Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 112022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35311642

RESUMO

We recently identified a missense mutation in Nucleoporin107 (Nup107; D447N) underlying XX-ovarian-dysgenesis, a rare disorder characterized by underdeveloped and dysfunctional ovaries. Modeling of the human mutation in Drosophila or specific knockdown of Nup107 in the gonadal soma resulted in ovarian-dysgenesis-like phenotypes. Transcriptomic analysis identified the somatic sex-determination gene doublesex (dsx) as a target of Nup107. Establishing Dsx as a primary relevant target of Nup107, either loss or gain of Dsx in the gonadal soma is sufficient to mimic or rescue the phenotypes induced by Nup107 loss. Importantly, the aberrant phenotypes induced by compromising either Nup107 or dsx are reminiscent of bone morphogenetic protein (BMP signaling hyperactivation). Remarkably, in this context, the metalloprotease AdamTS-A, a transcriptional target of both Dsx and Nup107, is necessary for the calibration of BMP signaling. As modulation of BMP signaling is a conserved critical determinant of soma-germline interaction, the sex- and tissue-specific deployment of Dsx-F by Nup107 seems crucial for the maintenance of the homeostatic balance between the germ cells and somatic gonadal cells.


Assuntos
Aquaporinas , Proteínas de Drosophila , Animais , Proteínas de Ligação a DNA/metabolismo , Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Diferenciação Sexual/genética
2.
Front Mol Neurosci ; 14: 693967, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34290587

RESUMO

Dehydrodolichyl diphosphate synthase (DHDDS) is a ubiquitously expressed enzyme that catalyzes cis-prenyl chain elongation to produce the poly-prenyl backbone of dolichol. It appears in all tissues including the nervous system and it is a highly conserved enzyme that can be found in all animal species. Individuals who have biallelic missense mutations in the DHDDS gene are presented with non-syndromic retinitis pigmentosa with unknown underlying mechanism. We have used the Drosophila model to compromise DHDDS ortholog gene (CG10778) in order to look for cellular and molecular mechanisms that, when defective, might be responsible for this retinal disease. The Gal4/UAS system was used to suppress the expression of CG10778 via RNAi-mediated-knockdown in various tissues. The resulting phenotypes were assessed using q-RT-PCR, transmission-electron-microscopy (TEM), electroretinogram, antibody staining and Western blot analysis. Targeted knockdown of CG10778-mRNA in the early embryo using the actin promoter or in the developing wings using the nub promoter resulted in lethality, or wings loss, respectively. Targeted expression of CG10778-RNAi using the glass multiple reporter (GMR)-Gal4 driver (GMR-DHDDS-RNAi) in the larva eye disc and pupal retina resulted in a complex phenotype: (a) TEM retinal sections revealed a unique pattern of retinal-degeneration, where photoreceptors R2 and R5 exhibited a nearly normal structure of their signaling-compartment (rhabdomere), but only at the region of the nucleus, while all other photoreceptors showed retinal degeneration at all regions. (b) Western blot analysis revealed a drastic reduction in rhodopsin levels in GMR-DHDDS-RNAi-flies and TEM sections showed an abnormal accumulation of endoplasmic reticulum (ER). To conclude, compromising DHDDS in the developing retina, while allowing formation of the retina, resulted in a unique pattern of retinal degeneration, characterized by a dramatic reduction in rhodopsin protein level and an abnormal accumulation of ER membranes in the photoreceptors cells, thus indicating that DHDDS is essential for normal retinal formation.

3.
Biol Proced Online ; 22: 14, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32684853

RESUMO

BACKGROUND: The generation of point mutations is a major tool for evaluating the roles of specific nucleotides or amino acids within the regulatory or functional landscape. However, examination of these mutations in vivo requires the generation of animals carrying only the relevant point mutations at the endogenous genomic loci, which is technically challenging. The CRISPR-Cas9 based genome editing greatly facilitates the generation of such genetically modified animals; however, most of the described methods use double-strand DNA (dsDNA) as the donor template. The dsDNA plasmids frequently undergo undesired integration events into the targeted genomic locus. The use of a single-strand oligodeoxynucleotide (ssODN) as the donor template prevents this complication and is therefore the preferred choice for introducing point mutations, as well as short sequences such as protein tags. RESULTS: We successfully applied the CRISPR-based white co-conversion strategy with a ssODN template, instead of the originally described dsDNA plasmid, to create genetically modified Drosophila melanogaster strains. We used the technique to easily introduce point mutations in two distinct chromosomes. Using the generated flies, we were able to demonstrate the in vivo importance of the respective mutations. For the Nucleoporin107 (Nup107) gene, the 1090G > A mutation was confirmed to affect ovarian development, while for the tinman (tin) gene, the regulatory role of the downstream core promoter element (DPE) was demonstrated within the developing Drosophila melanogaster embryo. CONCLUSIONS: The described approach has facilitated the successful generation of point mutations in two different chromosomes, by two different labs. Distinct phenotypes associated with the newly-generated genotype were identified, thus exemplifying the importance of investigating the in vivo role of specific nucleotides. In addition, detailed guidelines, recommendations and crossing schemes are provided in order to support the generation of additional genetically modified animals by the scientific community.

