Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cell ; 153(5): 941-3, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706731

ABSTRACT

Enhancers have been intensely studied as the sequences determining spatial and temporal gene expression during development. Lagha et al. now put the focus back on the promoter as the critical element coordinating gene expression across a cell population.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , Animals
2.
PLoS Biol ; 21(1): e3001956, 2023 01.
Article in English | MEDLINE | ID: mdl-36649329

ABSTRACT

Regulation of mRNA degradation is critical for a diverse array of cellular processes and developmental cell fate decisions. Many methods for determining mRNA half-lives rely on transcriptional inhibition or metabolic labelling. Here, we use a non-invasive method for estimating half-lives for hundreds of mRNAs in the early Drosophila embryo. This approach uses the intronic and exonic reads from a total RNA-seq time series and Gaussian process regression to model the dynamics of premature and mature mRNAs. We show how regulation of mRNA stability is used to establish a range of mature mRNA dynamics during embryogenesis, despite shared transcription profiles. Using single-molecule imaging, we provide evidence that, for the mRNAs tested, there is a correlation between short half-life and mRNA association with P-bodies. Moreover, we detect an enrichment of mRNA 3' ends in P-bodies in the early embryo, consistent with 5' to 3' degradation occurring in P-bodies for at least a subset of mRNAs. We discuss our findings in relation to recently published data suggesting that the primary function of P-bodies in other biological contexts is mRNA storage.


Subject(s)
Drosophila , Processing Bodies , Animals , Drosophila/genetics , Drosophila/metabolism , Single Molecule Imaging , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA Stability/genetics
3.
Development ; 148(18)2021 09 15.
Article in English | MEDLINE | ID: mdl-33722899

ABSTRACT

The Hunchback (Hb) transcription factor is crucial for anterior-posterior patterning of the Drosophila embryo. The maternal hb mRNA acts as a paradigm for translational regulation due to its repression in the posterior of the embryo. However, little is known about the translatability of zygotically transcribed hb mRNAs. Here, we adapt the SunTag system, developed for imaging translation at single-mRNA resolution in tissue culture cells, to the Drosophila embryo to study the translation dynamics of zygotic hb mRNAs. Using single-molecule imaging in fixed and live embryos, we provide evidence for translational repression of zygotic SunTag-hb mRNAs. Whereas the proportion of SunTag-hb mRNAs translated is initially uniform, translation declines from the anterior over time until it becomes restricted to a posterior band in the expression domain. We discuss how regulated hb mRNA translation may help establish the sharp Hb expression boundary, which is a model for precision and noise during developmental patterning. Overall, our data show how use of the SunTag method on fixed and live embryos is a powerful combination for elucidating spatiotemporal regulation of mRNA translation in Drosophila.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila/genetics , Protein Biosynthesis/genetics , RNA, Messenger, Stored/genetics , Transcription Factors/genetics , Animals , Body Patterning/genetics , Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental/genetics , Morphogenesis/genetics , Zygote/physiology
4.
Bioinformatics ; 38(4): 1030-1036, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34788793

ABSTRACT

MOTIVATION: The MS2-MCP (MS2 coat protein) live imaging system allows for visualization of transcription dynamics through the introduction of hairpin stem-loop sequences into a gene. A fluorescent signal at the site of nascent transcription in the nucleus quantifies mRNA production. Computational modelling can be used to infer the promoter states along with the kinetic parameters governing transcription, such as promoter switching frequency and polymerase loading rate. However, modelling of the fluorescent trace presents a challenge due its persistence; the observed fluorescence at a given time point depends on both current and previous promoter states. A compound state Hidden Markov Model (cpHMM) was recently introduced to allow inference of promoter activity from MS2-MCP data. However, the computational time for inference scales exponentially with gene length and the cpHMM is therefore not currently practical for application to many eukaryotic genes. RESULTS: We present a scalable implementation of the cpHMM for fast inference of promoter activity and transcriptional kinetic parameters. This new method can model genes of arbitrary length through the use of a time-adaptive truncated compound state space. The truncated state space provides a good approximation to the full state space by retaining the most likely set of states at each time during the forward pass of the algorithm. Testing on MS2-MCP fluorescent data collected from early Drosophila melanogaster embryos indicates that the method provides accurate inference of kinetic parameters within a computationally feasible timeframe. The inferred promoter traces generated by the model can also be used to infer single-cell transcriptional parameters. AVAILABILITY AND IMPLEMENTATION: Python implementation is available at https://github.com/ManchesterBioinference/burstInfer, along with code to reproduce the examples presented here. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Algorithms , Drosophila melanogaster , Animals , Drosophila melanogaster/genetics , Time Factors , Computer Simulation
5.
Genes Dev ; 27(10): 1146-58, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23699410

