Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
RNA ; 29(7): 1020-1032, 2023 07.
Article in English | MEDLINE | ID: mdl-37041032

ABSTRACT

The level of each RNA species depends on the balance between its rates of production and decay. Although previous studies have measured RNA decay across the genome in tissue culture and single-celled organisms, few experiments have been performed in intact complex tissues and organs. It is therefore unclear whether the determinants of RNA decay found in cultured cells are preserved in an intact tissue, and whether they differ between neighboring cell types and are regulated during development. To address these questions, we measured RNA synthesis and decay rates genome wide via metabolic labeling of whole cultured Drosophila larval brains using 4-thiouridine. Our analysis revealed that decay rates span a range of more than 100-fold, and that RNA stability is linked to gene function, with mRNAs encoding transcription factors being much less stable than mRNAs involved in core metabolic functions. Surprisingly, among transcription factor mRNAs there was a clear demarcation between more widely used transcription factors and those that are expressed only transiently during development. mRNAs encoding transient transcription factors are among the least stable in the brain. These mRNAs are characterized by epigenetic silencing in most cell types, as shown by their enrichment with the histone modification H3K27me3. Our data suggest the presence of an mRNA destabilizing mechanism targeted to these transiently expressed transcription factors to allow their levels to be regulated rapidly with high precision. Our study also demonstrates a general method for measuring mRNA transcription and decay rates in intact organs or tissues, offering insights into the role of mRNA stability in the regulation of complex developmental programs.


Subject(s)
Drosophila , Transcription Factors , Animals , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Drosophila/genetics , Larva/genetics , Larva/metabolism , Brain/metabolism , RNA Stability
2.
Genes Dev ; 28(16): 1772-85, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25081352

ABSTRACT

Splicing of pre-mRNAs results in the deposition of the exon junction complex (EJC) upstream of exon-exon boundaries. The EJC plays crucial post-splicing roles in export, translation, localization, and nonsense-mediated decay of mRNAs. It also aids faithful splicing of pre-mRNAs containing large introns, albeit via an unknown mechanism. Here, we show that the core EJC plus the accessory factors RnpS1 and Acinus aid in definition and efficient splicing of neighboring introns. This requires prior deposition of the EJC in close proximity to either an upstream or downstream splicing event. If present in isolation, EJC-dependent introns are splicing-defective also in wild-type cells. Interestingly, the most affected intron belongs to the piwi locus, which explains the reported transposon desilencing in EJC-depleted Drosophila ovaries. Based on a transcriptome-wide analysis, we propose that the dependency of splicing on the EJC is exploited as a means to control the temporal order of splicing events.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/genetics , Drosophila/metabolism , Introns/genetics , RNA Splicing/physiology , Animals , Argonaute Proteins/genetics , Cells, Cultured , DNA Transposable Elements/genetics , Drosophila/classification , Drosophila Proteins/genetics , Female , Gene Expression Regulation , Gene Silencing , Ovary/metabolism , RNA Precursors/metabolism
3.
Development ; 144(19): 3454-3464, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28851709

ABSTRACT

The termination of the proliferation of Drosophila neural stem cells, also known as neuroblasts (NBs), requires a 'decommissioning' phase that is controlled in a lineage-specific manner. Most NBs, with the exception of those of the mushroom body (MB), are decommissioned by the ecdysone receptor and mediator complex, causing them to shrink during metamorphosis, followed by nuclear accumulation of Prospero and cell cycle exit. Here, we demonstrate that the levels of Imp and Syp RNA-binding proteins regulate NB decommissioning. Descending Imp and ascending Syp expression have been shown to regulate neuronal temporal fate. We show that Imp levels decline slower in the MB than in other central brain NBs. MB NBs continue to express Imp into pupation, and the presence of Imp prevents decommissioning partly by inhibiting the mediator complex. Late-larval induction of transgenic Imp prevents many non-MB NBs from decommissioning in early pupae. Moreover, the presence of abundant Syp in aged NBs permits Prospero accumulation that, in turn, promotes cell cycle exit. Together, our results reveal that progeny temporal fate and progenitor decommissioning are co-regulated in protracted neuronal lineages.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Neural Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Nucleus/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Larva/metabolism , Mediator Complex/metabolism , Models, Biological , Mushroom Bodies/cytology , Mushroom Bodies/metabolism , Neural Stem Cells/cytology , Protein Binding , Pupa/metabolism , RNA-Binding Proteins/genetics
4.
EMBO J ; 33(14): 1582-98, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24920579