4.
Development ; 146(10)2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31092503

RESUMO

During embryonic gonad coalescence, primordial germ cells (PGCs) follow a carefully choreographed migratory route circumscribed by guidance signals towards somatic gonadal precursor cells (SGPs). In Drosophila melanogaster, SGP-derived Hedgehog (Hh), which serves as a guidance cue for the PGCs, is potentiated by mesodermally restricted HMGCoA-reductase (Hmgcr) and the ABC transporter Multi-drug-resistant-49 (Mdr49). Given the importance of cholesterol modification in the processing and long-distance transmission of the Hh ligand, we have analyzed the involvement of the Niemann-Pick disease type C-1a (NPC1a) protein, a cholesterol transporter, in germ cell migration and Hedgehog signaling. We show that mesoderm-specific inactivation of Npc1a results in germ cell migration defects. Similar to Mdr49, PGC migration defects in the Npc1a embryos are ameliorated by a cholesterol-rich diet. Consistently, reduction in Npc1a weakens the ability of ectopic HMG Coenzyme A reductase (Hmgcr) to induce germ cell migration defects. Moreover, compromising Npc1a levels influences Hh signaling adversely during wing development, a process that relies upon long-range Hh signaling. Last, doubly heterozygous embryos (Mdr49/Npc1a) display enhanced germ cell migration defects when compared with single mutants (Npc1a/+ or Mdr49/+), supporting cooperative interaction between the two.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas de Membrana/metabolismo , Animais , Movimento Celular/genética , Movimento Celular/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células Germinativas/metabolismo , Heterozigoto , Proteínas de Membrana/genética , Neurônios/metabolismo , Proteína C1 de Niemann-Pick , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
5.
N Engl J Med ; 379(11): 1042-1049, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30207912

RESUMO

The causes of ovarian dysgenesis remain incompletely understood. Two sisters with XX ovarian dysgenesis carried compound heterozygous truncating mutations in the BRCA2 gene that led to reduced BRCA2 protein levels and an impaired response to DNA damage, which resulted in chromosomal breakage and the failure of RAD51 to be recruited to double-stranded DNA breaks. The sisters also had microcephaly, and one sister was in long-term remission from leukemia, which had been diagnosed when she was 5 years old. Drosophila mutants that were null for an orthologue of BRCA2 were sterile, and gonadal dysgenesis was present in both sexes. These results revealed a new role for BRCA2 and highlight the importance to ovarian development of genes that are critical for recombination during meiosis. (Funded by the Israel Science Foundation and others.).


Assuntos
Proteína BRCA2/deficiência , Quebra Cromossômica , Reparo do DNA , Genes BRCA2 , Disgenesia Gonadal/genética , Ovário/crescimento & desenvolvimento , Adolescente , Animais , Proteína BRCA2/fisiologia , Quebra Cromossômica/efeitos dos fármacos , Análise Mutacional de DNA , Drosophila melanogaster , Feminino , Humanos , Hipogonadismo/genética , Masculino , Microcefalia/genética , Mitomicina/farmacologia , Modelos Animais , Ovário/fisiologia , Linhagem , Irmãos , Adulto Jovem
6.
Fly (Austin) ; 11(3): 200-207, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28300473