ABSTRACT

Cascades of zygotic gene expression pattern the anterior-posterior (AP) and dorsal-ventral (DV) axes of the early Drosophila embryo. Here, we used the global run-on sequencing assay (GRO-seq) to map the genome-wide RNA polymerase distribution during early Drosophila embryogenesis, thus providing insights into how genes are regulated. We identify widespread promoter-proximal pausing yet show that the presence of paused polymerase does not necessarily equate to direct regulation through pause release to productive elongation. Our data reveal that a subset of early Zelda-activated genes is regulated at the level of polymerase recruitment, whereas other Zelda target and axis patterning genes are predominantly regulated through pause release. In contrast to other signaling pathways, we found that bone morphogenetic protein (BMP) target genes are collectively more highly paused than BMP pathway components and show that BMP target gene expression requires the pause-inducing negative elongation factor (NELF) complex. Our data also suggest that polymerase pausing allows plasticity in gene activation throughout embryogenesis, as transiently repressed and transcriptionally silenced genes maintain and lose promoter polymerases, respectively. Finally, we provide evidence that the major effect of pausing is on the levels, rather than timing, of transcription. These data are discussed in terms of the efficiency of transcriptional activation required across cell populations during developmental time constraints.


Subject(s)
Body Patterning/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , RNA Polymerase II/metabolism , Transcription, Genetic , Animals , Drosophila Proteins/metabolism , Female , Nuclear Proteins , Transcription Factors/metabolism , Zygote/metabolism
6.
PLoS Genet ; 12(7): e1006164, 2016 07.
Article in English | MEDLINE | ID: mdl-27379389

ABSTRACT

The BMP signaling pathway has a conserved role in dorsal-ventral axis patterning during embryonic development. In Drosophila, graded BMP signaling is transduced by the Mad transcription factor and opposed by the Brinker repressor. In this study, using the Drosophila embryo as a model, we combine RNA-seq with Mad and Brinker ChIP-seq to decipher the BMP-responsive transcriptional network underpinning differentiation of the dorsal ectoderm during dorsal-ventral axis patterning. We identify multiple new BMP target genes, including positive and negative regulators of EGF signaling. Manipulation of EGF signaling levels by loss- and gain-of-function studies reveals that EGF signaling negatively regulates embryonic BMP-responsive transcription. Therefore, the BMP gene network has a self-regulating property in that it establishes a balance between its activity and that of the antagonistic EGF signaling pathway to facilitate correct patterning. In terms of BMP-dependent transcription, we identify key roles for the Zelda and Zerknüllt transcription factors in establishing the resulting expression domain, and find widespread binding of insulator proteins to the Mad and Brinker-bound genomic regions. Analysis of embryos lacking the BEAF-32 insulator protein shows reduced transcription of a peak BMP target gene and a reduction in the number of amnioserosa cells, the fate specified by peak BMP signaling. We incorporate our findings into a model for Mad-dependent activation, and discuss its relevance to BMP signal interpretation in vertebrates.


Subject(s)
Bone Morphogenetic Proteins/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , Body Patterning/genetics , Bone Morphogenetic Proteins/biosynthesis , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Embryonic Development/genetics , Epidermal Growth Factor/genetics , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Nuclear Proteins , Signal Transduction/genetics
7.
Development ; 142(19): 3362-73, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26293305

