metabolismo , Mapeo de Interacción de Proteínas , Proteínas Quinasas/metabolismo , Espermatocitos/metabolismo , Animales , Dominio Catalítico , Fraccionamiento Celular , Núcleo Celular/química , Inmunoprecipitación , Lisina/metabolismo , Masculino , Metilación , Microscopía Fluorescente , Unión Proteica
14.
Genetica ; 117(2-3): 239-45, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12723703

RESUMEN

The mechanism of silencing of testis expressed X-linked Stellate repeats by homologous Y-linked Suppressor of Stellate [Su(Ste)] repeats localized in the crystal locus was studied. The double stranded RNA as a product of symmetrical transcription of Su(Ste) repeat and small interference Su(Ste) siRNA were revealed suggesting the mechanism of RNA interference (RNAi) for Stellate silencing. The relief of Stellate silencing as a result of impaired complementarity between the sequences of putative target Stellate transcripts and Su(Ste) repeats was shown. The role of RNAi mechanism in the silencing of heterochromatic retrotransposon GATE inserted in Stellate cluster was revealed. The studies of cis-effects of Stellate tandem repeats causing variegated expression of juxtaposed reporter genes were extended and the lacZ variegation in imaginal disc was shown. The exceptional case of a non-variegated expression of mini-white gene juxtaposed to Stellate repeats in a construct inserted into the 39C region was shown to be accompanied by trans-activation in homozygous state. Trans-activation effect was retained after transposition of this construct into heterochromatic environment in spite of strong variegation of a mini-white gene.


Asunto(s)
Drosophila melanogaster/genética , Silenciador del Gen , Proteínas de Insectos/genética , Proteínas Quinasas/genética , Activación Transcripcional/genética , Transportadoras de Casetes de Unión a ATP/genética , Animales , Proteínas de Drosophila/genética , Proteínas del Ojo/genética , Heterocromatina , Proteínas Represoras/genética , Secuencias Repetidas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...