RESUMO

In Drosophila melanogaster the progenitors of the germ-line stem cells, the primordial germ cells (PGCs) are formed on the outside surface of the early embryo, while the somatic gonadal precursor cells (SGPs) are specified during mid-embryogenesis. To form the primitive embryonic gonad, the PGCs travel from outside of the embryo, across the mid-gut and then migrate through the mesoderm to the SGPs. The migratory path of PGCs is dictated by a series of attractive and repulsive cues. Studies in our laboratory have shown that one of the key chemoattractants is the Hedgehog (Hh) ligand. Although, Hh is expressed in other cell types, the long-distance transmission of this ligand is specifically potentiated in the SGPs by the hmgcr isoprenoid biosynthetic pathway. The distant transmission of the Hh ligand is gated by restricting expression of hmgcr to the SGPs. This is particularly relevant in light of the recent findings that an ABC transporter, mdr49 also acts in a mesoderm specific manner to release the germ cell attractant. Our studies have demonstrated that mdr49 functions in hh signaling likely via its role in the transport of cholesterol. Given the importance of cholesterol in the processing and long distance transmission of the Hh ligand, this observation has opened up an exciting avenue concerning the possible role of components of the sterol transport machinery in PGC migration.


Assuntos
Movimento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Células Germinativas/fisiologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Animais , Colesterol/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Embrião não Mamífero/citologia , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Proteínas Hedgehog/metabolismo , Transdução de Sinais
7.
Development ; 143(12): 2111-20, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27122170

RESUMO

Coalescence of the embryonic gonad in Drosophila melanogaster requires directed migration of primordial germ cells (PGCs) towards somatic gonadal precursor cells (SGPs). It was recently proposed that the ATP-binding cassette (ABC) transporter Mdr49 functions in the embryonic mesoderm to facilitate the transmission of the PGC attractant from the SGPs; however, the precise molecular identity of the Mdr49-dependent guidance signal remained elusive. Employing the loss- and gain-of-function strategies, we show that Mdr49 is a component of the Hedgehog (hh) pathway and it potentiates the signaling activity. This function is direct because in Mdr49 mutant embryos the Hh ligand is inappropriately sequestered in the hh-expressing cells. Our data also suggest that the role of Mdr49 is to provide cholesterol for the correct processing of the Hh precursor protein. Supporting this conclusion, PGC migration defects in Mdr49 embryos are substantially ameliorated by a cholesterol-rich diet.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Movimento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Células Germinativas/citologia , Células Germinativas/metabolismo , Proteínas Hedgehog/metabolismo , Alelos , Animais , Colesterol/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Células Epidérmicas , Epiderme/embriologia , Epistasia Genética , Comportamento Alimentar , Duplicação Gênica , Regulação da Expressão Gênica no Desenvolvimento , Homozigoto , Ligantes , Mutação/genética , Transdução de Sinais , Asas de Animais/anormalidades , Asas de Animais/metabolismo , Zigoto/metabolismo
8.
J Clin Invest ; 125(11): 4295-304, 2015 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-26485283

RESUMO

Ovarian development and maintenance are poorly understood; however, diseases that affect these processes can offer insights into the underlying mechanisms. XX female gonadal dysgenesis (XX-GD) is a rare, genetically heterogeneous disorder that is characterized by underdeveloped, dysfunctional ovaries, with subsequent lack of spontaneous pubertal development, primary amenorrhea, uterine hypoplasia, and hypergonadotropic hypogonadism. Here, we report an extended consanguineous family of Palestinian origin, in which 4 females exhibited XX-GD. Using homozygosity mapping and whole-exome sequencing, we identified a recessive missense mutation in nucleoporin-107 (NUP107, c.1339G>A, p.D447N). This mutation segregated with the XX-GD phenotype and was not present in available databases or in 150 healthy ethnically matched controls. NUP107 is a component of the nuclear pore complex, and the NUP107-associated protein SEH1 is required for oogenesis in Drosophila. In Drosophila, Nup107 knockdown in somatic gonadal cells resulted in female sterility, whereas males were fully fertile. Transgenic rescue of Drosophila females bearing the Nup107D364N mutation, which corresponds to the human NUP107 (p.D447N), resulted in almost complete sterility, with a marked reduction in progeny, morphologically aberrant eggshells, and disintegrating egg chambers, indicating defective oogenesis. These results indicate a pivotal role for NUP107 in ovarian development and suggest that nucleoporin defects may play a role in milder and more common conditions such as premature ovarian failure.