ABSTRACT

Precise control of the range of signalling molecule action is crucial for correct cell fate patterning during development. For example, Drosophila ovarian germline stem cells (GSCs) are maintained by exquisitely short-range BMP signalling from the niche. In the absence of BMP signalling, one GSC daughter differentiates into a cystoblast (CB) and this fate is stabilised by Brain tumour (Brat) and Pumilio (Pum)-mediated post-transcriptional repression of mRNAs, including that encoding the Dpp transducer, Mad. However, the identity of other repressed mRNAs and the mechanism of post-transcriptional repression are currently unknown. Here, we identify the Medea and schnurri mRNAs, which encode transcriptional regulators required for activation and/or repression of Dpp target genes, as additional Pum-Brat targets, suggesting that tripartite repression of the transducers is deployed to desensitise the CB to Dpp. In addition, we show that repression by Pum-Brat requires recruitment of the CCR4 and Pop2 deadenylases, with knockdown of deadenylases in vivo giving rise to ectopic GSCs. Consistent with this, Pum-Brat repression leads to poly(A) tail shortening and mRNA degradation in tissue culture cells, and we detect a reduced number of Mad and shn transcripts in the CB relative to the GSC based on single molecule mRNA quantitation. Finally, we show generality of the mechanism by demonstrating that Brat also attenuates pMad and Dpp signalling range in the early embryo. Together our data serve as a platform for understanding how post-transcriptional repression restricts interpretation of BMPs and other cell signals in order to allow robust cell fate patterning during development.


Subject(s)
Adult Stem Cells/physiology , Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/growth & development , Gene Expression Regulation, Developmental/physiology , Ovary/cytology , Smad4 Protein/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Differentiation/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization
8.
Cell Mol Life Sci ; 74(11): 1937-1956, 2017 06.
Article in English | MEDLINE | ID: mdl-27999899

ABSTRACT

Tissue patterning, through the concerted activity of a small number of signaling pathways, is critical to embryonic development. While patterning can involve signaling between neighbouring cells, in other contexts signals act over greater distances by traversing complex cellular landscapes to instruct the fate of distant cells. In this review, we explore different strategies adopted by cells to modulate signaling molecule range to allow correct patterning. We describe mechanisms for restricting signaling range and highlight how such short-range signaling can be exploited to not only control the fate of adjacent cells, but also to generate graded signaling within a field of cells. Other strategies include modulation of signaling molecule action by tissue architectural properties and the use of cellular membranous structures, such as signaling filopodia and exosomes, to actively deliver signaling ligands to target cells. Signaling filopodia can also be deployed to reach out and collect particular signals, thereby precisely controlling their site of action.


Subject(s)
Body Patterning , Embryonic Development , Signal Transduction , Animals , Cell Surface Extensions/metabolism , Extracellular Vesicles/metabolism , Humans , Ligands
9.
Biochem Soc Trans ; 44(5): 1465-1473, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27911728

ABSTRACT

The bone morphogenetic protein (BMP) pathway is a major conserved signalling pathway with diverse roles in development and homeostasis. Given that cells exist in three-dimensional environments, one important area is to understand how the BMP pathway operates within such complex cellular environments. The extracellular matrix contains information regarding tissue architecture and its mechanical properties that is transmitted to the cell via integrin receptors. In this review, I describe various examples of modulation of the BMP pathway by integrins. In the case of the Drosophila embryo and some cell line-based studies, integrins have been found to enhance BMP responses through different mechanisms, such as enhancement of BMP ligand-receptor binding and effects on Smad phosphorylation or stability. In these contexts, BMP-dependent activation of integrins is a common theme. However, I also discuss examples where integrins inhibit the BMP pathway, highlighting the context-dependent nature of integrin-BMP cross-talk.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Extracellular Matrix/metabolism , Integrins/metabolism , Signal Transduction , Animals , Extracellular Matrix Proteins/metabolism , Humans , Models, Biological , Protein Binding , Smad Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 109(28): 11222-7, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733779

ABSTRACT

In the Drosophila embryo, formation of a bone morphogenetic protein (BMP) morphogen gradient requires transport of a heterodimer of the BMPs Decapentaplegic (Dpp) and Screw (Scw) in a protein shuttling complex. Although the core components of the shuttling complex--Short Gastrulation (Sog) and Twisted Gastrulation (Tsg)--have been identified, key aspects of this shuttling system remain mechanistically unresolved. Recently, we discovered that the extracellular matrix protein collagen IV is important for BMP gradient formation. Here, we formulate a molecular mechanism of BMP shuttling that is catalyzed by collagen IV. We show that Dpp is the only BMP ligand in Drosophila that binds collagen IV. A collagen IV binding-deficient Dpp mutant signals at longer range in vivo, indicating that collagen IV functions to immobilize free Dpp in the embryo. We also provide in vivo evidence that collagen IV functions as a scaffold to promote shuttling complex assembly in a multistep process. After binding of Dpp/Scw and Sog to collagen IV, protein interactions are remodeled, generating an intermediate complex in which Dpp/Scw-Sog is poised for release by Tsg through specific disruption of a collagen IV-Sog interaction. Because all components are evolutionarily conserved, we propose that regulation of BMP shuttling and immobilization through extracellular matrix interactions is widely used, both during development and in tissue homeostasis, to achieve a precise extracellular BMP distribution.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/cytology , Gene Expression Regulation, Developmental , Amino Acid Sequence , Animals , Binding Sites , Collagen Type IV/chemistry , Collagen Type IV/metabolism , Dimerization , Extracellular Matrix/metabolism , Gastrula/physiology , Glutathione Transferase/metabolism , Ligands , Molecular Sequence Data , Protein Conformation , Protein Transport , Sequence Homology, Amino Acid
11.
Nature ; 455(7209): 72-7, 2008 Sep 04.
Article in English | MEDLINE | ID: mdl-18701888

