Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
MicroPubl Biol ; 20232023.
Article in English | MEDLINE | ID: mdl-38094098

ABSTRACT

CUTie2 is a FRET-based cGMP biosensor tested so far only in cells. To expand its use to multicellular organisms we generated two transgenic Drosophila melanogaster strains that express the biosensor in a tissue-dependent manner. CUTie2 expression and subcellular localization was verified by confocal microscopy. The performance of CUTie2 was analyzed on dissected larval brains by hyperspectral microscopy and flow cytometry. Both approaches confirmed its responsivity, and the latter showed a rapid and reversible change in the fluorescence of the FRET acceptor upon cGMP treatment. This validated reporter system may prove valuable for studying cGMP signaling at organismal level.

2.
PLoS Genet ; 16(11): e1008913, 2020 11.
Article in English | MEDLINE | ID: mdl-33211700

ABSTRACT

Mitotic divisions depend on the timely assembly and proper orientation of the mitotic spindle. Malfunctioning of these processes can considerably delay mitosis, thereby compromising tissue growth and homeostasis, and leading to chromosomal instability. Loss of functional Mms19 drastically affects the growth and development of mitotic tissues in Drosophila larvae and we now demonstrate that Mms19 is an important factor that promotes spindle and astral microtubule (MT) growth, and MT stability and bundling. Mms19 function is needed for the coordination of mitotic events and for the rapid progression through mitosis that is characteristic of neural stem cells. Surprisingly, Mms19 performs its mitotic activities through two different pathways. By stimulating the mitotic kinase cascade, it triggers the localization of the MT regulatory complex TACC/Msps (Transforming Acidic Coiled Coil/Minispindles, the homolog of human ch-TOG) to the centrosome. This activity of Mms19 can be rescued by stimulating the mitotic kinase cascade. However, other aspects of the Mms19 phenotypes cannot be rescued in this way, pointing to an additional mechanism of Mms19 action. We provide evidence that Mms19 binds directly to MTs and that this stimulates MT stability and bundling.


Subject(s)
Drosophila Proteins/metabolism , Microtubules/metabolism , Neural Stem Cells/metabolism , Spindle Apparatus/metabolism , Animals , Cell Cycle/physiology , Centrosome/metabolism , Drosophila melanogaster , Microtubules/physiology , Mitosis/physiology , Neural Stem Cells/physiology , Spindle Apparatus/genetics , Spindle Poles/genetics , Spindle Poles/metabolism , Transcription Factors/metabolism
3.
Biol Open ; 9(8)2020 08 18.
Article in English | MEDLINE | ID: mdl-32816692

ABSTRACT

Environmental factors such as the availability of oxygen are instructive cues that regulate stem cell maintenance and differentiation. We used a genetically encoded biosensor to monitor the hypoxic state of neural cells in the larval brain of Drosophila The biosensor reveals brain compartment and cell-type specific levels of hypoxia. The values correlate with differential tracheolation that is observed throughout development between the central brain and the optic lobe. Neural stem cells in both compartments show the strongest hypoxia response while intermediate progenitors, neurons and glial cells reveal weaker responses. We demonstrate that the distance between a cell and the next closest tracheole is a good predictor of the hypoxic state of that cell. Our study indicates that oxygen availability appears to be the major factor controlling the hypoxia response in the developing Drosophila brain and that cell intrinsic and cell-type specific factors contribute to modulate the response in an unexpected manner.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Brain/growth & development , Brain/pathology , Cell Compartmentation , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Hypoxia/pathology , Animals , Biosensing Techniques , Cell Differentiation , Cell Hypoxia/drug effects , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Glycolysis/genetics , Green Fluorescent Proteins/metabolism , Hypoxia/genetics , Larva/drug effects , Neuroepithelial Cells/drug effects , Neuroepithelial Cells/metabolism , Optic Lobe, Nonmammalian/pathology , Oxygen/pharmacology
4.
Methods Mol Biol ; 2047: 153-160, 2020.
Article in English | MEDLINE | ID: mdl-31552654

ABSTRACT

Live cell imaging gives valuable insights into the dynamic biological processes within and between cells. An important aspect of live cell imaging is to keep the cells under best physiological condition and to prevent abnormal cellular behavior, which might be caused by phototoxicity during microscopy. In this chapter we describe a protocol to visualize division patterns of neural stem cells in live whole mount brains of Drosophila larvae. We also present a newly developed live cell chamber that allows us to control the environmental air during live cell imaging. The protocol can be adapted to look at a wide range of cellular and tissue behavior in the Drosophila model system.


Subject(s)
Brain/cytology , Brain/physiology , Drosophila/cytology , Drosophila/physiology , Embryo, Nonmammalian/cytology , Animals , Brain/metabolism , Drosophila/metabolism , Drosophila melanogaster , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/physiology , In Situ Hybridization , Larva/cytology , Larva/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism
5.
Cell Rep ; 27(11): 3152-3166.e7, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31189102

ABSTRACT

After a peripheral nerve lesion, distal ends of injured axons disintegrate into small fragments that are subsequently cleared by Schwann cells and later by macrophages. Axonal debris clearing is an early step of the repair process that facilitates regeneration. We show here that Schwann cells promote distal cut axon disintegration for timely clearing. By combining cell-based and in vivo models of nerve lesion with mouse genetics, we show that this mechanism is induced by distal cut axons, which signal to Schwann cells through PlGF mediating the activation and upregulation of VEGFR1 in Schwann cells. In turn, VEGFR1 activates Pak1, leading to the formation of constricting actomyosin spheres along unfragmented distal cut axons to mediate their disintegration. Interestingly, oligodendrocytes can acquire a similar behavior as Schwann cells by enforced expression of VEGFR1. These results thus identify controllable molecular cues of a neuron-glia crosstalk essential for timely clearing of damaged axons.


Subject(s)
Actins/metabolism , Axons/metabolism , Peripheral Nerve Injuries/metabolism , Schwann Cells/metabolism , Animals , Cell Line , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Oligodendroglia/metabolism , Placenta Growth Factor/genetics , Placenta Growth Factor/metabolism , Rats , Rats, Wistar , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
6.
Neural Dev ; 13(1): 25, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30466475

ABSTRACT

BACKGROUND: Neural stem cells generate all of the neurons and glial cells in the central nervous system, both during development and in the adult to maintain homeostasis. In the Drosophila optic lobe, neuroepithelial cells progress through two transient progenitor states, PI and PII, before transforming into neuroblasts. Here we analyse the role of Notch signalling in the transition from neuroepithelial cells to neuroblasts. RESULTS: We observed dynamic regulation of Notch signalling: strong activity in PI progenitors, low signalling in PII progenitors, and increased activity after neuroblast transformation. Ectopic expression of the Notch ligand Delta induced the formation of ectopic PI progenitors. Interestingly, we show that the E3 ubiquitin ligase, Neuralized, regulates Delta levels and Notch signalling activity at the transition zone. We demonstrate that the proneural transcription factor, Lethal of scute, is essential to induce expression of Neuralized and promote the transition from the PI progenitor to the PII progenitor state. CONCLUSIONS: Our results show dynamic regulation of Notch signalling activity in the transition from neuroepithelial cells to neuroblasts. We propose a model in which Lethal of scute activates Notch signalling in a non-cell autonomous manner by regulating the expression of Neuralized, thereby promoting the progression between different neural stem cell states.


Subject(s)
Drosophila Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Optic Lobe, Nonmammalian/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Drosophila , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neural Stem Cells/cytology , Neuroepithelial Cells/cytology , Neuroepithelial Cells/metabolism , Neurons/cytology , Neurons/metabolism , Receptors, Notch/genetics
7.
Elife ; 72018 10 22.
Article in English | MEDLINE | ID: mdl-30346271

ABSTRACT

Lasting changes in gene expression are critical for the formation of long-term memories (LTMs), depending on the conserved CrebB transcriptional activator. While requirement of distinct neurons in defined circuits for different learning and memory phases have been studied in detail, only little is known regarding the gene regulatory changes that occur within these neurons. We here use the fruit fly as powerful model system to study the neural circuits of CrebB-dependent appetitive olfactory LTM. We edited the CrebB locus to create a GFP-tagged CrebB conditional knockout allele, allowing us to generate mutant, post-mitotic neurons with high spatial and temporal precision. Investigating CrebB-dependence within the mushroom body (MB) circuit we show that MB α/ß and α'/ß' neurons as well as MBON α3, but not in dopaminergic neurons require CrebB for LTM. Thus, transcriptional memory traces occur in different neurons within the same neural circuit.


Subject(s)
Appetite/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Mushroom Bodies/innervation , Mushroom Bodies/metabolism , Neurons/metabolism , Trans-Activators/metabolism , Alleles , Animals , Gene Knockout Techniques , Memory, Long-Term , Reproducibility of Results
8.
PLoS Genet ; 14(4): e1007353, 2018 04.
Article in English | MEDLINE | ID: mdl-29677185

ABSTRACT

The central nervous system develops from monolayered neuroepithelial sheets. In a first step patterning mechanisms subdivide the seemingly uniform epithelia into domains allowing an increase of neuronal diversity in a tightly controlled spatial and temporal manner. In Drosophila, neuroepithelial patterning of the embryonic optic placode gives rise to the larval eye primordium, consisting of two photoreceptor (PR) precursor types (primary and secondary), as well as the optic lobe primordium, which during larval and pupal stages develops into the prominent optic ganglia. Here, we characterize a genetic network that regulates the balance between larval eye and optic lobe precursors, as well as between primary and secondary PR precursors. In a first step the proneural factor Atonal (Ato) specifies larval eye precursors, while the orphan nuclear receptor Tailless (Tll) is crucial for the specification of optic lobe precursors. The Hedgehog and Notch signaling pathways act upstream of Ato and Tll to coordinate neural precursor specification in a timely manner. The correct spatial placement of the boundary between Ato and Tll in turn is required to control the precise number of primary and secondary PR precursors. In a second step, Notch signaling also controls a binary cell fate decision, thus, acts at the top of a cascade of transcription factor interactions to define PR subtype identity. Our model serves as an example of how combinatorial action of cell extrinsic and cell intrinsic factors control neural tissue patterning.


Subject(s)
Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Eye/growth & development , Eye/metabolism , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Genes, Insect , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroepithelial Cells/metabolism , Optic Lobe, Nonmammalian/growth & development , Optic Lobe, Nonmammalian/metabolism , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction
9.
Biol Open ; 6(2): 296-304, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28011628

ABSTRACT

Cells experience different oxygen concentrations depending on location, organismal developmental stage, and physiological or pathological conditions. Responses to reduced oxygen levels (hypoxia) rely on the conserved hypoxia-inducible factor 1 (HIF-1). Understanding the developmental and tissue-specific responses to changing oxygen levels has been limited by the lack of adequate tools for monitoring HIF-1 in vivo. To visualise and analyse HIF-1 dynamics in Drosophila, we used a hypoxia biosensor consisting of GFP fused to the oxygen-dependent degradation domain (ODD) of the HIF-1 homologue Sima. GFP-ODD responds to changing oxygen levels and to genetic manipulations of the hypoxia pathway, reflecting oxygen-dependent regulation of HIF-1 at the single-cell level. Ratiometric imaging of GFP-ODD and a red-fluorescent reference protein reveals tissue-specific differences in the cellular hypoxic status at ambient normoxia. Strikingly, cells in the larval brain show distinct hypoxic states that correlate with the distribution and relative densities of respiratory tubes. We present a set of genetic and image analysis tools that enable new approaches to map hypoxic microenvironments, to probe effects of perturbations on hypoxic signalling, and to identify new regulators of the hypoxia response.

10.
Cell Rep ; 14(5): 1100-1113, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26804909

ABSTRACT

Centrosome asymmetry has been implicated in stem cell fate maintenance in both flies and vertebrates, but the underlying molecular mechanisms are incompletely understood. Here, we report that loss of CG7337, the fly ortholog of WDR62, compromises interphase centrosome asymmetry in fly neural stem cells (neuroblasts). Wdr62 maintains an active interphase microtubule-organizing center (MTOC) by stabilizing microtubules (MTs), which are necessary for sustained recruitment of Polo/Plk1 to the pericentriolar matrix (PCM) and downregulation of Pericentrin-like protein (Plp). The loss of an active MTOC in wdr62 mutants compromises centrosome positioning, spindle orientation, and biased centrosome segregation. wdr62 mutant flies also have an ∼40% reduction in brain size as a result of cell-cycle delays. We propose that CG7337/Wdr62, a microtubule-associated protein, is required for the maintenance of interphase microtubules, thereby regulating centrosomal Polo and Plp levels. Independent of this function, Wdr62 is also required for the timely mitotic entry of neural stem cells.


Subject(s)
Centrosome/metabolism , Drosophila melanogaster/metabolism , Microcephaly/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Drosophila Proteins/metabolism , Humans , Interphase , Microtubule-Organizing Center/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Sequence Homology, Amino Acid , Spindle Apparatus/metabolism , Polo-Like Kinase 1
11.
PLoS Biol ; 13(9): e1002258, 2015.
Article in English | MEDLINE | ID: mdl-26406915

ABSTRACT

The pathogenesis of peripheral neuropathies in adults is linked to maintenance mechanisms that are not well understood. Here, we elucidate a novel critical maintenance mechanism for Schwann cell (SC)-axon interaction. Using mouse genetics, ablation of the transcriptional regulators histone deacetylases 1 and 2 (HDAC1/2) in adult SCs severely affected paranodal and nodal integrity and led to demyelination/remyelination. Expression levels of the HDAC1/2 target gene myelin protein zero (P0) were reduced by half, accompanied by altered localization and stability of neurofascin (NFasc)155, NFasc186, and loss of Caspr and septate-like junctions. We identify P0 as a novel binding partner of NFasc155 and NFasc186, both in vivo and by in vitro adhesion assay. Furthermore, we demonstrate that HDAC1/2-dependent P0 expression is crucial for the maintenance of paranodal/nodal integrity and axonal function through interaction of P0 with neurofascins. In addition, we show that the latter mechanism is impaired by some P0 mutations that lead to late onset Charcot-Marie-Tooth disease.


Subject(s)
Cell Adhesion Molecules/metabolism , Charcot-Marie-Tooth Disease/genetics , Myelin P0 Protein/genetics , Myelin Sheath/physiology , Nerve Growth Factors/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Charcot-Marie-Tooth Disease/enzymology , Gene Knockout Techniques , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Mice
12.
Dev Biol ; 405(2): 202-13, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26111972

ABSTRACT

Brain development goes through phases of proliferative growth and differentiation to ensure the formation of correct number and variety of neurons. How and when naïve neuroepithelial cells decide to enter a differentiation pathway remains poorly understood. In the Drosophila visual system, four optic ganglia emerge from neuroepithelia of the inner (IPC) and outer (OPC) proliferation centers. Here we demonstrate that the orphan nuclear receptor Tailless (Tll) is a key factor for the development of all optic ganglia. We describe tll expression during larval optic lobe development in unprecedented detail and find a spatiotemporally dynamic pattern. In the larval OPC, symmetrically dividing neuroepithelial cells transform into asymmetrically dividing medulla neuroblast and into lamina precursor cells in a precisely regulated fashion. Using genetic manipulations we found that tll is required for proper neuroepithelium morphology and neuroepithelial cell survival. We show that tll regulates the precise timing of the transition from neuroepithelial cells to medulla neuroblasts. In particular, however, we demonstrate that tll has a crucial role for the specification of lamina precursor cells. We propose that the Tll/Tlx transcription factors have an evolutionary conserved role in regulating neural precursor cell states in the Drosophila optic lobe and in the mammalian retina.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Optic Lobe, Nonmammalian/physiology , Repressor Proteins/physiology , Animals , Cell Proliferation , Cell Survival , Crosses, Genetic , Epithelial Cells/cytology , Green Fluorescent Proteins/metabolism , Neural Stem Cells/cytology , Neuroepithelial Cells/cytology , Neurons/cytology , Receptors, Notch/metabolism , Retina/embryology , Stem Cells/cytology
13.
Methods Mol Biol ; 1082: 71-8, 2014.
Article in English | MEDLINE | ID: mdl-24048927

ABSTRACT

The Drosophila visual system is an excellent model system to study the switch from proliferating to differentiating neural stem cells. In the developing larval optic lobe, symmetrically dividing neuroepithelial cells transform to asymmetrically dividing neuroblasts in a highly ordered and sequential manner. This chapter presents a protocol to visualize neural stem cell types in the Drosophila optic lobe by fluorescence confocal microscopy. A main focus is given on how to dissect, fix, immunolabel, and mount brains to reveal cellular morphology during early larval brain development.


Subject(s)
Drosophila melanogaster/cytology , Fluorescent Antibody Technique/methods , Neural Stem Cells/cytology , Optic Lobe, Nonmammalian/cytology , Animals , Brain/cytology , Female , Genotype , Neural Stem Cells/metabolism , Polylysine/metabolism , Time Factors , Tissue Fixation
14.
Dev Cell ; 26(1): 101-12, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23792147

ABSTRACT

Cell-type-specific transcriptional profiling often requires the isolation of specific cell types from complex tissues. We have developed "TaDa," a technique that enables cell-specific profiling without cell isolation. TaDa permits genome-wide profiling of DNA- or chromatin-binding proteins without cell sorting, fixation, or affinity purification. The method is simple, sensitive, highly reproducible, and transferable to any model system. We show that TaDa can be used to identify transcribed genes in a cell-type-specific manner with considerable temporal precision, enabling the identification of differential gene expression between neuroblasts and the neuroepithelial cells from which they derive. We profile the genome-wide binding of RNA polymerase II in these adjacent, clonally related stem cells within intact Drosophila brains. Our data reveal expression of specific metabolic genes in neuroepithelial cells, but not in neuroblasts, and highlight gene regulatory networks that may pattern neural stem cell fates.


Subject(s)
Brain/metabolism , Chromatin/metabolism , Gene Expression Profiling/methods , Neural Stem Cells/enzymology , RNA Polymerase II/analysis , Animals , Brain/cytology , Cell Separation , Chromatin/genetics , DNA Methylation , Drosophila/enzymology , Drosophila/genetics , Gene Regulatory Networks , Genes, Insect , Neural Stem Cells/cytology , Neuroepithelial Cells/cytology , Neuroepithelial Cells/enzymology , Protein Binding , RNA Polymerase II/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Reproducibility of Results , Sensitivity and Specificity , Transcription, Genetic
15.
Nat Protoc ; 8(5): 958-65, 2013 May.
Article in English | MEDLINE | ID: mdl-23598446

ABSTRACT

Cell culture systems are widely used for molecular, genetic and biochemical studies. Primary cell cultures of animal tissues offer the advantage that specific cell types can be studied in vitro outside of their normal environment. We provide a detailed protocol for generating primary neural cell cultures derived from larval brains of Drosophila melanogaster. The developing larval brain contains stem cells such as neural precursors and intermediate neural progenitors, as well as fully differentiated and functional neurons and glia cells. We describe how to analyze these cell types in vitro by immunofluorescent staining and scanning confocal microscopy. Cell type-specific fluorescent reporter lines and genetically encoded calcium sensors allow the monitoring of developmental, cellular processes and neuronal activity in living cells in vitro. The protocol provides a basis for functional studies of wild-type or genetically manipulated primary neural cells in culture, both in fixed and living samples. The entire procedure takes ∼3 weeks.


Subject(s)
Cell Culture Techniques , Drosophila melanogaster/cytology , Embryo, Nonmammalian/cytology , Neurons/cytology , Animals , Drosophila melanogaster/embryology , Larva/cytology , Microscopy, Fluorescence
16.
Neural Dev ; 7: 14, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22554060

ABSTRACT

In Drosophila, most neurogenetic research is carried out in vivo. Mammalian research demonstrates that primary cell culture techniques provide a powerful model to address cell autonomous and non-autonomous processes outside their endogenous environment. We developed a cell culture system in Drosophila using wildtype and genetically manipulated primary neural tissue for long-term observations. We assessed the molecular identity of distinct neural cell types by immunolabeling and genetically expressed fluorescent cell markers. We monitored mitotic activity of cell cultures derived from wildtype and tumorous larval brains. Our system provides a powerful approach to unveil developmental processes in the nervous system and to complement studies in vivo.


Subject(s)
Apoptosis/physiology , Cell Culture Techniques , Cell Shape/physiology , Drosophila melanogaster/cytology , Mitosis/physiology , Neurons/cytology , Animals , Cell Culture Techniques/methods , Cell Differentiation/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Larva/cytology , Larva/genetics , Larva/metabolism , Neurons/metabolism , Primary Cell Culture
17.
Fly (Austin) ; 5(3): 237-41, 2011.
Article in English | MEDLINE | ID: mdl-21502820

ABSTRACT

Stem cells proliferate through symmetric division or self-renew through asymmetric division whilst generating differentiating cell types. The balance between symmetric and asymmetric division requires tight control to either expand a stem cell pool or to generate cell diversity. In the Drosophila optic lobe, symmetrically dividing neuroepithelial cells transform into asymmetrically dividing neuroblasts. The switch from neuroepithelial cells to neuroblasts is triggered by a proneural wave that sweeps across the neuroepithelium. Here we review recent findings showing that the orchestrated action of the Notch, EGFR, Fat-Hippo, and JAK/STAT signalling pathways controls the progression of the proneural wave and the sequential transition from symmetric to asymmetric division. The neuroepithelial to neuroblast transition in the optic lobe bears many similarities to the switch from neuroepithelial cell to radial glial cell in the developing mammalian cerebral cortex. The Notch signalling pathway has a similar role in the transition from proliferating to differentiating stem cell pools in the developing vertebrate retina and in the neural tube. Therefore, findings in the Drosophila optic lobe provide insights into the transitions between proliferative and differentiative division in the stem cell pools of higher organisms.


Subject(s)
Cell Division , Drosophila/growth & development , Neural Stem Cells/physiology , Optic Lobe, Nonmammalian/growth & development , Animals , Cell Differentiation , Drosophila/cytology , Optic Lobe, Nonmammalian/cytology , Signal Transduction
18.
Development ; 137(18): 2981-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20685734

ABSTRACT

The proper balance between symmetric and asymmetric stem cell division is crucial both to maintain a population of stem cells and to prevent tumorous overgrowth. Neural stem cells in the Drosophila optic lobe originate within a polarised neuroepithelium, where they divide symmetrically. Neuroepithelial cells are transformed into asymmetrically dividing neuroblasts in a precisely regulated fashion. This cell fate transition is highly reminiscent of the switch from neuroepithelial cells to radial glial cells in the developing mammalian cerebral cortex. To identify the molecules that mediate the transition, we microdissected neuroepithelial cells and compared their transcriptional profile with similarly obtained optic lobe neuroblasts. We find genes encoding members of the Notch pathway expressed in neuroepithelial cells. We show that Notch mutant clones are extruded from the neuroepithelium and undergo premature neurogenesis. A wave of proneural gene expression is thought to regulate the timing of the transition from neuroepithelium to neuroblast. We show that the proneural wave transiently suppresses Notch activity in neuroepithelial cells, and that inhibition of Notch triggers the switch from symmetric, proliferative division, to asymmetric, differentiative division.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Neurons/metabolism , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/metabolism , Receptors, Notch/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Division , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Mutation , Receptors, Notch/genetics , Signal Transduction
19.
Neural Dev ; 4: 9, 2009 Mar 02.
Article in English | MEDLINE | ID: mdl-19254370

ABSTRACT

BACKGROUND: The production of new neurons during adulthood and their subsequent integration into a mature central nervous system have been shown to occur in all vertebrate species examined to date. However, the situation in insects is less clear and, in particular, it has been reported that there is no proliferation in the Drosophila adult brain. RESULTS: We report here, using clonal analysis and 5'-bromo-2'-deoxyuridine (BrdU) labelling, that cell proliferation does occur in the Drosophila adult brain. The majority of clones cluster on the ventrolateral side of the antennal lobes, as do the BrdU-positive cells. Of the BrdU-labelled cells, 86% express the glial gene reversed polarity (repo), and 14% are repo negative. CONCLUSION: We have observed cell proliferation in the Drosophila adult brain. The dividing cells may be adult stem cells, generating glial and/or non-glial cell types.


Subject(s)
Brain/cytology , Bromodeoxyuridine/chemistry , Cell Proliferation , Neuroglia/metabolism , Neurons/metabolism , Animals , Brain/anatomy & histology , Cell Count/methods , Drosophila , Immunohistochemistry
20.
Semin Cell Dev Biol ; 19(3): 283-93, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18328747

ABSTRACT

Asymmetric cell division is an important and conserved strategy in the generation of cellular diversity during animal development. Many of our insights into the underlying mechanisms of asymmetric cell division have been gained from Drosophila, including the establishment of polarity, orientation of mitotic spindles and segregation of cell fate determinants. Recent studies are also beginning to reveal the connection between the misregulation of asymmetric cell division and cancer. What we are learning from Drosophila as a model system has implication both for stem cell biology and also cancer research.


Subject(s)
Cell Division , Drosophila melanogaster/cytology , Stem Cells/cytology , Animals , Cell Lineage , Neoplasms/pathology , Spindle Apparatus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...