Assuntos
Aquaporinas/fisiologia , Proteínas de Drosophila/fisiologia , Disgenesia Gonadal 46 XX/genética , Mutação de Sentido Incorreto , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Ovário/patologia , Adolescente , Adulto , Animais , Animais Geneticamente Modificados , Aquaporinas/deficiência , Aquaporinas/genética , Consanguinidade , Modelos Animais de Doenças , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Humanos , Infertilidade Feminina/genética , Masculino , Camundongos , Modelos Moleculares , Morfogênese , Complexos Multiproteicos , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Oogênese/genética , Óvulo/patologia , Linhagem , Conformação Proteica
9.
J Cell Sci ; 125(Pt 23): 5811-8, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22956540

RESUMO

BMP-dependent patterning in the Drosophila melanogaster wing imaginal disc serves as a paradigm to understand how morphogens specify cell fates. The observed profile of the transcriptional response to the graded signal of BMP relies upon two counter-active gradients of pMad and Brinker (Brk). This patterning model is inadequate to explain the expression of target genes, like vestigial and spalt, in lateral regions of the wing disc where BMP signals decline and Brk levels peak. Here, we show that in contrast to the reciprocal repressor gradient mechanism, where Brk represses BMP targets in medial regions, target expression in lateral regions is downregulated by BMP signalling and activated by Brk. Brk induces lateral expression indirectly, apparently through repression of a negative regulator. Our findings provide a model explaining how the expression of an established BMP target is differentially and inversely regulated along the anterior-posterior axis of the wing disc.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Drosophila/metabolismo , Animais , Padronização Corporal/genética , Padronização Corporal/fisiologia , Proteínas Morfogenéticas Ósseas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Asas de Animais/embriologia , Asas de Animais/metabolismo
10.
J Biol Chem ; 286(17): 15556-64, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21385866

RESUMO

Apoptosis operates to eliminate damaged or potentially dangerous cells. This loss is often compensated by extra proliferation of neighboring cells. Studies in Drosophila imaginal discs suggest that the signal for the additional growth emanates from the dying cells. In particular, it was suggested that the initiator caspase Dronc mediates compensatory proliferation (CP) through Dp53 in wing discs. However, the exact mechanism that governs this CP remained poorly understood. We have previously shown that elimination of misspecified cells due to reduced Dpp signaling is achieved by the interaction of the co-repressor NAB with the transcriptional repressor Brk, which in turn induces Jun N-terminal kinase-dependent apoptosis. Here, we performed a systematic in vivo loss- and gain-of-function analysis to study NAB-induced death and CP. Our findings indicate that the NAB primary signal activates JNK, which in turn transmits two independent signals. One triggers apoptosis through the pro-apoptotic proteins Reaper and Hid, which in turn promote activation of caspases by the apoptosome components Ark and Dronc. The other signal induces CP in a manner that is independent of the death signal, Dronc, or Dp53. Once induced, the apoptotic pathway further activates a CP response. Our data suggest that JNK is the candidate factor that differentiates between apoptosis that involves CP and apoptosis that does not.


Assuntos
Apoptose , Proliferação de Células , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Animais , Apoptossomas/metabolismo , Neuropeptídeos/metabolismo
11.
Fly (Austin) ; 5(1): 25-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21057219

RESUMO

During metazoan development, a small number of signaling pathways are iteratively used to orchestrate diverse processes such as cell division, cell fate specification and survival. Temporal and spatial regulation of these pathways underlies the final cellular makeup, size and shape of organs. In Drosophila melanogaster, the master switch gene Sex-lethal (Sxl) orchestrates all aspects of female development and behavior by modulating gene expression. Many of the sex-specific differences in gene expression and morphology are controlled through a gene activity cascade that involves Sxl→tra→dsx-fru. However, various aspects of somatic sexual development appear to be independent of this cascade. Consistent with this idea, Sxl protein, on its own, was recently implicated in the regulation of both Hh and Notch signaling to shape some of the sexually dimorphic traits. Paradoxically, however, Sxl activity is essential in every female cell to prevent the activation of the male-specific dosage compensation system and thus to ensure the proper level of X-linked gene expression. This raises a key question as to how the sex-specific effects of Sxl on major signaling pathways are prevented in monomorphic tissues during female development. We have elucidated a novel mechanism where Hrp48, an abundant essential hnRNP functions to restrict Sxl expression in monomorphic tissues and thus allow for proper development. Our findings bring into focus the critical role played by general homeostatic factors in specification of diverse cell fates and morphogenesis.


Assuntos
Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Animais , Mecanismo Genético de Compensação de Dose , Proteínas de Drosophila/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Homeostase , Masculino , Proteínas de Ligação a RNA/metabolismo , Diferenciação Sexual
12.
Neuro Oncol ; 12(5): 422-33, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20406893

RESUMO

Gliomas express many genes that play a role in neural precursor cells (NPCs), but no direct comparison between glioma and stem cell (SC) gene expression profiles has been performed. To investigate the similarities and differences between gliomas and SCs, we compared the microRNA (miRNA) expression signatures of glial tumors, embryonic SCs (ESCs), NPCs, and normal adult brains from both human and mouse tissues. We demonstrated that both human gliomas (regardless of their grade) and methylcholanthrene-induced mouse glioma shared an miRNA expression profile that is reminiscent of NPCs. About half of the miRNAs expressed in the shared profile clustered in seven genomic regions susceptible to genetic/epigenetic alterations in various cancers. These clusters comprised the miR17 family, mir183-182, and the SC-specific clusters mir367-302 and mir371-373, which are upregulated in gliomas, ESCs, and NPCs. The bipartite cluster of 7 + 46 miRNAs on chromosome 14q32.31, which might represent the largest tumor suppressor miRNA cluster, was downregulated in the shared expression profile. This study provides the first evidence for association between these clusters and gliomas. Despite the broad similarity in the miRNA expression profiles, 15 miRNAs showed disparate expression between SC and gliomas. Ten miRNAs belong to the 2 SC-specific clusters and the remaining (mir135b, mir141, mir205, mir200C, and mir301a) have been previously shown to associate with malignancies. Our finding showed that all gliomas displayed NPC-like miRNA signatures, which may have implications for studies of glioma origins. Furthermore, careful study of the 15 miRNAs that differ in expression between SCs and gliomas, particularly those 5 that are not SC-specific, may enhance our understanding of gliomagenesis.


Assuntos
Neoplasias Encefálicas/genética , Perfilação da Expressão Gênica , Glioma/genética , Neurônios/metabolismo , RNA Mensageiro/análise , Células-Tronco/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Perda de Heterozigosidade , Camundongos , Camundongos Endogâmicos C57BL
13.
Proc Natl Acad Sci U S A ; 107(15): 6930-5, 2010 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-20351283

RESUMO

Different signaling pathways are deployed in specific developmental contexts to generate sexually dimorphic traits. Recently, Sex-lethal (Sxl), the female determinant in Drosophila melanogaster, was shown to down-regulate Notch (N) signaling to accomplish sex-specific patterning. Paradoxically, however, both Sxl and N are ubiquitously expressed in all of the female cells. This raises a key question as to how, during monomorphic female development, N signaling escapes the negative impact of Sxl. Here, we uncover a regulatory loop involving Hrp48, an abundant Drosophila hnRNP, Sxl and N. Phenotypic consequences of the partial loss of hrp48 resemble that of N but are more pronounced in females than in males. Likewise, N levels are drastically diminished only in females. Interestingly, monomorphic female tissues including wing, eye and antennal discs display considerable increase in Sxl amounts. Finally, female-specific attenuation of N signaling is rescued upon simultaneous removal of Sxl. Thus, our data demonstrate that in monomorphic contexts, Hrp48 functions as a moderator of Sxl expression to achieve adequate levels of N receptor production and signaling. We propose that it is critical to modulate the activities of the master determinant underling sexual dimorphism, to ensure that it does not function inappropriately in monomorphic tissues and disrupt their development.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Ribonucleoproteínas Nucleares Heterogêneas/fisiologia , Proteínas de Ligação a RNA/metabolismo , Receptores Notch/metabolismo , Alelos , Animais , Cruzamentos Genéticos , Feminino , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Masculino , Mitose , Modelos Biológicos , Fatores Sexuais , Transdução de Sinais , Asas de Animais/embriologia
14.
Development ; 136(7): 1137-45, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19270172

RESUMO

The proper development of tissues requires morphogen activity that dictates the appropriate growth and differentiation of each cell according to its position within a developing field. Elimination of underperforming cells that are less efficient in receiving/transducing the morphogenetic signal is thought to provide a general fail-safe mechanism to avoid developmental misspecification. In the developing Drosophila wing, the morphogen Dpp provides cells with growth and survival cues. Much of the regulation of transcriptional output by Dpp is mediated through repression of the transcriptional repressor Brinker (Brk), and thus through the activation of target genes. Mutant cells impaired for Dpp reception or transduction are lost from the wing epithelium. At the molecular level, reduced Dpp signaling results in Brk upregulation that triggers apoptosis through activation of the JNK pathway. Here we show that the transcriptional co-regulator dNAB is a Dpp target in the developing wing that interacts with Brk to eliminate cells with reduced Dpp signaling through the JNK pathway. We further show that both dNAB and Brk are required for cell elimination induced by differential dMyc expression, a process that depends on reduced Dpp transduction in outcompeted cells. We propose a novel mechanism whereby the morphogen Dpp regulates the responsiveness to its own survival signal by inversely controlling the expression of a repressor, Brk, and its co-repressor, dNAB.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas Repressoras/metabolismo , Animais , Animais Geneticamente Modificados , Apoptose/genética , Apoptose/fisiologia , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/genética , Padronização Corporal/fisiologia , Primers do DNA/genética , Drosophila/genética , Proteínas de Drosophila/genética , Epistasia Genética , Genes de Insetos , Técnicas In Vitro , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Repressoras/genética , Transdução de Sinais , Asas de Animais/crescimento & desenvolvimento , Asas de Animais/metabolismo
15.
Development ; 136(6): 995-1006, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19211674

RESUMO

Cell competition is a short-range cell-cell interaction leading to the proliferation of winner cells at the expense of losers, although either cell type shows normal growth in homotypic environments. Drosophila Myc (dMyc; Dm-FlyBase) is a potent inducer of cell competition in wing epithelia, but its role in the ovary germline stem cell niche is unknown. Here, we show that germline stem cells (GSCs) with relative lower levels of dMyc are replaced by GSCs with higher levels of dMyc. By contrast, dMyc-overexpressing GSCs outcompete wild-type stem cells without affecting total stem cell numbers. We also provide evidence for a naturally occurring cell competition border formed by high dMyc-expressing stem cells and low dMyc-expressing progeny, which may facilitate the concentration of the niche-provided self-renewal factor BMP/Dpp in metabolically active high dMyc stem cells. Genetic manipulations that impose uniform dMyc levels across the germline produce an extended Dpp signaling domain and cause uncoordinated differentiation events. We propose that dMyc-induced competition plays a dual role in regulating optimal stem cell pools and sharp differentiation boundaries, but is potentially harmful in the case of emerging dmyc duplications that facilitate niche occupancy by pre-cancerous stem cells. Moreover, competitive interactions among stem cells may be relevant for the successful application of stem cell therapies in humans.


Assuntos
Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Ovário/citologia , Ovário/metabolismo , Nicho de Células-Tronco/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Mutação/genética , Nicho de Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
17.
Genes Dev ; 16(9): 1055-9, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12000788

RESUMO

Secreted peptide signals control many fundamental processes during animal development. Proper responses to these signals require cognate inducible feedback antagonists. Here we report the identification of a novel Drosophila Wingless (Wg) target gene, wingful (wf), and show that it encodes a potent extracellular feedback inhibitor of Wg. In contrast to the cytoplasmic protein Naked cuticle (Nkd), the only known Wg feedback antagonist, Wf functions during larval stages, when Nkd function is dispensable. We propose that Wf may provide feedback control for the long-range morphogen activities of Wg.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Proteínas Proto-Oncogênicas/metabolismo , Sequência de Aminoácidos , Animais , Drosophila/crescimento & desenvolvimento , Esterases/genética , Esterases/metabolismo , Matriz Extracelular/metabolismo , Retroalimentação Fisiológica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Insetos/metabolismo , Larva , Dados de Sequência Molecular , Mutação , Proteínas Proto-Oncogênicas/genética , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Proteína Wnt1
18.
Int J Dev Biol ; 46(1): 173-6, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11902680

RESUMO

The development of Drosophila imaginal discs serves as a model system to understand how genes determine the shape and size of an organ. The identification of genes involved in this process is an important step towards this goal. Here we describe a P-element based enhancer trap screen for genes expressed in the larval imaginal discs. Our aim was to establish a large collection of enhancer trap lines each showing expression of Gal4 in imaginal discs. To this end, we improved the well established P-element vector pGawB in order to obtain higher in vivo transposition frequencies. In addition we chose an F1-screening approach using UAS-GFP as a reporter gene. This system permits the efficient screening of larval and pupal stages of living animals and the detection of imaginal gene expression patterns through the transparent cuticle. The procedure has been optimized for high-throughput. 2'000 P-element insertions have been established which exhibit expression in imaginal discs.


Assuntos
Drosophila/genética , Elementos Facilitadores Genéticos , Técnicas Genéticas , Proteínas de Saccharomyces cerevisiae , Animais , Proteínas de Ligação a DNA , Proteínas Fúngicas/genética , Vetores Genéticos , Proteínas de Fluorescência Verde , Proteínas Luminescentes/metabolismo , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...