ABSTRACT

We have identified a new function for the dynein adaptor Bicaudal D homolog 1 (BICD1) by screening a siRNA library for genes affecting the dynamics of neurotrophin receptor-containing endosomes in motor neurons (MNs). Depleting BICD1 increased the intracellular accumulation of brain-derived neurotrophic factor (BDNF)-activated TrkB and p75 neurotrophin receptor (p75(NTR)) by disrupting the endosomal sorting, reducing lysosomal degradation and increasing the co-localisation of these neurotrophin receptors with retromer-associated sorting nexin 1. The resulting re-routing of active receptors increased their recycling to the plasma membrane and altered the repertoire of signalling-competent TrkB isoforms and p75(NTR) available for ligand binding on the neuronal surface. This resulted in attenuated, but more sustained, AKT activation in response to BDNF stimulation. These data, together with our observation that Bicd1 expression is restricted to the developing nervous system when neurotrophin receptor expression peaks, indicate that BICD1 regulates neurotrophin signalling by modulating the endosomal sorting of internalised ligand-activated receptors.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Endosomes/metabolism , Models, Biological , Motor Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Cytoskeletal Proteins/genetics , Fluorescent Antibody Technique , Immunohistochemistry , Luminescent Proteins , Mice , Microscopy, Electron, Transmission , Protein Transport/physiology , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Red Fluorescent Protein
5.
Methods ; 126: 166-176, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28651965

ABSTRACT

RNA in situ hybridization is a powerful method to investigate post-transcriptional regulation, but analysis of intracellular mRNA distributions in thick, complex tissues like the brain poses significant challenges. Here, we describe the application of single-molecule fluorescent in situ hybridization (smFISH) to quantitate primary nascent transcription and post-transcriptional regulation in whole-mount Drosophila larval and adult brains. Combining immunofluorescence and smFISH probes for different regions of a single gene, i.e., exons, 3'UTR, and introns, we show examples of a gene that is regulated post-transcriptionally and one that is regulated at the level of transcription. Our simple and rapid protocol can be used to co-visualise a variety of different transcripts and proteins in neuronal stem cells as well as deep brain structures such as mushroom body neuropils, using conventional confocal microscopy. Finally, we introduce the use of smFISH as a sensitive alternative to immunofluorescence for labelling specific neural stem cell populations in the brain.


Subject(s)
Brain/cytology , Brain/physiology , Drosophila/cytology , Drosophila/physiology , In Situ Hybridization, Fluorescence/methods , Single Molecule Imaging/methods , Animals , Drosophila/chemistry , RNA Processing, Post-Transcriptional/physiology
6.
Development ; 141(8): 1780-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24715465

ABSTRACT

Formation of somites, the rudiments of vertebrate body segments, is an oscillatory process governed by a gene-expression oscillator, the segmentation clock. This operates in each cell of the presomitic mesoderm (PSM), but the individual cells drift out of synchrony when Delta/Notch signalling fails, causing gross anatomical defects. We and others have suggested that this is because synchrony is maintained by pulses of Notch activation, delivered cyclically by each cell to its neighbours, that serve to adjust or reset the phase of the intracellular oscillator. This, however, has never been proved. Here, we provide direct experimental evidence, using zebrafish containing a heat-shock-driven transgene that lets us deliver artificial pulses of expression of the Notch ligand DeltaC. In DeltaC-defective embryos, in which endogenous Notch signalling fails, the artificial pulses restore synchrony, thereby rescuing somite formation. The spacing of segment boundaries produced by repetitive heat-shocking varies according to the time interval between one heat-shock and the next. The induced synchrony is manifest both morphologically and at the level of the oscillations of her1, a core component of the intracellular oscillator. Thus, entrainment of intracellular clocks by periodic activation of the Notch pathway is indeed the mechanism maintaining cell synchrony during somitogenesis.


Subject(s)
Biological Clocks , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptor, Notch1/metabolism , Somites/cytology , Somites/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Count , Embryo, Nonmammalian/metabolism , Heat-Shock Response , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Models, Biological , Time Factors , Transgenes , Zebrafish/embryology , Zebrafish/metabolism
7.
Proc Natl Acad Sci U S A ; 110(46): E4316-24, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24151332

ABSTRACT

Sequential production of body segments in vertebrate embryos is regulated by a molecular oscillator (the segmentation clock) that drives cyclic transcription of genes involved in positioning intersegmental boundaries. Mathematical modeling indicates that the period of the clock depends on the total delay kinetics of a negative feedback circuit, including those associated with the synthesis of transcripts encoding clock components [Lewis J (2003) Curr Biol 13(16):1398-1408]. Here, we measure expression delays for three transcripts [Lunatic fringe, Hes7/her1, and Notch-regulated-ankyrin-repeat-protein (Nrarp)], that cycle during segmentation in the zebrafish, chick, and mouse, and provide in vivo measurements of endogenous splicing and export kinetics. We show that mRNA splicing and export are much slower than transcript elongation, with the longest delay (about 16 min in the mouse) being due to mRNA export. We conclude that the kinetics of mRNA and protein production and destruction can account for much of the clock period, and provide strong support for delayed autorepression as the underlying mechanism of the segmentation clock.


Subject(s)
Biological Clocks/physiology , Body Patterning/physiology , Gene Expression Regulation, Developmental/physiology , Models, Biological , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Chick Embryo , Glycosyltransferases/metabolism , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins , Mice , Proteins/metabolism , RNA Splicing/physiology , Species Specificity , Time Factors , Zebrafish
8.
Development ; 137(21): 3591-601, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20876653

ABSTRACT

Integrins act at signalling crossroads, and their interactions with other signal transduction pathways are key to the regulation of normal and pathological cell cytoarchitecture and behaviour. Here, we describe a signalling cascade that acts during the formation of the defining segmental features of the vertebrate body - the somites - in which ß1-integrin activity regulates epithelialisation by controlling downstream Wnt and Notch activity crucial for somite border formation. Using in vivo transcriptional inhibition in the developing chick embryo, we show that ß1-integrin in the anterior presomitic mesoderm activates canonical Wnt signalling in a cell-autonomous, `outside-inside' manner. Signalling is mediated by integrin-linked kinase (ILK), leading to modulation of glycogen synthase kinase 3ß (GSK3ß) phosphorylation, and activates Notch signalling in the anterior presomitic mesoderm. The two signalling pathways then cooperate to promote somite formation via cMESO1/Mesp2. Our results show that ß1-integrin can regulate cell shape and tissue morphogenesis indirectly, by regulation of downstream signalling cascades.


Subject(s)
Integrin beta1/physiology , Receptors, Notch/metabolism , Somites/embryology , Wnt Proteins/metabolism , Animals , Animals, Genetically Modified , Cells, Cultured , Chick Embryo , Embryo Culture Techniques , Gene Knockdown Techniques , Integrin beta1/genetics , Models, Biological , Organogenesis/genetics , Organogenesis/physiology , Receptors, Notch/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Somites/metabolism , Wnt Proteins/physiology
10.
Biol Open ; 9(2)2020 02 11.
Article in English | MEDLINE | ID: mdl-31988092

