Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 68
1.
Development ; 151(11)2024 Jun 01.
Article En | MEDLINE | ID: mdl-38819456

Drosophila nervous system development progresses through a series of well-characterized steps in which homeodomain transcription factors (HDTFs) play key roles during most, if not all, phases. Strikingly, although some HDTFs have only one role, many others are involved in multiple steps of the developmental process. Most Drosophila HDTFs engaged in nervous system development are conserved in vertebrates and often play similar roles during vertebrate development. In this Spotlight, we focus on the role of HDTFs during embryogenesis, where they were first characterized.


Drosophila Proteins , Homeodomain Proteins , Nervous System , Transcription Factors , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Nervous System/metabolism , Nervous System/embryology , Transcription Factors/metabolism , Transcription Factors/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Drosophila/genetics , Drosophila/metabolism , Drosophila/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism
2.
Neurosci Lett ; 824: 137675, 2024 Feb 28.
Article En | MEDLINE | ID: mdl-38355003

The rapid evolution of different imaging modalities in the last two decades has enabled the investigation of the role of different genes in development and disease to be studied in a range of model organisms. However, selection of the appropriate imaging technique depends on a number of constraints, including cost, time, image resolution, size of the sample, computational complexity and processing power. Here, we use the adult mouse central nervous system to investigate whether High-Resolution Episcopic Microscopy (HREM) can provide an effective means to study the volume of individual subregions within the brain. We find that HREM can provide precise volume quantification of different structures within the mouse brain, albeit with limitations regarding the time involved for analysis and the necessity of some estimations.


Imaging, Three-Dimensional , Microscopy , Mice , Animals , Microscopy/methods , Imaging, Three-Dimensional/methods
3.
J Cell Sci ; 136(21)2023 11 01.
Article En | MEDLINE | ID: mdl-37921122

The covalent modification of histones is critical for many biological functions in mammals, including gene regulation and chromatin structure. Posttranslational histone modifications are added and removed by specialised 'writer' and 'eraser' enzymes, respectively. One such writer protein implicated in a wide range of cellular processes is SET domain-containing 2 (SETD2), a histone methyltransferase that catalyses the trimethylation of lysine 36 on histone H3 (H3K36me3). Recently, SETD2 has also been found to modify proteins other than histones, including actin and tubulin. The emerging roles of SETD2 in the development and function of the mammalian central nervous system (CNS) are of particular interest as several SETD2 variants have been implicated in neurodevelopmental disorders, such as autism spectrum disorder and the overgrowth disorder Luscan-Lumish syndrome. Here, we summarise the numerous roles of SETD2 in mammalian cellular functions and development, with a focus on the CNS. We also provide an overview of the consequences of SETD2 variants in human disease and discuss future directions for understanding essential cellular functions of SETD2.


Autism Spectrum Disorder , Histones , Animals , Humans , Histones/metabolism , Autism Spectrum Disorder/genetics , Methylation , Chromatin , Central Nervous System/metabolism , Mammals/metabolism
4.
Bioinformatics ; 39(7)2023 07 01.
Article En | MEDLINE | ID: mdl-37449901

MOTIVATION: Identification of cell types using single-cell RNA-seq is revolutionizing the study of multicellular organisms. However, typical single-cell RNA-seq analysis often involves post hoc manual curation to ensure clusters are transcriptionally distinct, which is time-consuming, error-prone, and irreproducible. RESULTS: To overcome these obstacles, we developed Cytocipher, a bioinformatics method and scverse compatible software package that statistically determines significant clusters. Application of Cytocipher to normal tissue, development, disease, and large-scale atlas data reveals the broad applicability and power of Cytocipher to generate biological insights in numerous contexts. This included the identification of cell types not previously described in the datasets analysed, such as CD8+ T cell subtypes in human peripheral blood mononuclear cells; cell lineage intermediate states during mouse pancreas development; and subpopulations of luminal epithelial cells over-represented in prostate cancer. Cytocipher also scales to large datasets with high-test performance, as shown by application to the Tabula Sapiens Atlas representing >480 000 cells. Cytocipher is a novel and generalizable method that statistically determines transcriptionally distinct and programmatically reproducible clusters from single-cell data. AVAILABILITY AND IMPLEMENTATION: The software version used for this manuscript has been deposited on Zenodo (https://doi.org/10.5281/zenodo.8089546), and is also available via github (https://github.com/BradBalderson/Cytocipher).


Algorithms , Gene Expression Profiling , Animals , Mice , Humans , Sequence Analysis, RNA/methods , Gene Expression Profiling/methods , Leukocytes, Mononuclear , Single-Cell Gene Expression Analysis , Single-Cell Analysis , Software
5.
Cell Tissue Res ; 391(2): 269-286, 2023 Feb.
Article En | MEDLINE | ID: mdl-36512054

The central nervous system contains a daunting number of different cell types. Understanding how each cell acquires its fate remains a major challenge for neurobiology. The developing embryonic ventral nerve cord (VNC) of Drosophila melanogaster has been a powerful model system for unraveling the basic principles of cell fate specification. This pertains specifically to neuropeptide neurons, which typically are stereotypically generated in discrete subsets, allowing for unambiguous single-cell resolution in different genetic contexts. Here, we study the specification of the OrcoA-LA neurons, characterized by the expression of the neuropeptide Orcokinin A and located laterally in the A1-A5 abdominal segments of the VNC. We identified the progenitor neuroblast (NB; NB5-3) and the temporal window (castor/grainyhead) that generate the OrcoA-LA neurons. We also describe the role of the Ubx, abd-A, and Abd-B Hox genes in the segment-specific generation of these neurons. Additionally, our results indicate that the OrcoA-LA neurons are "Notch Off" cells, and neither programmed cell death nor the BMP pathway appears to be involved in their specification. Finally, we performed a targeted genetic screen of 485 genes known to be expressed in the CNS and identified nab, vg, and tsh as crucial determinists for OrcoA-LA neurons. This work provides a new neuropeptidergic model that will allow for addressing new questions related to neuronal specification mechanisms in the future.


Drosophila Proteins , Neuropeptides , Animals , Drosophila , Drosophila melanogaster/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Neurons/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Repressor Proteins
6.
Cells ; 11(15)2022 08 02.
Article En | MEDLINE | ID: mdl-35954220

Nuclear factor one X (NFIX) is a transcription factor required for normal ependymal development. Constitutive loss of Nfix in mice (Nfix-/-) is associated with hydrocephalus and sloughing of the dorsal ependyma within the lateral ventricles. Previous studies have implicated NFIX in the transcriptional regulation of genes encoding for factors essential to ependymal development. However, the cellular and molecular mechanisms underpinning hydrocephalus in Nfix-/- mice are unknown. To investigate the role of NFIX in hydrocephalus, we examined ependymal cells in brains from postnatal Nfix-/- and control (Nfix+/+) mice using a combination of confocal and electron microscopy. This revealed that the ependymal cells in Nfix-/- mice exhibited abnormal cilia structure and disrupted localisation of adhesion proteins. Furthermore, we modelled ependymal cell adhesion using epithelial cell culture and revealed changes in extracellular matrix and adherens junction gene expression following knockdown of NFIX. Finally, the ablation of Nfix from ependymal cells in the adult brain using a conditional approach culminated in enlarged ventricles, sloughing of ependymal cells from the lateral ventricles and abnormal localisation of adhesion proteins, which are phenotypes observed during development. Collectively, these data demonstrate a pivotal role for NFIX in the regulation of cell adhesion within ependymal cells of the lateral ventricles.


Ependyma , Hydrocephalus , NFI Transcription Factors , Animals , Cell Physiological Phenomena , Hydrocephalus/genetics , Lateral Ventricles , Mice , NFI Transcription Factors/genetics , Neuroglia
7.
Dev Neurobiol ; 82(6): 495-504, 2022 09.
Article En | MEDLINE | ID: mdl-35796156

A striking feature of the nervous system pertains to the appearance of different neural cell subtypes at different axial levels. Studies in the Drosophila central nervous system reveal that one mechanism underlying such segmental differences pertains to the segment-specific removal of cells by programmed cell death (PCD). One group of genes involved in segment-specific PCD is the Hox homeotic genes. However, while segment-specific PCD is highly precise, Hox gene expression is evident in gradients, raising the issue of how the Hox gene function is precisely gated to trigger PCD in specific segments at the outer limits of Hox expression. The Drosophila Va neurons are initially generated in all nerve cord segments but removed by PCD in posterior segments. Va PCD is triggered by the posteriorly expressed Hox gene Abdominal-B (Abd-B). However, Va PCD is highly reproducible despite exceedingly weak Abd-B expression in the anterior frontiers of its expression. Here, we found that the transcriptional cofactor Dachshund supports Abd-B-mediated PCD in its anterior domain. In vivo bimolecular fluorescence complementation analysis lends support to the idea that the Dachshund/Abd-B interplay may involve physical interactions. These findings provide an example of how combinatorial codes of transcription factors ensure precision in Hox-mediated PCD in specific segments at the outer limits of Hox expression.


Drosophila Proteins , Drosophila , Animals , Apoptosis , Central Nervous System/metabolism , Dogs , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
8.
PLoS Genet ; 18(6): e1010255, 2022 06.
Article En | MEDLINE | ID: mdl-35737938

The MCM2-7 complex is a highly conserved hetero-hexameric protein complex, critical for DNA unwinding at the replicative fork during DNA replication. Overexpression or mutation in MCM2-7 genes is linked to and may drive several cancer types in humans. In mice, mutations in MCM2-7 genes result in growth retardation and mortality. All six MCM2-7 genes are also expressed in the developing mouse CNS, but their role in the CNS is not clear. Here, we use the central nervous system (CNS) of Drosophila melanogaster to begin addressing the role of the MCM complex during development, focusing on the specification of a well-studied neuropeptide expressing neuron: the Tv4/FMRFa neuron. In a search for genes involved in the specification of the Tv4/FMRFa neuron we identified Mcm5 and find that it plays a highly specific role in the specification of the Tv4/FMRFa neuron. We find that other components of the MCM2-7 complex phenocopies Mcm5, indicating that the role of Mcm5 in neuronal subtype specification involves the MCM2-7 complex. Surprisingly, we find no evidence of reduced progenitor proliferation, and instead find that Mcm5 is required for the expression of the type I BMP receptor Tkv, which is critical for the FMRFa expression. These results suggest that the MCM2-7 complex may play roles during CNS development outside of its well-established role during DNA replication.


Bone Morphogenetic Proteins , Cell Cycle Proteins , Drosophila Proteins , Neurons , Protein Serine-Threonine Kinases , Receptors, Cell Surface , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Replication/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Mice , Minichromosome Maintenance Proteins/genetics , Neurons/cytology , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction
9.
Development ; 149(5)2022 03 01.
Article En | MEDLINE | ID: mdl-35245348

The hypothalamus displays staggering cellular diversity, chiefly established during embryogenesis by the interplay of several signalling pathways and a battery of transcription factors. However, the contribution of epigenetic cues to hypothalamus development remains unclear. We mutated the polycomb repressor complex 2 gene Eed in the developing mouse hypothalamus, which resulted in the loss of H3K27me3, a fundamental epigenetic repressor mark. This triggered ectopic expression of posteriorly expressed regulators (e.g. Hox homeotic genes), upregulation of cell cycle inhibitors and reduced proliferation. Surprisingly, despite these effects, single cell transcriptomic analysis revealed that most neuronal subtypes were still generated in Eed mutants. However, we observed an increase in glutamatergic/GABAergic double-positive cells, as well as loss/reduction of dopamine, hypocretin and Tac2-Pax6 neurons. These findings indicate that many aspects of the hypothalamic gene regulatory flow can proceed without the key H3K27me3 epigenetic repressor mark, but points to a unique sensitivity of particular neuronal subtypes to a disrupted epigenomic landscape.


Embryonic Development/physiology , Hypothalamus/physiology , Neurons/physiology , Polycomb Repressive Complex 2/genetics , Polycomb-Group Proteins/genetics , Animals , Cell Proliferation/genetics , Epigenetic Repression/genetics , Female , Male , Mice , Mutation/genetics , Transcriptome/genetics
10.
Nucleic Acids Res ; 50(3): 1280-1296, 2022 02 22.
Article En | MEDLINE | ID: mdl-35048973

A prominent aspect of most, if not all, central nervous systems (CNSs) is that anterior regions (brain) are larger than posterior ones (spinal cord). Studies in Drosophila and mouse have revealed that Polycomb Repressor Complex 2 (PRC2), a protein complex responsible for applying key repressive histone modifications, acts by several mechanisms to promote anterior CNS expansion. However, it is unclear what the full spectrum of PRC2 action is during embryonic CNS development and how PRC2 intersects with the epigenetic landscape. We removed PRC2 function from the developing mouse CNS, by mutating the key gene Eed, and generated spatio-temporal transcriptomic data. To decode the role of PRC2, we developed a method that incorporates standard statistical analyses with probabilistic deep learning to integrate the transcriptomic response to PRC2 inactivation with epigenetic data. This multi-variate analysis corroborates the central involvement of PRC2 in anterior CNS expansion, and also identifies several unanticipated cohorts of genes, such as proliferation and immune response genes. Furthermore, the analysis reveals specific profiles of regulation via PRC2 upon these gene cohorts. These findings uncover a differential logic for the role of PRC2 upon functionally distinct gene cohorts that drive CNS anterior expansion. To support the analysis of emerging multi-modal datasets, we provide a novel bioinformatics package that integrates transcriptomic and epigenetic datasets to identify regulatory underpinnings of heterogeneous biological processes.


Central Nervous System/embryology , Polycomb Repressive Complex 2 , Animals , Embryo, Mammalian/metabolism , Histones/genetics , Histones/metabolism , Mice , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
11.
Development ; 148(12)2021 06 15.
Article En | MEDLINE | ID: mdl-34128986

The balance between stem cell potency and lineage specification entails the integration of both extrinsic and intrinsic cues, which ultimately influence gene expression through the activity of transcription factors. One example of this is provided by the Hippo signalling pathway, which plays a central role in regulating organ size during development. Hippo pathway activity is mediated by the transcriptional co-factors Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), which interact with TEA domain (TEAD) proteins to regulate gene expression. Although the roles of YAP and TAZ have been intensively studied, the roles played by TEAD proteins are less well understood. Recent studies have begun to address this, revealing that TEADs regulate the balance between progenitor self-renewal and differentiation throughout various stages of development. Furthermore, it is becoming apparent that TEAD proteins interact with other co-factors that influence stem cell biology. This Primer provides an overview of the role of TEAD proteins during development, focusing on their role in Hippo signalling as well as within other developmental, homeostatic and disease contexts.


Disease Susceptibility , Embryonic Development , Gene Expression Regulation, Developmental , Multigene Family , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Homeostasis , Humans , Molecular Targeted Therapy , Organ Specificity , Regeneration , Species Specificity , Stem Cells/cytology , Stem Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Vertebrates
12.
Neurobiol Dis ; 146: 105079, 2020 12.
Article En | MEDLINE | ID: mdl-32961270

Microtubule Associated Protein Tau (MAPT) forms proteopathic aggregates in several diseases. The G273R tau mutation, located in the first repeat region, was found by exome sequencing in a patient who presented with dementia and parkinsonism. We herein return to pathological examination which demonstrated tau immunoreactivity in neurons and glia consistent of mixed progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) features. To rationalize the pathological findings, we used molecular biophysics to characterize the mutation in more detail in vitro and in Drosophila. The G273R mutation increases the aggregation propensity of 4-repeat (4R) tau and alters the tau binding affinity towards microtubules (MTs) and F-actin. Tau aggregates in PSP and CBD are predominantly 4R tau. Our data suggest that the G273R mutation induces a shift in pool of 4R tau by lower F-actin affinity, alters the conformation of MT bound 4R tau, while increasing chaperoning of 3R tau by binding stronger to F-actin. The mutation augmented fibrillation of 4R tau initiation in vitro and in glial cells in Drosophila and showed preferential seeding of 4R tau in vitro suggestively causing a late onset 4R tauopathy reminiscent of PSP and CBD.


Brain/pathology , Neurons/metabolism , Supranuclear Palsy, Progressive/metabolism , Tauopathies/pathology , Animals , Basal Ganglia Diseases/metabolism , Brain/metabolism , Drosophila , Mutation/genetics , Neuroglia/metabolism
13.
Curr Top Dev Biol ; 137: 333-361, 2020.
Article En | MEDLINE | ID: mdl-32143748

In bilaterally-symmetric animals (Bilateria), condensation of neurons and ganglia into a centralized nervous system (CNS) constitutes a salient feature. In most, if not all, Bilateria another prominent aspect is that the anterior regions of the CNS are typically larger than the posterior ones. Detailed studies in Drosophila melanogaster (Drosophila) have revealed that anterior expansion in this species stems from three major developmental features: the generation of more progenitors anteriorly, an extended phase of proliferation of anterior progenitors, and more proliferative daughter cells in anterior regions. These brain-specific features combine to generate a larger average lineage size and higher cell numbers in the brain, when compared to more posterior regions. Genetic studies reveal that these anterior-posterior (A-P) differences are controlled by the modulation of temporal programs, common to all progenitors, as well as by Hox homeotic genes, expressed in the nerve cord, and brain-specific factors. All of these regulatory features are gated by the action of the PRC2 epigenetic complex. Studies in mammals indicate that most, if not all of these anterior expansion principles and the underlying genetic programs are evolutionarily conserved. These findings further lend support for the recently proposed idea that the brain and nerve cord may have originated from different parts of the nervous system present in the Bilaterian ancestor. This brain-nerve cord "fusion" concept may help explain a number of the well-known fundamental differences in the biology of the brain, when compared to the nerve cord.


Central Nervous System/cytology , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Embryo, Nonmammalian/cytology , Gene Expression Regulation, Developmental , Nervous System Physiological Phenomena , Animals , Cell Proliferation , Central Nervous System/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Embryo, Nonmammalian/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
14.
Adv Exp Med Biol ; 1218: 39-58, 2020.
Article En | MEDLINE | ID: mdl-32060870

During central nervous system (CNS) development, a complex series of events play out, starting with the establishment of neural progenitor cells, followed by their asymmetric division and formation of lineages and the differentiation of neurons and glia. Studies in the Drosophila melanogaster embryonic CNS have revealed that the Notch signal transduction pathway plays at least five different and distinct roles during these events. Herein, we review these many faces of Notch signalling and discuss the mechanisms that ensure context-dependent and compartment-dependent signalling. We conclude by discussing some outstanding issues regarding Notch signalling in this system, which likely have bearing on Notch signalling in many species.


Central Nervous System/embryology , Central Nervous System/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals
15.
Cell Rep ; 29(11): 3636-3651.e3, 2019 Dec 10.
Article En | MEDLINE | ID: mdl-31825841

In the developing Drosophila central nervous system (CNS), neural progenitor (neuroblast [NB]) selection is gated by lateral inhibition, controlled by Notch signaling and proneural genes. However, proneural mutants still generate many NBs, indicating the existence of additional proneural genes. Moreover, recent studies reveal involvement of key epithelial-mesenchymal transition (EMT) genes in NB selection, but the regulatory interplay between Notch signaling and the EMT machinery is unclear. We find that SoxNeuro (SoxB family) and worniu (Snail family) are integrated with the Notch pathway, and constitute the missing proneural genes. Notch signaling, the proneural, SoxNeuro, and worniu genes regulate key EMT genes to orchestrate the NB selection process. Hence, we uncover an expanded lateral inhibition network for NB selection and demonstrate its link to key players in the EMT machinery. The evolutionary conservation of the genes involved suggests that the Notch-SoxB-Snail-EMT network may control neural progenitor selection in many other systems.


Drosophila Proteins/metabolism , Epithelial-Mesenchymal Transition , Neural Stem Cells/metabolism , Receptors, Notch/metabolism , SOX Transcription Factors/metabolism , Transcription Factors/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neural Stem Cells/cytology , Neurogenesis , Receptors, Notch/genetics , SOX Transcription Factors/genetics , Signal Transduction , Transcription Factors/genetics
17.
Elife ; 82019 07 04.
Article En | MEDLINE | ID: mdl-31271353

During CNS development, there is prominent expansion of the anterior region, the brain. In Drosophila, anterior CNS expansion emerges from three rostral features: (1) increased progenitor cell generation, (2) extended progenitor cell proliferation, (3) more proliferative daughters. We find that tailless (mouse Nr2E1/Tlx), otp/Rx/hbn (Otp/Arx/Rax) and Doc1/2/3 (Tbx2/3/6) are important for brain progenitor generation. These genes, and earmuff (FezF1/2), are also important for subsequent progenitor and/or daughter cell proliferation in the brain. Brain TF co-misexpression can drive brain-profile proliferation in the nerve cord, and can reprogram developing wing discs into brain neural progenitors. Brain TF expression is promoted by the PRC2 complex, acting to keep the brain free of anti-proliferative and repressive action of Hox homeotic genes. Hence, anterior expansion of the Drosophila CNS is mediated by brain TF driven 'super-generation' of progenitors, as well as 'hyper-proliferation' of progenitor and daughter cells, promoted by PRC2-mediated repression of Hox activity.


Brain/embryology , Cell Proliferation , Drosophila/embryology , Gene Expression Regulation, Developmental , Stem Cells/physiology , Transcription Factors/metabolism , Animals
18.
Development ; 146(13)2019 07 09.
Article En | MEDLINE | ID: mdl-31289041

During cell cycle progression, the activity of the CycE-Cdk2 complex gates S-phase entry. CycE-Cdk2 is inhibited by CDK inhibitors (CKIs) of the Cip/Kip family, which include the human p21Cip1 and Drosophila Dacapo (Dap) proteins. Both the CycE and Cip/Kip family proteins are under elaborate control via protein degradation, mediated by the Cullin-RING ligase (CRL) family of ubiquitin ligase complexes. The CRL complex SCFFbxw7/Ago targets phosphorylated CycE, whereas p21Cip1 and Dap are targeted by the CRL4Cdt2 complex, binding to the PIP degron. The role of CRL-mediated degradation of CycE and Cip/Kip proteins during CNS development is not well understood. Here, we analyse the role of ago (Fbxw7)-mediated CycE degradation, and of Dap and p21Cip1 degradation during Drosophila CNS development. We find that ago mutants display over-proliferation, accompanied by elevated CycE expression levels. By contrast, expression of PIP degron mutant Dap and p21Cip1 transgenes inhibit proliferation. However, surprisingly, this is also accompanied by elevated CycE levels. Hence, ago mutation and PIP degron Cip/Kip transgenic expression trigger opposite effects on proliferation, but similar effects on CycE levels.


Cell Proliferation/genetics , Cyclin E/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , F-Box Proteins/genetics , Mutation , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Animals , Animals, Genetically Modified , Central Nervous System/cytology , Central Nervous System/embryology , Cyclin E/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/physiology , Drosophila melanogaster , Embryo, Mammalian , F-Box Proteins/physiology , Mutation/physiology , Nuclear Proteins/chemistry , Peptide Fragments/chemistry , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Protein Interaction Domains and Motifs/physiology , Protein Stability
19.
Development ; 146(6)2019 03 15.
Article En | MEDLINE | ID: mdl-30837222

The nervous system displays a daunting cellular diversity. Neuronal subtypes differ from each other in several aspects, including their neurotransmitter expression and axon projection. These aspects can converge, but can also diverge, such that neurons expressing the same neurotransmitter may project axons to different targets. It is not well understood how regulatory programs converge/diverge to associate/dissociate different cell fate features. Studies of the Drosophila Tv1 neurons have identified a regulatory cascade, ladybird early→collier→apterous/eyes absent→dimmed, that specifies Tv1 neurotransmitter expression. Here, we conduct genetic and transcriptome analysis to address how other aspects of Tv1 cell fate are governed. We find that an initiator terminal selector gene triggers a feedforward loop that branches into different subroutines, each of which establishes different features of this one unique neuronal cell fate.


Drosophila melanogaster/genetics , Gene Regulatory Networks , Neurons/cytology , Animals , Axons/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Lineage , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/genetics , Microscopy, Confocal , Neurotransmitter Agents/genetics , Sequence Analysis, RNA , Signal Transduction , Transcription Factors/genetics , Transcriptome
20.
PLoS Biol ; 17(2): e3000163, 2019 02.
Article En | MEDLINE | ID: mdl-30807568

During central nervous system (CNS) development, genetic programs establish neural stem cells and drive both stem and daughter cell proliferation. However, the prominent anterior expansion of the CNS implies anterior-posterior (A-P) modulation of these programs. In Drosophila, a set of neural stem cell factors acts along the entire A-P axis to establish neural stem cells. Brain expansion results from enhanced stem and daughter cell proliferation, promoted by a Polycomb Group (PcG)->Homeobox (Hox) homeotic network. But how does PcG->Hox modulate neural-stem-cell-factor activity along the A-P axis? We find that the PcG->Hox network creates an A-P expression gradient of neural stem cell factors, thereby driving a gradient of proliferation. PcG mutants can be rescued by misexpression of the neural stem cell factors or by mutation of one single Hox gene. Hence, brain expansion results from anterior enhancement of core neural-stem-cell-factor expression, mediated by PcG repression of brain Hox expression.


Brain/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Neural Stem Cells/metabolism , Polycomb-Group Proteins/genetics , Stem Cell Factor/genetics , Animals , Brain/growth & development , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/metabolism , Mutation , Neural Stem Cells/cytology , Neurogenesis/genetics , Polycomb-Group Proteins/metabolism , Signal Transduction , Stem Cell Factor/metabolism
...