ABSTRACT

Dorsal-ventral patterning in vertebrate and invertebrate embryos is mediated by a conserved system of secreted proteins that establishes a bone morphogenetic protein (BMP) gradient. Although the Drosophila embryonic Decapentaplegic (Dpp) gradient has served as a model to understand how morphogen gradients are established, no role for the extracellular matrix has been previously described. Here we show that type IV collagen extracellular matrix proteins bind Dpp and regulate its signalling in both the Drosophila embryo and ovary. We provide evidence that the interaction between Dpp and type IV collagen augments Dpp signalling in the embryo by promoting gradient formation, yet it restricts the signalling range in the ovary through sequestration of the Dpp ligand. Together, these results identify a critical function of type IV collagens in modulating Dpp in the extracellular space during Drosophila development. On the basis of our findings that human type IV collagen binds BMP4, we predict that this role of type IV collagens will be conserved.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Collagen Type IV/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Signal Transduction , Animals , Body Patterning , Bone Morphogenetic Proteins/genetics , Cell Count , Collagen Type IV/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Female , Male , Ovary/cytology , Ovary/metabolism , Protein Binding , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Nat Commun ; 15(1): 4976, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862520

ABSTRACT

Twisted gastrulation (TWSG1) is an evolutionarily conserved secreted glycoprotein which controls signaling by Bone Morphogenetic Proteins (BMPs). TWSG1 binds BMPs and their antagonist Chordin to control BMP signaling during embryonic development, kidney regeneration and cancer. We report crystal structures of TWSG1 alone and in complex with a BMP ligand, Growth Differentiation Factor 5. TWSG1 is composed of two distinct, disulfide-rich domains. The TWSG1 N-terminal domain occupies the BMP type 1 receptor binding site on BMPs, whereas the C-terminal domain binds to a Chordin family member. We show that TWSG1 inhibits BMP function in cellular signaling assays and mouse colon organoids. This inhibitory function is abolished in a TWSG1 mutant that cannot bind BMPs. The same mutation in the Drosophila TWSG1 ortholog Tsg fails to mediate BMP gradient formation required for dorsal-ventral axis patterning of the early embryo. Our studies reveal the evolutionarily conserved mechanism of BMP signaling inhibition by TWSG1.


Subject(s)
Bone Morphogenetic Proteins , Signal Transduction , Animals , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Mice , Humans , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/chemistry , Glycoproteins/metabolism , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Binding Sites , Protein Domains , Protein Binding , Organoids/metabolism , Organoids/embryology , HEK293 Cells , Gastrulation/genetics , Mutation , Crystallography, X-Ray , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Proteins
13.
J Biol Chem ; 287(8): 5942-53, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22199351

ABSTRACT

Bone morphogenetic proteins (BMPs) are synthesized as proproteins that undergo proteolytic processing by furin/subtilisin proprotein convertases to release the active ligand. Here we study processing of BMP5/6/7/8 proteins, including the Drosophila orthologs Glass Bottom Boat (Gbb) and Screw (Scw) and human BMP7. Gbb and Scw have three functional furin/subtilisin proprotein convertase cleavage sites; two between the prodomain and ligand domain, which we call the Main and Shadow sites, and one within the prodomain, which we call the Pro site. In Gbb each site can be cleaved independently, although efficient cleavage at the Shadow site requires cleavage at the Main site, and remarkably, none of the sites is essential for Gbb function. Rather, Gbb must be processed at either the Pro or Main site to produce a functional ligand. Like Gbb, the Pro and Main sites in Scw can be cleaved independently, but cleavage at the Shadow site is dependent on cleavage at the Main site. However, both Pro and Main sites are essential for Scw function. Thus, Gbb and Scw have different processing requirements. The BMP7 ligand rescues gbb mutants in Drosophila, but full-length BMP7 cannot, showing that functional differences in the prodomain limit the BMP7 activity in flies. Furthermore, unlike Gbb, cleavage-resistant BMP7, although non-functional in rescue assays, activates the downstream signaling cascade and thus retains some functionality. Our data show that cleavage requirements evolve rapidly, supporting the notion that changes in post-translational processing are used to create functional diversity between BMPs within and between species.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Proteolysis , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Animals , Binding Sites , Bone Morphogenetic Protein 5/chemistry , Bone Morphogenetic Protein 5/metabolism , Bone Morphogenetic Protein 6/chemistry , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Proteins/chemistry , Conserved Sequence , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Evolution, Molecular , Humans , Ligands , Molecular Sequence Data , Mutation , Proprotein Convertases/metabolism , Protein Structure, Tertiary , Signal Transduction , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/genetics
14.
EMBO Rep ; 12(6): 519-26, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21546910

ABSTRACT

Drosophila ovarian germline stem cells (GSCs) are maintained by the extracellular BMP2/4 orthologue Dpp, which is produced from the surrounding somatic niche. The Dpp signal has a short range; it induces a response in GSCs within the niche, but is rapidly extinguished in their progeny only one cell-diameter away. To ensure the correct balance between stem-cell maintenance and differentiation, several regulatory mechanisms that modulate the Dpp signal at many stages of the pathway have been described. Here, we discuss the nature of the ovarian Dpp signal and review the catalogue of mechanisms that regulate it, demonstrating how the exquisite modulation of Dpp signalling in this context can result in precise and robust control of stem-cell fate. This modulation is applicable to other stem-cell environments that use BMPs as a niche signal, and the regulatory mechanisms are conceptually relevant to several other stem-cell systems.


Subject(s)
Drosophila/metabolism , Signal Transduction , Stem Cell Niche/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Drosophila/cytology , Extracellular Space/metabolism , Female , Germ Cells/metabolism , Humans , Intracellular Space/metabolism , Ovary/cytology , Ovary/metabolism
15.
Cell Rep ; 42(4): 112382, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37060568

ABSTRACT

Dosage compensation, the balancing of X-linked gene expression between sexes and to the autosomes, is critical to an organism's fitness and survival. In Drosophila, dosage compensation involves hypertranscription of the male X chromosome. Here, we use quantitative live imaging and modeling at single-cell resolution to study X chromosome dosage compensation in Drosophila. We show that the four X chromosome genes studied undergo transcriptional bursting in male and female embryos. Mechanistically, our data reveal that transcriptional upregulation of male X chromosome genes is primarily mediated by a higher RNA polymerase II initiation rate and burst amplitude across the expression domain. In contrast, burst frequency is spatially modulated in nuclei within the expression domain in response to different transcription factor concentrations to tune the transcriptional response. Together, these data show how the local and global regulation of distinct burst parameters can establish the complex transcriptional outputs underpinning developmental patterning.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Male , Female , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , X Chromosome/metabolism , Dosage Compensation, Genetic
16.
Biol Open ; 11(6)2022 06 15.
Article in English | MEDLINE | ID: mdl-35603711

ABSTRACT

A BMP gradient is essential for patterning the dorsal-ventral axis of invertebrate and vertebrate embryos. The extracellular BMP binding protein Short Gastrulation (Sog) in Drosophila plays a key role in BMP gradient formation. In this study, we combine genome editing, structural and developmental approaches to study Sog function in Drosophila. We generate a sog knockout fly stock, which allows simple reintegration of altered versions of the sog coding sequence. As proof-of-principle, we test the requirement for two cysteine residues that were previously identified as targets for palmitoylation, which has been proposed to enhance Sog secretion. However, we show that the sogC27,28S mutant is viable with only very mild phenotypes, indicating that these residues and their potential modification are not critical for Sog secretion in vivo. Additionally, we use experimental negative stain EM imaging and hydrodynamic data to validate the AlphaFold structure prediction for Sog. The model suggests a more compact shape than the vertebrate ortholog Chordin and conformational flexibility between the C-terminal von Willebrand C domains. We discuss how this altered compactness may contribute to mechanistic differences in Sog and Chordin function during BMP gradient formation. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Bone Morphogenetic Proteins/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/metabolism , Gastrulation , Humans , Signal Transduction
17.
RNA Biol ; 8(5): 904-12, 2011.
Article in English | MEDLINE | ID: mdl-21788736

ABSTRACT

During Drosophila development, translational control plays a crucial role in regulating gene expression, and is particularly important during pre-patterning of the maturing oocyte. A critical step in translation initiation is the binding of the eukaryotic translation initiation factor 4E (eIF4E) to the mRNA cap structure, which ultimately leads to recruitment of the ribosome. d4EHP is a translational repressor that prevents translation initiation by out-competing eIF4E on the cap structure for a subset of mRNAs. However, only two examples of mRNAs subject to d4EHP translation repression in Drosophila are known. Here we show that the belle (bel) mRNA is translationally repressed by the d4EHP protein in the Drosophila ovary. Consistent with this regulation, d4EHP overexpression in the ovary phenocopies the bel mutant. We also provide evidence that the Bel protein binds to eIF4E and may itself function as a translation repressor protein, with bruno as a potential target for Bel repression in the oocyte. Bruno is known to repress the mRNA of the key oocyte axis determinant oskar (osk) during oogenesis, and we find that an increase in the level of Bruno protein in bel mutant ovaries is associated with a reduction in Osk protein. Overall, our data suggest that a translational regulatory network exists in which consecutive translational repression events act to correctly pattern the Drosophila oocyte.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Eukaryotic Initiation Factor-4E/metabolism , Oocytes/physiology , RNA Helicases/genetics , Animals , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/biosynthesis , Gene Expression Regulation, Developmental , Oogenesis/genetics , Protein Binding , Protein Biosynthesis/genetics , RNA Caps/metabolism , RNA Helicases/biosynthesis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
18.
STAR Protoc ; 2(1): 100379, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33778778

ABSTRACT

Visualizing transcription live in Drosophila is providing important new insights into the spatiotemporal regulation of transcription. Here, we describe a protocol to visualize and quantitate transcription from gene loci that are tagged with MS2 stem-loop sequences in the Drosophila embryo. MS2 stem-loop sequences are recognized by a coat protein fused to a fluorescent protein and visualized with microscopy. We also describe an analysis pipeline to extract and subsequently quantify transcription dynamics. For complete details on the use and execution of this protocol, please refer to Hoppe et al. (2020).


Subject(s)
Drosophila/embryology , Gene Expression Profiling/methods , Microscopy, Fluorescence/methods , Animals , Capsid Proteins/genetics , Inverted Repeat Sequences/genetics , RNA, Messenger/genetics , Tomography, X-Ray Computed/methods
19.
STAR Protoc ; 2(1): 100380, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33786461

ABSTRACT

CRISPR-Cas9 genome editing has transformed biology by enabling site-specific genome modifications to be simply engineered. Here, we describe two CRISPR-Cas9 approaches to introduce MS2 stem-loop sequences into endogenous gene loci in Drosophila. This can facilitate live imaging of nascent transcription in Drosophila. For complete details on the use and execution of this protocol, please refer to Hoppe et al. (2020).


Subject(s)
Drosophila/embryology , Gene Editing/methods , Gene Expression Profiling/methods , Animals , CRISPR-Cas Systems , Genome , Inverted Repeat Sequences/genetics , RNA, Messenger/genetics
20.
STAR Protoc ; 2(3): 100812, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34585149

ABSTRACT

Significant regulation of gene expression is mediated at the translation level. Here, we describe protocols for imaging and analysis of translation at single mRNA resolution in both fixed and living Drosophila embryos. These protocols use the SunTag system, in which the protein of interest is visualized by inserting a peptide array that is recognized by a single chain antibody. Simultaneous detection of individual mRNAs using the MS2/MCP system or by smFISH allows translation sites to be identified and quantified. For complete information on the generation and use of this protocol, please refer to Vinter et al. (2021).


Subject(s)
Embryo, Nonmammalian/metabolism , Protein Biosynthesis/genetics , RNA, Messenger/analysis , Single Molecule Imaging/methods , Animals , Drosophila/genetics , Drosophila/metabolism , Embryo, Nonmammalian/chemistry , Female , In Situ Hybridization, Fluorescence , Male , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Single-Chain Antibodies
SELECTION OF CITATIONS
SEARCH DETAIL