ABSTRACT

During vertebrate embryonic development, the formation of axial structures is driven by a population of stem-like cells that reside in a region of the tailbud called the chordoneural hinge (CNH). We have compared the mouse CNH transcriptome with those of surrounding tissues and shown that the CNH and tailbud mesoderm are transcriptionally similar, and distinct from the presomitic mesoderm. Amongst CNH-enriched genes are several that are required for axial elongation, including Wnt3a, Cdx2, Brachyury/T and Fgf8, and androgen/oestrogen receptor nuclear signalling components such as Greb1 We show that the pattern and duration of tailbud Greb1 expression is conserved in mouse, zebrafish and chicken embryos, and that Greb1 is required for axial elongation and somitogenesis in zebrafish embryos. The axial truncation phenotype of Greb1 morphant embryos can be explained by much reduced expression of No tail (Ntl/Brachyury), which is required for axial progenitor maintenance. Posterior segmentation defects in the morphants (including misexpression of genes such as mespb, myoD and papC) appear to result, in part, from lost expression of the segmentation clock gene, her7.


Subject(s)
Embryonic Development/genetics , Membrane Proteins/genetics , Morphogenesis/genetics , Neoplasm Proteins/genetics , Zebrafish Proteins/genetics , Animals , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Phenotype , Stem Cells/cytology , Stem Cells/metabolism , Transcriptome
11.
Elife ; 92020 01 14.
Article in English | MEDLINE | ID: mdl-31934860

ABSTRACT

The numerous neurons and glia that form the brain originate from tightly controlled growth and division of neural stem cells, regulated systemically by important known stem cell-extrinsic signals. However, the cell-intrinsic mechanisms that control the distinctive proliferation rates of individual neural stem cells are unknown. Here, we show that the size and division rates of Drosophila neural stem cells (neuroblasts) are controlled by the highly conserved RNA binding protein Imp (IGF2BP), via one of its top binding targets in the brain, myc mRNA. We show that Imp stabilises myc mRNA leading to increased Myc protein levels, larger neuroblasts, and faster division rates. Declining Imp levels throughout development limit myc mRNA stability to restrain neuroblast growth and division, and heterogeneous Imp expression correlates with myc mRNA stability between individual neuroblasts in the brain. We propose that Imp-dependent regulation of myc mRNA stability fine-tunes individual neural stem cell proliferation rates.


The brain is a highly complex organ made up of huge numbers of different cell types that connect up to form a precise network. All these different cell types are generated from the repeated division of a relatively small pool of cells called neural stem cells. The division of these cells needs to be carefully regulated so that the correct number and type of nerve cells are produced at the right time and place. But it remains unclear how the division rate of individual neural stem cells is controlled during development. Controlling these divisions requires the activity of countless genes to be tightly regulated over space and time. When a gene is active, it is copied via a process called transcription into a single-stranded molecule known as messenger RNA (or mRNA for short). This molecule provides the instructions needed to build the protein encoded within the gene. Proteins are the functional building blocks of all cells. The conventional way of controlling protein levels is to vary the number of mRNA molecules made by transcription. Now, Samuels et al. reveal a second mechanism of determining protein levels in the brain, through regulating the stability of mRNA after it is transcribed. Samuels et al. discovered that a key regulatory protein called Imp controls the growth and division of individual neural stem cells in the brains of developing fruit flies. The experiments showed that Imp binds to mRNA molecules that contain the code for a protein called Myc, which is known to drive cell growth and division in many different cell types. Both human Imp and Myc have been implicated in cancer. Using a technique that images single molecules of mRNA, Samuels et al. showed that the Imp protein in stem cells stabilises the mRNA molecule coding for Myc. This means that when more Imp is present, more Myc protein gets produced. Thus, the level of Imp in each individual neural stem cell fine-tunes the rate at which the cell grows and divides: the higher the level of Imp, the larger the stem cell and the faster it divides. These findings underscore how important post-transcriptional processes are for regulating gene activity in the developing brain. The methods used in this study to study mRNA molecules in single cells also provide new insights that could not be derived from the average measurements of many cells. Similar methods could also be applied to other developmental systems in the future.


Subject(s)
Brain/embryology , DNA-Binding Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Neural Stem Cells/cytology , RNA Stability , RNA-Binding Proteins/chemistry , Transcription Factors/metabolism , Animals , Brain/cytology , Cell Differentiation , Cell Proliferation , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , In Situ Hybridization, Fluorescence , Larva , Male , Mushroom Bodies/cytology , Mushroom Bodies/metabolism , Neural Stem Cells/metabolism , Protein Binding , RNA Interference , RNA, Messenger/metabolism , Signal Transduction
12.
J Cell Biol ; 219(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-32040548

ABSTRACT

Memory and learning involve activity-driven expression of proteins and cytoskeletal reorganization at new synapses, requiring posttranscriptional regulation of localized mRNA a long distance from corresponding nuclei. A key factor expressed early in synapse formation is Msp300/Nesprin-1, which organizes actin filaments around the new synapse. How Msp300 expression is regulated during synaptic plasticity is poorly understood. Here, we show that activity-dependent accumulation of Msp300 in the postsynaptic compartment of the Drosophila larval neuromuscular junction is regulated by the conserved RNA binding protein Syncrip/hnRNP Q. Syncrip (Syp) binds to msp300 transcripts and is essential for plasticity. Single-molecule imaging shows that msp300 is associated with Syp in vivo and forms ribosome-rich granules that contain the translation factor eIF4E. Elevated neural activity alters the dynamics of Syp and the number of msp300:Syp:eIF4E RNP granules at the synapse, suggesting that these particles facilitate translation. These results introduce Syp as an important early acting activity-dependent regulator of a plasticity gene that is strongly associated with human ataxias.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Neuronal Plasticity , RNA-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Developmental , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Skeletal/embryology , Neuromuscular Junction/embryology , Neuromuscular Junction/genetics , RNA-Binding Proteins/genetics , Time Factors
13.
Biol Open ; 9(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32205310

ABSTRACT

During Drosophila and vertebrate brain development, the conserved transcription factor Prospero/Prox1 is an important regulator of the transition between proliferation and differentiation. Prospero level is low in neural stem cells and their immediate progeny, but is upregulated in larval neurons and it is unknown how this process is controlled. Here, we use single molecule fluorescent in situ hybridisation to show that larval neurons selectively transcribe a long prospero mRNA isoform containing a 15 kb 3' untranslated region, which is bound in the brain by the conserved RNA-binding protein Syncrip/hnRNPQ. Syncrip binding increases the stability of the long prospero mRNA isoform, which allows an upregulation of Prospero protein production. Adult flies selectively lacking the long prospero isoform show abnormal behaviour that could result from impaired locomotor or neurological activity. Our findings highlight a regulatory strategy involving alternative polyadenylation followed by differential post-transcriptional regulation.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Drosophila Proteins/genetics , Drosophila/physiology , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Neurons/metabolism , Nuclear Proteins/genetics , Polyadenylation , RNA, Messenger/genetics , Transcription Factors/genetics , 3' Untranslated Regions , Animals , Drosophila Proteins/metabolism , Immunohistochemistry , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Organ Specificity/genetics , RNA Stability , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism
14.
Dev Cell ; 3(1): 63-74, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12110168

ABSTRACT

A molecular oscillator regulates the pace of vertebrate segmentation. Here, we show that the oscillator (clock) controls cyclic initiation of transcription in the unsegmented presomitic mesoderm (PSM). We identify an evolutionarily conserved 2.3 kb region in the murine Lunatic fringe (Lfng) promoter that drives periodic expression in the PSM. This region includes conserved blocks required for enhancing and repressing cyclic Lfng transcription, and to prevent continued expression in formed somites. We also show that dynamic expression in the cycling PSM is lost in the total absence of Notch signaling, and that Notch signaling acts directly via CBF1/RBP-Jkappa binding sites to regulate Lfng. These results are consistent with a model in which oscillatory Notch signaling underlies the segmentation clock and directly activates and indirectly represses Lfng expression.


Subject(s)
Biological Clocks/genetics , Body Patterning/genetics , Embryo, Mammalian/embryology , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/physiology , Glycosyltransferases/genetics , Membrane Proteins/genetics , Transcriptional Activation/genetics , Animals , Avian Proteins , Chick Embryo , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Evolution, Molecular , Genes, Reporter/genetics , Glycosyltransferases/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/metabolism , Periodicity , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Receptors, Notch , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Transcription, Genetic/physiology , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
15.
Curr Biol ; 14(21): 1950-6, 2004 Nov 09.
Article in English | MEDLINE | ID: mdl-15530398

ABSTRACT

Drosophila neuroblasts undergo asymmetric divisions along the apicobasal axis to produce two daughter cells of unequal size and different developmental fate. Inscuteable (Insc) protein functions as part of an apically localized complex to coordinate orientation of the mitotic spindle and basal sorting of cell fate determinants. insc mRNA transcripts also localize apically in neuroblasts, yet the mechanism underpinning this process and its developmental significance are unknown. Here, we show that the Egalitarian (Egl)/Bicaudal-D (BicD)/dynein mRNA transport machinery mediates apical localization of insc mRNA transcripts in neuroblasts, and we provide evidence that insc localization is required for efficient apical targeting of Insc protein. egl and BicD mutant neuroblasts display defects in apicobasal polarity, which is consistent with apical Insc activity being reduced. Also, we observe shortened mitotic spindles at metaphase in egl, BicD, and insc mutant neuroblasts and demonstrate a previously unknown, dose-dependent requirement for Insc in augmenting metaphase spindle length. We conclude that localization of insc mRNA transcripts in neuroblasts confers maximal levels of apical Insc activity, which is required for accurate control of metaphase spindle length, division orientation, and asymmetric cell division.


Subject(s)
Cell Differentiation/physiology , Cytoskeletal Proteins/metabolism , Neurons/physiology , RNA, Messenger/metabolism , Spindle Apparatus/physiology , Animals , Biological Transport , Cytoskeletal Proteins/genetics , Drosophila , Drosophila Proteins/metabolism , Dyneins/metabolism , Fluorescence , Immunohistochemistry , In Situ Hybridization , Neurons/metabolism , Neuropeptides
16.
Curr Biol ; 14(18): 1680-4, 2004 Sep 21.
Article in English | MEDLINE | ID: mdl-15380072

ABSTRACT

Segmental patterning in Drosophila relies on a cascade of transcription factors that subdivide the embryo into successively more precise domains. We have identified a missense mutation (W049) in the gene encoding the transcriptional elongation factor Spt5 (reviewed in ) which, when homozygous in the maternal germ line, leads to defects in segmental patterning of the embryo. W049 alters the C-terminal domain of Spt5 and affects its activity in vitro, impairing its abilities to confer sensitivity to the transcriptional inhibitor DRB and to stimulate transcription at limiting nucleotide concentrations. In vivo, W049 shows locus-specific effects on transcription: expression of gap genes remains wild-type, but striped patterning of the primary pair-rule genes even-skipped and runt is disrupted. even-skipped stripes are broadened in the mutant embryos indicating that Spt5 is likely to be a direct, negative regulator of this target gene. Our results suggest control of transcriptional elongation by repressors contributes to striped gene expression in the embryo. By contrast, expression of heat shock-induced proteins is reduced in the mutant embryos. These results provide genetic evidence for Spt5 function during heat shock induction and demonstrate that Spt5 acts both positively and negatively on transcription in vivo depending on context.


Subject(s)
Body Patterning/genetics , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Developmental , Transcriptional Activation/genetics , Transcriptional Elongation Factors/genetics , Animals , DNA Mutational Analysis , DNA Primers , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/metabolism , Heat-Shock Response/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Mutation, Missense/genetics , Nuclear Proteins , Polymorphism, Single-Stranded Conformational , Transcription Factors/metabolism
17.
Genetics ; 170(2): 697-708, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15834145

ABSTRACT

We describe the maternal-effect and zygotic phenotypes of null mutations in the Drosophila gene for the epsilon-subunit of mitochondrial ATP synthase, stunted (sun). Loss of zygotic sun expression leads to a dramatic delay in the growth rate of first instar larvae and ultimately death. Embryos lacking maternally supplied sun (sun embryos) have a sixfold reduction in ATP synthase activity. Cellular analysis of sun embryos shows defects only after the nuclei have migrated to the cortex. During the cortical divisions the actin-based metaphase and cellularization furrows do not form properly, and the nuclei show abnormal spacing and division failures. The most striking abnormality is that nuclei and spindles form lines and clusters, instead of adopting a regular spacing. This is reflected in a failure to properly position neighboring nonsister centrosomes during the telophase-to-interphase transition of the cortical divisions. Our study is consistent with a role for Sun in mitochondrial ATP synthesis and suggests that reduced ATP levels selectively affect molecular motors. As Sun has been identified as the ligand for the Methuselah receptor that regulates aging, Sun may function both within and outside mitochondria.


Subject(s)
Drosophila/embryology , Mitochondrial Proton-Translocating ATPases/chemistry , Actins/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Cell Movement , Cell Nucleus/metabolism , Centrosome/ultrastructure , Cytoskeleton/metabolism , DNA/metabolism , Drosophila/physiology , Female , Interphase , Ligands , Male , Microscopy, Fluorescence , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/physiology , Models, Genetic , Models, Molecular , Molecular Motor Proteins , Molecular Sequence Data , Phenotype , Sequence Homology, Amino Acid , Spindle Apparatus , Telophase
19.
Int J Dev Biol ; 47(4): 263-72, 2003 May.
Article in English | MEDLINE | ID: mdl-12755331

ABSTRACT

Notch signalling plays a major role in many invertebrate and vertebrate patterning systems. In this paper, we use high-titre, non-replicative pseudotype viruses to show that the two Notch ligands, Delta1 and Serrate1 (Jagged1), have differing activities in the developing chick spinal cord and hindbrain. In the walls of the neural tube, Serrate1 appears not to affect neurogenesis, in contrast to Delta1 which mediates lateral inhibition as elsewhere in the nervous system. In the floorplate we find that there is also a requirement for Notch, but with a different type of dependence on the two Notch ligands: cells with a floorplate character are lost when Notch activity is blocked with dominant-negative, truncated forms of either Delta1 or Serrate1. Our results are consistent with ligand-receptor specificity within the Notch signalling pathway, Serrate1 recognising selectively Notch2 (which is expressed in the floorplate), and Delta1 acting on both Notch2 and Notch1 (which is expressed in the walls of the neural tube).


Subject(s)
Membrane Proteins/physiology , Rhombencephalon/embryology , Spinal Cord/embryology , Transcription Factors , Animals , Base Sequence , Calcium-Binding Proteins , Chick Embryo , DNA/genetics , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Ligands , Membrane Proteins/genetics , Proteins/genetics , Proteins/physiology , Receptor, Notch1 , Receptor, Notch2 , Receptors, Cell Surface/physiology , Receptors, Notch , Retroviridae/genetics , Serrate-Jagged Proteins , Signal Transduction , Transfection
20.
Gene Expr Patterns ; 3(3): 325-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12799079

ABSTRACT

To study the role of receptor tyrosine phosphatases in vertebrate development, in particular somitogenesis, we have cloned chick receptor tyrosine phosphatase Psi (cRPTPPsi). cRPTPPsi is expressed in a dynamic fashion in the somites to-be-formed and uniformly throughout the presomitic mesoderm. In differentiating somites, cRPTPPsi expression gets restricted to the dermomyotome. In addition cRPTPPsi is expressed in the developing intermediate mesoderm, in neurogenic and sensory organs, the limb bud and the developing heart.


Subject(s)
Protein Tyrosine Phosphatases/genetics , Somites/metabolism , Amino Acid Sequence , Animals , Chick Embryo , Humans , In Situ Hybridization , Mice , Molecular Sequence Data , Protein Tyrosine Phosphatases/biosynthesis , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL