Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
G3 (Bethesda) ; 10(11): 3949-3958, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32972998

ABSTRACT

The R7 and R8 photoreceptor cells of the Drosophila compound eye mediate color vision. Throughout the majority of the eye, these cells occur in two principal types of ommatidia. Approximately 35% of ommatidia are of the pale type and express Rh3 in R7 cells and Rh5 in R8 cells. The remaining 65% are of the yellow type and express Rh4 in R7 cells and Rh6 in R8 cells. The specification of an R8 cell in a pale or yellow ommatidium depends on the fate of the adjacent R7 cell. However, pale and yellow R7 cells are specified by a stochastic process that requires the genes spineless, tango and klumpfuss To identify additional genes involved in this process we performed genetic screens using a collection of 480 P{EP} transposon insertion strains. We identified genes in gain of function and loss of function screens that significantly altered the percentage of Rh3 expressing R7 cells (Rh3%) from wild-type. 36 strains resulted in altered Rh3% in the gain of function screen where the P{EP} insertion strains were crossed to a sevEP-GAL4 driver line. 53 strains resulted in altered Rh3% in the heterozygous loss of function screen. 4 strains showed effects that differed between the two screens, suggesting that the effect found in the gain of function screen was either larger than, or potentially masked by, the P{EP} insertion alone. Analyses of homozygotes validated many of the candidates identified. These results suggest that R7 cell fate specification is sensitive to perturbations in mRNA transcription, splicing and localization, growth inhibition, post-translational protein modification, cleavage and secretion, hedgehog signaling, ubiquitin protease activity, GTPase activation, actin and cytoskeletal regulation, and Ser/Thr kinase activity, among other diverse signaling and cell biological processes.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Cell Differentiation , Drosophila/genetics , Drosophila Proteins/genetics , Hedgehog Proteins , Photoreceptor Cells, Invertebrate
2.
PLoS One ; 15(10): e0240451, 2020.
Article in English | MEDLINE | ID: mdl-33052948

ABSTRACT

Cell differentiation and cell fate determination in sensory systems are essential for stimulus discrimination and coding of environmental stimuli. Color vision is based on the differential color sensitivity of retinal photoreceptors, however the developmental programs that control photoreceptor cell differentiation and specify color sensitivity are poorly understood. In Drosophila melanogaster, there is evidence that the color sensitivity of different photoreceptors in the compound eye is regulated by inductive signals between cells, but the exact nature of these signals and how they are propagated remains unknown. We conducted a genetic screen to identify additional regulators of this process and identified a novel mutation in the hibris gene, which encodes an irre cell recognition module protein (IRM). These immunoglobulin super family cell adhesion molecules include human KIRREL and nephrin (NPHS1). hibris is expressed dynamically in the developing Drosophila melanogaster eye and loss-of-function mutations give rise to a diverse range of mutant phenotypes including disruption of the specification of R8 photoreceptor cell diversity. We demonstrate that hibris is required within the retina, and that hibris over-expression is sufficient to disrupt normal photoreceptor cell patterning. These findings suggest an additional layer of complexity in the signaling process that produces paired expression of opsin genes in adjacent R7 and R8 photoreceptor cells.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Retina/growth & development , Animals , Cell Differentiation , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Imaginal Discs/metabolism , Mutation , Organ Specificity , Photoreceptor Cells, Invertebrate/cytology , Retina/metabolism
3.
Genetics ; 174(4): 1973-82, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17028348

ABSTRACT

Differential gene expression is the major mechanism underlying the development of specific body regions. Here we assessed the role of genes differentially expressed in the Drosophila wing imaginal disc, which gives rise to two distinct adult structures: the body wall and the wing. Reverse genetics was used to test the function of uncharacterized genes first identified in a microarray screen as having high levels of expression in the presumptive wing. Such genes could participate in elaborating the specific morphological characteristics of the wing. The activity of the genes was modulated using misexpression and RNAi-mediated silencing. Misexpression of eight of nine genes tested caused phenotypes. Of 12 genes tested, 10 showed effective silencing with RNAi transgenes, but only 3 of these had resulting phenotypes. The wing phenotypes resulting from RNAi suggest that CG8780 is involved in patterning the veins in the proximal region of the wing blade and that CG17278 and CG30069 are required for adhesion of wing surfaces. Venation and apposition of the wing surfaces are processes specific to wing development providing a correlation between the expression and function of these genes. The results show that a combination of expression profiling and tissue-specific gene silencing has the potential to identify new genes involved in wing development and hence to contribute to our understanding of this process. However, there are both technical and biological limitations to this approach, including the efficacy of RNAi and the role that gene redundancy may play in masking phenotypes.


Subject(s)
Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Wings, Animal/physiology , Amino Acid Sequence , Animals , Cells, Cultured , Crosses, Genetic , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , In Situ Hybridization , Male , Molecular Sequence Data , Phenotype , Pupa/cytology , Pupa/metabolism , RNA, Small Interfering/pharmacology , Sequence Homology, Amino Acid
4.
Genetics ; 167(2): 687-98, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15238521

ABSTRACT

Vein (Vn), a ligand for the Drosophila epidermal growth factor receptor (Egfr), has a complex structure including a PEST, Ig, and EGF domain. We analyzed the structure-function relationships of Vn by assaying deletion mutants. The results show that each conserved domain influences Vn activity. A PEST deletion increases Vn potency and genetic evidence suggests that Vn is regulated by proteasomal degradation. The Ig deletion causes toxic effects not seen following expression of native Vn, but the Ig domain is not required for Vn localization or for the activation of Egfr signaling in wing vein patterning. Remarkably, when the EGF domain is deleted, Vn functions as a dominant negative ligand, implying that Vn normally physically interacts with another factor to promote its activity. We identified additional highly conserved sequences and found several regions that affect Vn potency and one that may mediate the effect of dominant negative Vn molecules. Together the results show that the activity of Vn is controlled both positively and negatively, demonstrating the existence of additional levels at which Egfr signaling can be regulated.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Epidermal Growth Factor/genetics , ErbB Receptors/genetics , Amino Acid Sequence , Animals , Conserved Sequence , Ligands , Molecular Sequence Data , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Veins
5.
Genetics ; 201(4): 1319-28, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26500255

ABSTRACT

Transfection of transgenes into Drosophila cultured cells is a standard approach for studying gene function. However, the number of transgenes present in the cell following transient transfection or stable random integration varies, and the resulting differences in expression level affect interpretation. Here we developed a system for Drosophila cell lines that allows selection of cells with a single-copy transgene inserted at a specific genomic site using recombination-mediated cassette exchange (RMCE). We used the φC31 integrase and its target sites attP and attB for RMCE. Cell lines with an attP-flanked genomic cassette were transfected with donor plasmids containing a transgene of interest (UAS-x), a dihydrofolate reductase (UAS-DHFR) gene flanked by attB sequences, and a thymidine kinase (UAS-TK) gene in the plasmid backbone outside the attB sequences. In cells undergoing RMCE, UAS-x and UAS-DHFR were exchanged for the attP-flanked genomic cassette, and UAS-TK was excluded. These cells were selected using methotrexate, which requires DHFR expression, and ganciclovir, which causes death in cells expressing TK. Pure populations of cells with one copy of a stably integrated transgene were efficiently selected by cloning or mass culture in ∼6 weeks. Our results show that RMCE avoids the problems associated with current methods, where transgene number is not controlled, and facilitates the rapid generation of Drosophila cell lines in which expression from a single transgene can be studied.


Subject(s)
Gene Targeting/methods , Integrases/metabolism , Transfection/methods , Animals , Cell Line , Drosophila melanogaster/genetics , Female , Genes, Insect , Male , Recombination, Genetic , Tissue Culture Techniques , Transgenes
6.
Genetics ; 191(4): 1213-26, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22595244

ABSTRACT

The highly conserved epidermal growth factor receptor (Egfr) pathway is required in all animals for normal development and homeostasis; consequently, aberrant Egfr signaling is implicated in a number of diseases. Genetic analysis of Drosophila melanogaster Egfr has contributed significantly to understanding this conserved pathway and led to the discovery of new components and targets. Here we used microarray analysis of third instar wing discs, in which Egfr signaling was perturbed, to identify new Egfr-responsive genes. Upregulated transcripts included five known targets, suggesting the approach was valid. We investigated the function of 29 previously uncharacterized genes, which had pronounced responses. The Egfr pathway is important for wing-vein patterning and using reverse genetic analysis we identified five genes that showed venation defects. Three of these genes are expressed in vein primordia and all showed transcriptional changes in response to altered Egfr activity consistent with being targets of the pathway. Genetic interactions with Egfr further linked two of the genes, Sulfated (Sulf1), an endosulfatase gene, and CG4096, an A Disintegrin And Metalloproteinase with ThromboSpondin motifs (ADAMTS) gene, to the pathway. Sulf1 showed a strong genetic interaction with the neuregulin-like ligand vein (vn) and may influence binding of Vn to heparan-sulfated proteoglycans (HSPGs). How Drosophila Egfr activity is modulated by CG4096 is unknown, but interestingly vertebrate EGF ligands are regulated by a related ADAMTS protein. We suggest Sulf1 and CG4096 are negative feedback regulators of Egfr signaling that function in the extracellular space to influence ligand activity.


Subject(s)
Drosophila/metabolism , ErbB Receptors/metabolism , Feedback, Physiological , Signal Transduction , Animals , Body Patterning/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Epistasis, Genetic , ErbB Receptors/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Genotype , Ligands , Phenotype , Protein Binding , RNA Interference , Sulfatases/genetics , Sulfatases/metabolism , Transcriptome , Veins/metabolism , Wings, Animal/metabolism
7.
Fly (Austin) ; 2(6): 306-9, 2008.
Article in English | MEDLINE | ID: mdl-19077546

ABSTRACT

The in vivo analysis of Drosophila using genetics, with almost a hundred year history, has produced an immense body of knowledge about biology. In vitro analysis, while arguably the poor cousin to its in vivo relative, has a utility--in biochemical analyses and in cell-based screening, for example, with RNAi. A major block to the development of in vitro analysis has been the lack of an efficient genetic method to derive cell lines from mutant Drosophila strains. We recently discovered that expression of activated Ras (Ras(V12)) provides cells in vitro with both a survival and a proliferative advantage and hence promotes the generation of cell lines. In this addendum, we provide new data describing the genesis of seven cell lines corresponding to a rumi mutant, which demonstrate that the method can be used to derive lines and study genetic mutants in vitro.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Animals , Cell Culture Techniques , Cell Line , DNA Mutational Analysis , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Fibroblasts/cytology , Genotype , Glucosyltransferases/genetics , RNA Interference
8.
Development ; 130(4): 659-70, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12505997

ABSTRACT

The Drosophila wing disc is divided along the proximal-distal axis into regions giving rise to the body wall (proximal), wing hinge (central) and wing blade (distal). We applied DNA microarray analysis to discover genes with potential roles in the development of these regions. We identified a set of 94 transcripts enriched (two fold or greater) in the body wall and 56 transcripts enriched in the wing/hinge region. Transcripts that are known to have highly restricted expression patterns, such as pannier, twist and Bar-H1 (body wall) and knot, nubbin and Distal-less (wing/hinge), showed strong differential expression on the arrays. In situ hybridization for 50 previously uncharacterized genes similarly revealed that transcript enrichment identified by the array analysis was consistent with the observed spatial expression. There was a broad spectrum of patterns, in some cases suggesting that the genes could be targets of known signaling pathways. We show that three of these genes respond to wingless signaling. We also discovered genes likely to play specific roles in tracheal and myoblast cell types, as these cells are part of the body wall fragment. In summary, the identification of genes with restricted expression patterns using whole genome profiling suggests that many genes with potential roles in wing disc development remain to be characterized.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Gene Expression Regulation, Developmental , Wings, Animal/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/metabolism , Eye Proteins/genetics , Homeodomain Proteins/genetics , In Situ Hybridization , Larva , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Oligonucleotides/genetics , POU Domain Factors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription Factors/genetics , Twist-Related Protein 1 , Wings, Animal/cytology , Wings, Animal/growth & development , Wnt1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL