Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(6): e0305696, 2024.
Article in English | MEDLINE | ID: mdl-38913612

ABSTRACT

In Drosophila coordinated proliferation of two neural stem cells, neuroblasts (NB) and neuroepithelial (NE) cells, is pivotal for proper larval brain growth that ultimately determines the final size and performance of an adult brain. The larval brain growth displays two phases based on behaviors of NB and NEs: the first one in early larval stages, influenced by nutritional status and the second one in the last larval stage, promoted by ecdysone signaling after critical weight checkpoint. Mutations of the baboon (babo) gene that produces three isoforms (BaboA-C), all acting as type-I receptors of Activin-type transforming growth factor ß (TGF-ß) signaling, cause a small brain phenotype due to severely reduced proliferation of the neural stem cells. In this study we show that loss of babo function severely affects proliferation of NBs and NEs as well as conversion of NEs from both phases. By analyzing babo-null and newly generated isoform-specific mutants by CRISPR mutagenesis as well as isoform-specific RNAi knockdowns in a cell- and stage-specific manner, our data support differential contributions of the isoforms for these cellular events with BaboA playing the major role. Stage-specific expression of EcR-B1 in the brain is also regulated primarily by BaboA along with function of the other isoforms. Blocking EcR function in both neural stem cells results in a small brain phenotype that is more severe than baboA-knockdown alone. In summary, our study proposes that the Babo-mediated signaling promotes proper behaviors of the neural stem cells in both phases and achieves this by acting upstream of EcR-B1 expression in the second phase.


Subject(s)
Brain , Cell Proliferation , Drosophila Proteins , Larva , Neural Stem Cells , Neuroepithelial Cells , Protein Isoforms , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Larva/metabolism , Larva/genetics , Larva/growth & development , Protein Isoforms/metabolism , Protein Isoforms/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Brain/metabolism , Neuroepithelial Cells/metabolism , Neuroepithelial Cells/cytology , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Signal Transduction , Activin Receptors/metabolism , Activin Receptors/genetics
2.
PLoS Biol ; 20(5): e3001660, 2022 05.
Article in English | MEDLINE | ID: mdl-35594316

ABSTRACT

In polarized epithelial cells, receptor-ligand interactions can be restricted by different spatial distributions of the 2 interacting components, giving rise to an underappreciated layer of regulatory complexity. We explored whether such regulation occurs in the Drosophila wing disc, an epithelial tissue featuring the TGF-ß family member Decapentaplegic (Dpp) as a morphogen controlling growth and patterning. Dpp protein has been observed in an extracellular gradient within the columnar cell layer of the disc, but also uniformly in the disc lumen, leading to the question of how graded signaling is achieved in the face of 2 distinctly localized ligand pools. We find the Dpp Type II receptor Punt, but not the Type I receptor Tkv, is enriched at the basolateral membrane and depleted at the junctions and apical surface. Wit, a second Type II receptor, shows a markedly different behavior, with the protein detected on all membrane regions but enriched at the apical side. Mutational studies identified a short juxtamembrane sequence required for basolateral restriction of Punt in both wing discs and mammalian Madin-Darby canine kidney (MDCK) cells. This basolateral targeting (BLT) determinant can dominantly confer basolateral localization on an otherwise apical receptor. Rescue of punt mutants with transgenes altered in the targeting motif showed that flies expressing apicalized Punt due to the lack of a functional BLT displayed developmental defects, female sterility, and significant lethality. We also show that apicalized Punt does not produce an ectopic signal, indicating that the apical pool of Dpp is not a significant signaling source even when presented with Punt. Instead, we find that basolateral presentation of Punt is required for optimal signaling. Finally, we present evidence that the BLT acts through polarized sorting machinery that differs between types of epithelia. This suggests a code whereby each epithelial cell type may differentially traffic common receptors to enable distinctive responses to spatially localized pools of extracellular ligands.


Subject(s)
Drosophila Proteins , Drosophila , Transforming Growth Factor beta , Animals , Cell Membrane/metabolism , Dogs , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Ligands , Madin Darby Canine Kidney Cells , Protein Serine-Threonine Kinases , Receptors, Cell Surface/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
3.
Elife ; 112022 01 17.
Article in English | MEDLINE | ID: mdl-35037619

ABSTRACT

Hedgehog (Hh) and Bone Morphogenetic Proteins (BMPs) pattern the developing Drosophila wing by functioning as short- and long-range morphogens, respectively. Here, we show that a previously unknown Hh-dependent mechanism fine-tunes the activity of BMPs. Through genome-wide expression profiling of the Drosophila wing imaginal discs, we identify nord as a novel target gene of the Hh signaling pathway. Nord is related to the vertebrate Neuron-Derived Neurotrophic Factor (NDNF) involved in congenital hypogonadotropic hypogonadism and several types of cancer. Loss- and gain-of-function analyses implicate Nord in the regulation of wing growth and proper crossvein patterning. At the molecular level, we present biochemical evidence that Nord is a secreted BMP-binding protein and localizes to the extracellular matrix. Nord binds to Decapentaplegic (Dpp) or the heterodimer Dpp-Glass-bottom boat (Gbb) to modulate their release and activity. Furthermore, we demonstrate that Nord is a dosage-dependent BMP modulator, where low levels of Nord promote and high levels inhibit BMP signaling. Taken together, we propose that Hh-induced Nord expression fine-tunes both the range and strength of BMP signaling in the developing Drosophila wing.


Subject(s)
Body Patterning/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/metabolism , Wings, Animal/growth & development , Animals , Body Patterning/genetics , Drosophila Proteins/genetics , Hedgehog Proteins/genetics , Larva/growth & development , Larva/metabolism
4.
Nat Commun ; 11(1): 4468, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32901021

ABSTRACT

Speciation constrains the flow of genetic information between populations of sexually reproducing organisms. Gaining control over mechanisms of speciation would enable new strategies to manage wild populations of disease vectors, agricultural pests, and invasive species. Additionally, such control would provide safe biocontainment of transgenes and gene drives. Here, we demonstrate a general approach to create engineered genetic incompatibilities (EGIs) in the model insect Drosophila melanogaster. EGI couples a dominant lethal transgene with a recessive resistance allele. Strains homozygous for both elements are fertile and fecund when they mate with similarly engineered strains, but incompatible with wild-type strains that lack resistant alleles. EGI genotypes can also be tuned to cause hybrid lethality at different developmental life-stages. Further, we demonstrate that multiple orthogonal EGI strains of D. melanogaster can be engineered to be mutually incompatible with wild-type and with each other. EGI is a simple and robust approach in multiple sexually reproducing organisms.


Subject(s)
Drosophila melanogaster/genetics , Genetic Engineering/methods , Genetic Speciation , Animals , Animals, Genetically Modified , Crosses, Genetic , Female , Genes, Insect , Genes, Lethal , Genotype , Hybridization, Genetic , Male , Models, Genetic , Transgenes
5.
Elife ; 92020 07 07.
Article in English | MEDLINE | ID: mdl-32633716

ABSTRACT

Organ growth and size are finely tuned by intrinsic and extrinsic signaling molecules. In Drosophila, the BMP family member Dpp is produced in a limited set of imaginal disc cells and functions as a classic morphogen to regulate pattern and growth by diffusing throughout imaginal discs. However, the role of TGFß/Activin-like ligands in disc growth control remains ill-defined. Here, we demonstrate that Myoglianin (Myo), an Activin family member, and a close homolog of mammalian Myostatin (Mstn), is a muscle-derived extrinsic factor that uses canonical dSmad2-mediated signaling to regulate wing size. We propose that Myo is a myokine that helps mediate an allometric relationship between muscles and their associated appendages.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Imaginal Discs/growth & development , Smad Proteins, Receptor-Regulated/physiology , Transforming Growth Factor beta/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Imaginal Discs/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Muscles/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
6.
Cell ; 174(1): 18-20, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29958107

ABSTRACT

A new study combines detailed biochemical characterization with whole-animal genetics and computational transcriptome data mining to reveal how the LRRC33 milieu molecule imposes an exquisite level of spatial control on TGF-ß signaling in the CNS.


Subject(s)
Transforming Growth Factor beta/genetics , Animals , Cell Communication , Microglia , Signal Transduction , Transcriptome
7.
Cell Cycle ; 14(16): 2677-87, 2015.
Article in English | MEDLINE | ID: mdl-26125535

ABSTRACT

The transforming growth factor ß (TGF-ß) and bone morphogenetic protein (BMP) pathways transduce extracellular signals into tissue-specific transcriptional responses. During this process, signaling effector Smad proteins translocate into the nucleus to direct changes in transcription, but how and where they localize to DNA remain important questions. We have mapped Drosophila TGF-ß signaling factors Mad, dSmad2, Medea, and Schnurri genome-wide in Kc cells and find that numerous sites for these factors overlap with the architectural protein CTCF. Depletion of CTCF by RNAi results in the disappearance of a subset of Smad sites, suggesting Smad proteins localize to CTCF binding sites in a CTCF-dependent manner. Sensitive Smad binding sites are enriched at low occupancy CTCF peaks within topological domains, rather than at the physical domain boundaries where CTCF may function as an insulator. In response to Decapentaplegic, CTCF binding is not significantly altered, whereas Mad, Medea, and Schnurri are redirected from CTCF to non-CTCF binding sites. These results suggest that CTCF participates in the recruitment of Smad proteins to a subset of genomic sites and in the redistribution of these proteins in response to BMP signaling.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Repressor Proteins/physiology , Smad Proteins/metabolism , Animals , Base Sequence , Binding Sites , CCCTC-Binding Factor , Cell Line , Consensus Sequence , Drosophila melanogaster/genetics , Epigenesis, Genetic , Protein Transport , Signal Transduction , Transcription, Genetic , Transforming Growth Factor beta/physiology
8.
Methods ; 68(1): 183-93, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24680699

ABSTRACT

The TGF-ß pathway is an evolutionarily conserved signal transduction module that mediates diverse biological processes in animals. In Drosophila, both the BMP and Activin branches are required for viability. Studies rooted in classical and molecular genetic approaches continue to uncover new developmental roles for TGF-ß signaling. We present an overview of the secreted ligands, transmembrane receptors and cellular Smad transducer proteins that compose the core pathway in Drosophila. An assortment of tools have been developed to conduct tissue-specific loss- and gain-of-function experiments for these pathway components. We discuss the deployment of these reagents, with an emphasis on appropriate usage and limitations of the available tools. Throughout, we note reagents that are in need of further improvement or development, and signaling features requiring further study. A general theme is that comparison of phenotypes for ligands, receptors, and Smads can be used to map tissue interactions, and to separate canonical and non-canonical signaling activities. Core TGF-ß signaling components are subject to multiple layers of regulation, and are coupled to context-specific inputs and outputs. In addition to fleshing out how TGF-ß signaling serves the fruit fly, we anticipate that future studies will uncover new regulatory nodes and modes and will continue to advance paradigms for how TGF-ß signaling regulates general developmental processes.


Subject(s)
Signal Transduction , Transforming Growth Factor beta/genetics , Activins/genetics , Activins/metabolism , Animals , DNA-Binding Proteins/genetics , Developmental Biology/methods , Drosophila/genetics , Gene Expression Regulation, Developmental , Phosphorylation , Transforming Growth Factor beta/metabolism
9.
Development ; 140(3): 649-59, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23293296

ABSTRACT

Imaginal disc development in Drosophila requires coordinated cellular proliferation and tissue patterning. In our studies of TGFß superfamily signaling components, we found that a protein null mutation of Smad2, the only Activin subfamily R-Smad in the fruit fly, produces overgrown wing discs that resemble gain of function for BMP subfamily signaling. The wing discs are expanded specifically along the anterior-posterior axis, with increased proliferation in lateral regions. The morphological defect is not observed in mutants for the TGFß receptor baboon, and epistasis tests showed that baboon is epistatic to Smad2 for disc overgrowth. Rescue experiments indicate that Baboon binding, but not canonical transcription factor activity, of Smad2 is required for normal disc growth. Smad2 mutant discs generate a P-Mad stripe that is narrower and sharper than the normal gradient, and activation targets are correspondingly expressed in narrowed domains. Repression targets of P-Mad are profoundly mis-regulated, with brinker and pentagone reporter expression eliminated in Smad2 mutants. Loss of expression requires a silencer element previously shown to be controlled by BMP signaling. Epistasis experiments show that Baboon, Mad and Schnurri are required to mediate the ectopic silencer output in the absence of Smad2. Taken together, our results show that loss of Smad2 permits promiscuous Baboon activity, which represses genes subject to control by Mad-dependent silencer elements. The absence of Brinker and Pentagone in Smad2 mutants explains the compound wing disc phenotype. Our results highlight the physiological relevance of substrate inhibition of a kinase, and reveal a novel interplay between the Activin and BMP pathways.


Subject(s)
Activin Receptors/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Gene Expression Regulation, Developmental , Imaginal Discs/physiology , Smad2 Protein/metabolism , Wings, Animal/physiology , Activin Receptors/genetics , Alleles , Animals , Body Patterning , Bone Morphogenetic Proteins/metabolism , Cell Proliferation , Cell Size , Crosses, Genetic , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/genetics , Epistasis, Genetic , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Imaginal Discs/metabolism , Immunohistochemistry , Male , Mutagenesis, Site-Directed , Mutation , Organ Size , Phenotype , RNA Interference , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated , Smad2 Protein/genetics , Time Factors , Wings, Animal/metabolism
10.
PLoS One ; 7(5): e36548, 2012.
Article in English | MEDLINE | ID: mdl-22563507

ABSTRACT

Animals use TGF-ß superfamily signal transduction pathways during development and tissue maintenance. The superfamily has traditionally been divided into TGF-ß/Activin and BMP branches based on relationships between ligands, receptors, and R-Smads. Several previous reports have shown that, in cell culture systems, "BMP-specific" Smads can be phosphorylated in response to TGF-ß/Activin pathway activation. Using Drosophila cell culture as well as in vivo assays, we find that Baboon, the Drosophila TGF-ß/Activin-specific Type I receptor, can phosphorylate Mad, the BMP-specific R-Smad, in addition to its normal substrate, dSmad2. The Baboon-Mad activation appears direct because it occurs in the absence of canonical BMP Type I receptors. Wing phenotypes generated by Baboon gain-of-function require Mad, and are partially suppressed by over-expression of dSmad2. In the larval wing disc, activated Baboon cell-autonomously causes C-terminal Mad phosphorylation, but only when endogenous dSmad2 protein is depleted. The Baboon-Mad relationship is thus controlled by dSmad2 levels. Elevated P-Mad is seen in several tissues of dSmad2 protein-null mutant larvae, and these levels are normalized in dSmad2; baboon double mutants, indicating that the cross-talk reaction and Smad competition occur with endogenous levels of signaling components in vivo. In addition, we find that high levels of Activin signaling cause substantial turnover in dSmad2 protein, providing a potential cross-pathway signal-switching mechanism. We propose that the dual activity of TGF-ß/Activin receptors is an ancient feature, and we discuss several ways this activity can modulate TGF-ß signaling output.


Subject(s)
Activin Receptors/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Smad2 Protein/metabolism , Transcription Factors/metabolism , Activin Receptors/genetics , Animals , Blotting, Western , Cell Line , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Female , Larva/growth & development , Larva/metabolism , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptor Cross-Talk , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated , Smad2 Protein/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Wings, Animal/growth & development , Wings, Animal/metabolism
11.
Sci Signal ; 5(218): pe14, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22472647

ABSTRACT

Signaling molecules of the transforming growth factor (TGF)-ß family are generated from proprotein precursors containing prodomain sequences that are typically removed to allow signaling by the mature ligands. A form of a TGF-ß family ligand that remains covalently attached to its prodomain but retains signaling activity has been identified. Glass bottom boat (Gbb), a Drosophila homolog of the bone morphogenetic protein 5/6/7/8 subfamily, is active as a carboxyl-terminal fragment of the proprotein (Gbb15) that is generated by a conventional processing event common to TGF-ß ligands. Unexpectedly, a larger form (Gbb38) produced by processing at a newly identified furin site in the prodomain is also secreted and active. Contrary to the present paradigm in which TGF-ß ligands require dissociation of the entire prodomain for activity, Gbb38 is active in cell culture and in vivo without additional processing at conventional sites. The large form can restore the viability of gbb mutant animals but has distinct signaling properties compared with the conventional form. Production of multiple functional ligands from one proprotein is a potential mechanism to fine-tune TGF-ß signaling outputs. Mutations in TGF-ß family members have been linked to human diseases, several of which affect potential furin cleavage sites in prodomains. However, given the diversity of potential furin processing sites and prodomain functions, direct experimentation will be required to determine whether production of active jumbo ligands is a general feature of TGF-ß superfamily members.


Subject(s)
Drosophila Proteins/metabolism , Protein Precursors/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Binding Sites/genetics , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Furin/metabolism , Humans , Models, Biological , Mutation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Precursors/genetics , Protein Processing, Post-Translational , Transforming Growth Factor beta/genetics
12.
Stud Health Technol Inform ; 129(Pt 2): 1250-4, 2007.
Article in English | MEDLINE | ID: mdl-17911915

ABSTRACT

Many biological processes rely on remodeling of the transcriptional response of cells through activation of transcription factors. Although determination of the activity level of transcription factors from microarray data can provide insight into developmental and disease processes, it requires careful analysis because of the multiple regulation of genes. We present a novel approach that handles both the assignment of genes to multiple patterns, as required by multiple regulation, and the linking of genes in prior probability distributions according to their known transcriptional regulators. We demonstrate the power of this approach in simulations and by application to yeast cell cycle and deletion mutant data. The results of simulations in the presence of increasing noise showed improved recovery of patterns in terms of chi2 fit. Analysis of the yeast data led to improved inference of biologically meaningful groups in comparison to other techniques, as demonstrated with ROC analysis. The new algorithm provides an approach for estimating the levels of transcription factor activity from microarray data, and therefore provides insights into biological response.


Subject(s)
Algorithms , Gene Expression Regulation , Oligonucleotide Array Sequence Analysis , Transcription Factors/metabolism , Bayes Theorem , Computational Biology , Markov Chains , Models, Genetic , Monte Carlo Method , ROC Curve , Transcription, Genetic , Yeasts/genetics
13.
Methods Mol Biol ; 377: 243-54, 2007.
Article in English | MEDLINE | ID: mdl-17634621

ABSTRACT

Typical microarray or GeneChip experiments now provide genome-wide measurements on gene expression across many conditions. Analysis often focuses on only a few of the genes, looking for those that are "differentially expressed" between conditions or groups of conditions. However, the large number of measurements both present statistical problems to such single gene approaches and offers a tremendous amount of information for methods focused on biological processes rather than individual genes. Here we provide a method to utilize biological annotations in the form of gene ontologies to interpret the results of individual or multiple pattern recognition analyses of a microarray experiment.


Subject(s)
Data Interpretation, Statistical , Genes , Microarray Analysis/methods , Molecular Biology/methods , Animals , Cluster Analysis , Gene Expression , Genome , Humans , Pattern Recognition, Automated
14.
Genetics ; 167(3): 1225-39, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15280237

ABSTRACT

The Drosophila Sex Comb on Midleg (SCM) protein is a transcriptional repressor of the Polycomb group (PcG). Although genetic studies establish SCM as a crucial PcG member, its molecular role is not known. To investigate how SCM might link to PcG complexes, we analyzed the in vivo role of a conserved protein interaction module, the SPM domain. This domain is found in SCM and in another PcG protein, Polyhomeotic (PH), which is a core component of Polycomb repressive complex 1 (PRC1). SCM-PH interactions in vitro are mediated by their respective SPM domains. Yeast two-hybrid and in vitro binding assays were used to isolate and characterize >30 missense mutations in the SPM domain of SCM. Genetic rescue assays showed that SCM repressor function in vivo is disrupted by mutations that impair SPM domain interactions in vitro. Furthermore, overexpression of an isolated, wild-type SPM domain produced PcG loss-of-function phenotypes in flies. Coassembly of SCM with a reconstituted PRC1 core complex shows that SCM can partner with PRC1. However, gel filtration chromatography showed that the bulk of SCM is biochemically separable from PH in embryo nuclear extracts. These results suggest that SCM, although not a core component of PRC1, interacts and functions with PRC1 in gene silencing.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/genetics , Gene Silencing , Phenotype , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Chromatography, Gel , DNA Primers , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Gene Components , Immunoprecipitation , Molecular Sequence Data , Mutation, Missense/genetics , Nucleoproteins/genetics , Nucleoproteins/metabolism , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...