Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurosci Res ; 65(1): 17-23, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11433425

RESUMO

The aim of our study was to investigate whether a human neural cell line could be used as a reliable screening tool to examine the functional conservation, in humans, of transcription factors involved in neuronal or glial specification in other species. Gain-of-function experiments were performed on DEV cells, a cell line derived from a human medulloblastoma. Genes encoding nine different transcription factors were tested for their influence on the process of specification of human DEV cells towards a neuronal or glial fate. In a first series of experiments, DEV cells were transfected with murine genes encoding transcription factors known to be involved in the neuronal differentiation cascade. Neurogenins-1, -2, and -3; Mash-1; and NeuroD increased the differentiation of DEV cells towards a neuronal phenotype by a factor of 2-3.5. In a second series of experiments, we tested transcription factors involved in invertebrate glial specification. In the embryonic Drosophila CNS, the development of most glial cells depends on the master regulatory gene glial cell missing (gcm). Expression of gcm in DEV cells induced a twofold increase of astrocytic and a sixfold increase of oligodendroglial cell types. Interestingly, expression of tramtrack69, which is required in all Drosophila glial cells, resulted in a fourfold increase of only the oligodendrocyte phenotype. Expression of the related tramtrack88 protein, which is not expressed in the fly glia, or the C. elegans lin26 protein showed no effect. These results show that the Drosophila transcription factor genes tested can conserve their function upon transfection into the human DEV cells, qualifying this cell line as a screening tool to analyze the mechanisms of neuronal and glial specification.


Assuntos
Proteínas de Caenorhabditis elegans , Neoplasias Cerebelares , Proteínas de Drosophila , Meduloblastoma , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Células-Tronco/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Caenorhabditis elegans , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Meios de Cultura/farmacologia , Proteínas de Ligação a DNA/genética , Drosophila , Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde , Humanos , Indicadores e Reagentes/metabolismo , Proteínas Luminescentes/genética , Neuroglia/citologia , Neuropeptídeos/genética , Proteínas Repressoras/genética , Transativadores/genética , Fatores de Transcrição/genética , Transfecção , Células Tumorais Cultivadas
2.
Mech Dev ; 106(1-2): 25-36, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11472832

RESUMO

Axonal growth cones require an evolutionary conserved repulsive guidance system to ensure proper crossing of the CNS midline. In Drosophila, the Slit protein is a repulsive signal secreted by the midline glial cells. It binds to the Roundabout receptors, which are expressed on CNS axons in the longitudinal tracts but not in the commissural tracts. Here we present an analysis of the genes leak and kuzbanian and show that both genes are involved in the repulsive guidance system operating at the CNS midline. Mutations in leak, which encodes the Roundabout-2 Slit receptor, were first recovered by Nüsslein-Volhard and co-workers based on defects in the larval cuticle. Analysis of the head phenotype suggests that slit may be able to act as an attractive guidance cue while directing the movements of the dorsal ectodermal cell sheath. kuzbanian also regulates midline crossing of CNS axons. It encodes a metalloprotease of the ADAM family and genetically interacts with slit. Expression of a dominant negative Kuzbanian protein in the CNS midline cells results in an abnormal midline crossing of axons and prevents the clearance of the Roundabout receptor from commissural axons. Our analyses support a model in which Kuzbanian mediates the proteolytic activation of the Slit/Roundabout receptor complex.


Assuntos
Movimento Celular , Desintegrinas/fisiologia , Proteínas de Drosophila , Drosophila/embriologia , Ectoderma/citologia , Cones de Crescimento/fisiologia , Metaloendopeptidases/fisiologia , Animais , Axônios/fisiologia , Moléculas de Adesão Celular Neuronais/metabolismo , Sistema Nervoso Central/embriologia , Desintegrinas/genética , Drosophila/genética , Ectoderma/metabolismo , Desenvolvimento Embrionário , Genes de Insetos , Teste de Complementação Genética , Cabeça/embriologia , Metaloendopeptidases/genética , Modelos Biológicos , Morfogênese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Receptores Imunológicos/genética , Receptores Imunológicos/fisiologia , Proteínas Roundabout
3.
Trends Neurosci ; 24(8): 432-3, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11476870

RESUMO

A key problem in developmental neurobiology is how axons home in on their correct target tissue and establish the correct synaptic contacts. Recent work shows that in the developing Drosophila visual system a population of distinct lamina glial cells ensures correct target layer selection of retinal axons. In the absence of lamina neurons, photoreceptor axons terminate their growth in the correct zone, but when glial cell migration into the lamina is disrupted, as in nonstop mutants, growth cones advance into deeper layers of the brain.


Assuntos
Axônios/fisiologia , Movimento Celular/fisiologia , Neuroglia/citologia , Vias Visuais/citologia , Vias Visuais/embriologia , Animais , Drosophila , Células Fotorreceptoras de Invertebrados/citologia
4.
Int J Dev Neurosci ; 19(4): 373-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11378297

RESUMO

In the Drosophila central nervous system (CNS) about 10% of the cells are of glial nature. A set of molecular markers has allowed unraveling a number of genes controlling glial cell fate determination as well as genes required for glial cell differentiation. Here we focus on the embryonic CNS glia and review the recent progress in the field.


Assuntos
Drosophila melanogaster/citologia , Neuroglia/citologia , Animais , Biomarcadores , Diferenciação Celular , Linhagem da Célula , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Proteínas de Insetos/genética , Proteínas de Insetos/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Neuroglia/classificação , Neuroglia/fisiologia , Transdução de Sinais
6.
Mech Dev ; 100(2): 339-42, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11165494

RESUMO

Here we describe of a novel Drosophila LTR-type retrotransposon that is expressed in the embryonic CNS midline glia and in the embryonic germ cells. The element is related to the gypsy and burdock retrotransposons and was termed midline-jumper. In addition to cDNA clones generated from internal retrotransposon sequences, we have identified one cDNA clone that appears to reflect a transposition event, indicating that the midline-jumper retrotransposon is not only transcribed but also able to transpose during Drosophila development.


Assuntos
Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Retroelementos , Animais , DNA Complementar/metabolismo , Drosophila , Hibridização In Situ , Modelos Genéticos , Neuroglia/metabolismo
7.
Curr Biol ; 10(10): R388-91, 2000 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-10837218

RESUMO

Activation of the Drosophila EGF receptor requires the transmembrane TGF-alpha-like ligand Spitz. Recent studies have shed new light on the role of two transmembrane proteins, Star and Rhomboid, in the presentation and subsequent proteolytic processing of Spitz.


Assuntos
Proteínas de Drosophila , Drosophila/metabolismo , Fator de Crescimento Epidérmico , Receptores ErbB/genética , Receptores ErbB/metabolismo , Transdução de Sinais , Animais , Drosophila/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo
8.
Neuron ; 26(2): 357-70, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10839355

RESUMO

Here we report the description of the Drosophila gene futsch, which encodes a protein recognized by the monoclonal antibody 22C10 that has been widely used to visualize neuronal morphology and axonal projections. The Futsch protein is 5327 amino acids in length. It localizes to the microtubule compartment of the cell and associates with microtubules in vitro. The N- and C-terminal domains of Futsch are homologous to the vertebrate MAP1B microtubule-associated protein. The central domain of the Futsch protein is highly repetitive and shows sequence similarity to neurofilament proteins of which no Drosophila homologs have been reported. Loss-of-function analyses demonstrate that during embryogenesis Futsch is necessary for dendritic and axonal growth. Gain-of-function analyses demonstrate a functional interaction of Futsch with other MAPs. In addition, we show that during development, futsch expression is negatively regulated in nonneuronal tissues.


Assuntos
Axônios/fisiologia , Dendritos/fisiologia , Proteínas de Drosophila , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Fatores de Crescimento Neural/fisiologia , Envelhecimento/metabolismo , Sequência de Aminoácidos/genética , Animais , Humanos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/imunologia , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/fisiologia , Dados de Sequência Molecular , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/imunologia , Fatores de Crescimento Neural/metabolismo , Homologia de Sequência de Aminoácidos , Transmissão Sináptica/fisiologia
9.
Neuron ; 26(2): 371-82, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10839356

RESUMO

We present evidence that Futsch, a novel protein with MAP1B homology, controls synaptic growth at the Drosophila neuromuscularjunction through the regulation of the synaptic microtubule cytoskeleton. Futsch colocalizes with microtubules and identifies cytoskeletal loops that traverse the lateral margin of select synaptic boutons. An apparent rearrangement of microtubule loop architecture occurs during bouton division, and a genetic analysis indicates that Futsch is necessary for this process. futsch mutations disrupt synaptic microtubule organization, reduce bouton number, and increase bouton size. These deficits can be partially rescued by neuronal overexpression of a futsch MAP1B homology domain. Finally, genetic manipulations that increase nerve-terminal branching correlate with increased synaptic microtubule loop formation, and both processes require normal Futsch function. These data suggest a common microtubule-based growth mechanism at the synapse and growth cone.


Assuntos
Proteínas de Drosophila , Drosophila/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Microtúbulos/fisiologia , Fatores de Crescimento Neural/fisiologia , Sinapses/fisiologia , Animais , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Microtúbulos/ultraestrutura , Terminações Nervosas/fisiologia , Terminações Nervosas/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Sinapses/ultraestrutura , Distribuição Tecidual
10.
Genes Dev ; 14(7): 863-73, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10766742

RESUMO

In Drosophila, the correct formation of the segmental commissures depends on neuron-glial interactions at the midline. The VUM midline neurons extend axons along which glial cells migrate in between anterior and posterior commissures. Here, we show that the gene kette is required for the normal projection of the VUM axons and subsequently disrupts glial migration. Axonal projection defects are also found for many other moto- and interneurons. In addition, kette affects the cell morphology of mesodermal and epidermal derivatives, which show an abnormal actin cytoskeleton. The KETTE protein is homologous to the transmembrane protein HEM-2/NAP1 evolutionary conserved from worms to vertebrates. In vitro analysis has shown a specific interaction of the vertebrate HEM-2/NAP1 with the SH2-SH3 adapter protein NCK and the small GTPase RAC1, which both have been implicated in regulating cytoskeleton organization and axonal growth. Hypomorphic kette mutations lead to axonal defects similar to mutations in the Drosophila NCK homolog dreadlocks. Furthermore, we show that kette and dock mutants genetically interact. NCK is thought to interact with the small G proteins RAC1 and CDC42, which play a role in axonal growth. In line with these observations, a kette phenocopy can be obtained following directed expression of mutant DCDC42 or DRAC1 in the CNS midline. In addition, the kette mutant phenotype can be partially rescued by expression of an activated DRAC1 transgene. Our data suggest an important role of the HEM-2 protein in cytoskeletal organization during axonal pathfinding.


Assuntos
Axônios/fisiologia , Citoesqueleto/fisiologia , Drosophila/embriologia , Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Proteínas de Membrana/metabolismo , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/genética , Neuroglia/fisiologia , Neurônios/fisiologia , Proteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular , Sequência Conservada , Citoesqueleto/ultraestrutura , Proteínas de Drosophila , Embrião não Mamífero/fisiologia , Metanossulfonato de Etila , Mutagênese , Proteínas Nucleares , Proteína 1 de Modelagem do Nucleossomo , Vertebrados
11.
Mech Dev ; 91(1-2): 197-208, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10704844

RESUMO

In Drosophila lateral glial cell development is initiated by the transcription factor encoded by glial cells missing. glial cells missing activates downstream transcription factors such as repo and pointed which subsequently control terminal glial differentiation. The gene loco has been identified as a potential target gene of pointed and is involved in terminal glial differentiation. It encodes an RGS domain protein expressed specifically by the lateral glial cells in the developing embryonic CNS. Here we analyzed the loco promoter and the control of the glial-specific transcription pattern. Using promoter-reporter gene fusions we identified a 1.9 kb promoter element capable of directing the almost complete loco gene expression pattern. Sequence analysis suggested the presence of gcm and pointed DNA binding sites. Following in vitro mutagenesis of these sites we demonstrated their relevance in vivo. The expression of loco is initially dependent on gcm. During subsequent stages of embryonic development gcm and pointed appear to activate loco transcription synergistically. In addition, at least two other factors appear to repress loco expression in the ectoderm and in the CNS midline cells.


Assuntos
Proteínas de Drosophila , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Insetos/genética , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Transcrição Gênica , Animais , Sítios de Ligação , Proteínas de Ligação a DNA , Drosophila/embriologia , Ectoderma , Elementos Facilitadores Genéticos , Neuroglia , Neuropeptídeos/genética , Proteínas Proto-Oncogênicas/genética , Análise de Sequência , Transativadores/genética , Fatores de Transcrição , beta-Galactosidase/genética
12.
Curr Opin Neurobiol ; 9(5): 531-6, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10508736

RESUMO

Glial cells are pivotal players during the development and function of complex nervous systems. In Drosophila, recent genetic analyses have revealed several genes that control differentiation and function of CNS glial cells and their interactions with neurons can be studied in detail at the CNS midline, where it is essential for the correct establishment of the commissural axon pattern.


Assuntos
Sistema Nervoso Central/embriologia , Drosophila/embriologia , Neuroglia/fisiologia , Animais , Axônios/fisiologia , Diferenciação Celular/fisiologia , Senescência Celular/fisiologia , Embrião não Mamífero/fisiologia , Neuroglia/citologia
13.
Dev Biol ; 209(2): 381-98, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10328928

RESUMO

In the ventral nerve cord of Drosophila most axons are organized in a simple, ladder-like pattern. Two segmental commissures connect the hemisegments along the mediolateral and two longitudinal connectives connect individual neuromeres along the anterior-posterior axis. Cells located at the midline of the developing CNS first guide commissural growth cones toward and across the midline. In later stages, midline glial cells are required to separate anterior and posterior commissures into distinct axon bundles. To unravel the genes underlying the formation of axon pattern in the embryonic ventral nerve cord, we conducted a saturating ethylmethane sulfonate mutagenesis, screening for mutations which disrupt this process. Subsequent genetic and phenotypic analyses support a sequential model of axon pattern formation in the embryonic ventral nerve cord. Specification of midline cell lineages is brought about by the action of segment polarity genes. Five genes are necessary for the establishment of the commissures. In addition to commissureless, the netrin genes, and the netrin receptor encoded by the frazzled gene, two gene functions are required for the initial formation of commissural tracts. Over 20 genes appear to be required for correct development of the midline glial cells which are necessary for the formation of distinct segmental commissures.


Assuntos
Sistema Nervoso Central/embriologia , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Animais , Axônios/patologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Linhagem da Célula , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila , Drosophila melanogaster/genética , Drosophila melanogaster/ultraestrutura , Embrião não Mamífero/ultraestrutura , Desenvolvimento Embrionário , Metanossulfonato de Etila , Genes de Insetos , Genes Letais , Genótipo , Proteínas de Insetos/genética , Proteínas de Insetos/fisiologia , Morfogênese/genética , Mutagênese , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/fisiologia , Receptores de Netrina , Netrina-1 , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Fenótipo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Proteínas Supressoras de Tumor
14.
Development ; 126(8): 1781-91, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10079238

RESUMO

In Drosophila, glial cell development depends on the gene glial cells missing (gcm). gcm activates the expression of other transcription factors such as pointed and repo, which control subsequent glial differentiation. In order to better understand glial cell differentiation, we have screened for genes whose expression in glial cells depends on the activity of pointed. Using an enhancer trap approach, we have identified loco as such a gene. loco is expressed in most lateral CNS glial cells throughout development. Embryos lacking loco function have an normal overall morphology, but fail to hatch. Ultrastructural analysis of homozygous mutant loco embryos reveals a severe glial cell differentiation defect. Mutant glial cells fail to properly ensheath longitudinal axon tracts and do not form the normal glial-glial cell contacts, resulting in a disruption of the blood-brain barrier. Hypomorphic loco alleles were isolated following an EMS mutagenesis. Rare escapers eclose which show impaired locomotor capabilities. loco encodes the first two known Drosophila members of the family of Regulators of G-protein signalling (RGS) proteins, known to interact with the alpha subunits of G-proteins. loco specifically interacts with the Drosophila alphai-subunit. Strikingly, the interaction is not confined to the RGS domain. This interaction and the coexpression of LOCO and Galphai suggests a function of G-protein signalling for glial cell development.


Assuntos
Proteínas de Drosophila , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Insetos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/citologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Diferenciação Celular , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , DNA Complementar , Proteínas de Ligação a DNA , Drosophila/embriologia , Elementos Facilitadores Genéticos , Proteínas Ativadoras de GTPase , Expressão Gênica , Humanos , Proteínas de Insetos/genética , Dados de Sequência Molecular , Mutagênese , Proteínas do Tecido Nervoso/genética , Sistema Nervoso Periférico/embriologia , Sistema Nervoso Periférico/metabolismo , Fenótipo , Proteínas , Proteínas Proto-Oncogênicas/genética , Ratos , Fatores de Transcrição , beta-Galactosidase/biossíntese
15.
Development ; 126(4): 771-9, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9895324

RESUMO

Most of the neurons of the ventral nerve cord send out long projecting axons which cross the midline. In the Drosophila central nervous system (CNS) cells of the midline give rise to neuronal and glial lineages with different functions during the establishment of the commissural pattern. Here we present evidence that beside the previously known NETRIN/FRAZZLED (DCC) signalling system an additional attractive system(s) is operating in the developing embryonic nervous system of Drosophila. Attractive cues appear to be provided by the midline neurons. We show that the glial cells present repulsive signals to the previously described ROUNDABOUT receptor in addition to a permissive contact-dependent signal helping commissural growth cones across the midline. A novel repulsive component is encoded by the karussell gene. Furthermore the midline glial cells separate anterior and posterior commissures. By genetic criteria we demonstrate that some of the genes we have identified are acting in the midline glia whereas other genes are required in the midline neurons. The results lead to a detailed model relating different cellular functions to axonal patterning at the midline.


Assuntos
Sistema Nervoso Central/embriologia , Drosophila/embriologia , Animais , Padronização Corporal/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Sistema Nervoso Central/crescimento & desenvolvimento , Genótipo , Imuno-Histoquímica , Mutação/genética , Neuroglia/metabolismo , Fenótipo
16.
Adv Exp Med Biol ; 468: 23-32, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10635017

RESUMO

The major axon tracts in the embryonic CNS of Drosophila are organized in a simple, ladder like pattern. Each neuromere contains two commissures which connect the contra-lateral hemi-neuromeres and two longitudinal connectives which connect the different neuromeres along the anterior-posterior axis. The formation of these axon tracts occurs in close association with different glial cells. Loss of specific glial cells within the CNS leads to predictable defects in the organization of the CNS axon pattern. To unravel the genes underlying CNS glia development, we have conducted a saturating F2 EMS mutagenesis, screening for mutations, which disrupt axon pattern in the embryonic nervous system. We found a large number of mutations that lead to phenotypes indicative for glia defects. The analysis of the genes identified, show that glial cell differentiation requires the function of two independent regulatory pathways.


Assuntos
Drosophila/embriologia , Drosophila/genética , Sistema Nervoso/embriologia , Neuroglia/fisiologia , Animais , Axônios/fisiologia , Padronização Corporal , Metanossulfonato de Etila , Mutagênese , Mutação , Neuroglia/citologia
17.
Development ; 124(24): 4949-58, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9362458

RESUMO

Cells located at the midline of the developing central nervous system perform a number of conserved functions during the establishment of the lateral CNS. The midline cells of the Drosophila CNS were previously shown to be required for correct pattern formation in the ventral ectoderm and for the induction of specific mesodermal cells. Here we investigated whether the midline cells are required for the correct development of lateral CNS cells as well. Embryos that lack midline cells through genetic ablation show a 15% reduction in the number of cortical CNS cells. A similar thinning of the ventral nerve cord can be observed following mechanical ablation of the midline cells. We have identified a number of specific neuronal and glial cell markers that are reduced in CNS midline-less embryos (in single-minded embryos, in early heat-shocked Notch(ts1) embryos or in embryos where we mechanically ablated the midline cells). Genetic data suggest that both neuronal and glial midline cell lineages are required for differentiation of lateral CNS cells. We could rescue the lateral CNS phenotype of single-minded mutant embryos by transplantation of midline cells as well as by homotopic expression of single-minded, the master gene for midline development. Furthermore, ectopic midline cells are able to induce enhanced expression of some lateral CNS cell markers. We thus conclude that the CNS midline plays an important role in the differentiation or maintenance of the lateral CNS cortex.


Assuntos
Sistema Nervoso Central/embriologia , Proteínas de Drosophila , Drosophila/embriologia , Indução Embrionária/fisiologia , Fator de Crescimento Epidérmico , Neurônios/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Padronização Corporal/fisiologia , Transplante de Células , Sistema Nervoso Central/citologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Drosophila/genética , Ectoderma , Elementos Facilitadores Genéticos/genética , Receptores ErbB/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/fisiologia , Mutação , Neuroglia/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Transdução de Sinais/fisiologia
18.
Development ; 124(14): 2681-90, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9226439

RESUMO

The Drosophila ventral midline cells generate a discrete set of CNS lineages, required for proper patterning of the ventral ectoderm. Here we provide the first evidence that the CNS midline cells also exert inductive effects on the mesoderm. Mesodermal progenitors adjacent to the midline progenitor cells give rise to ventral somatic mucles and a pair of unique cells that come to lie dorsomedially on top of the ventral nerve cord, the so-called DM cells. Cell ablation as well as cell transplantation experiments indicate that formation of the DM cells is induced by midline progenitors in the early embryo. These results are corroborated by genetic analyses. Mutant single minded embryos lack the CNS midline as well as the DM cells. Embryos mutant for any of the spitz group genes, which primarily express defects in the midline glial cell lineages, show reduced formation of the DM cells. Conversely, directed overexpression of secreted SPITZ by some or all CNS midline cells leads to the formation of additional DM cells. Furthermore we show that DM cell development does not depend on the absolute concentration of a local inductor but appears to require a graded source of an inducing signal. Thus, the Drosophila CNS midline cells play a central inductive role in patterning the mesoderm as well as the underlying ectoderm.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster/embriologia , Fator de Crescimento Epidérmico , Mesoderma/citologia , Animais , Movimento Celular , Sistema Nervoso Central/embriologia , Proteínas de Ligação a DNA , Indução Embrionária , Proteínas do Olho/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/fisiologia , Morfogênese , Proteínas do Tecido Nervoso/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais , Fatores de Transcrição
19.
Development ; 124(12): 2307-16, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9199357

RESUMO

Two classes of glial cells are found in the embryonic Drosophila CNS, midline glial cells and lateral glial cells. Midline glial development is triggered by EGF-receptor signalling, whereas lateral glial development is controlled by the gcm gene. Subsequent glial cell differentiation depends partly on the pointed gene. Here we describe a novel component required for all CNS glia development. The tramtrack gene encodes two zinc-finger proteins, one of which, ttkp69, is expressed in all non-neuronal CNS cells. We show that ttkp69 is downstream of gcm and can repress neuronal differentiation. Double mutant analysis and coexpression experiments indicate that glial cell differentiation may depend on a dual process, requiring the activation of glial differentiation by pointed and the concomitant repression of neuronal development by tramtrack.


Assuntos
Sistema Nervoso Central/embriologia , Proteínas de Drosophila , Drosophila/embriologia , Drosophila/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Neuroglia/fisiologia , Proteínas Repressoras , Animais , Animais Geneticamente Modificados , Diferenciação Celular/genética , Sistema Nervoso Central/crescimento & desenvolvimento , Proteínas de Ligação a DNA/fisiologia , Drosophila/genética , Embrião não Mamífero/fisiologia , Indução Embrionária , Teste de Complementação Genética , Mutação , Proteínas do Tecido Nervoso , Neuroglia/ultraestrutura , Neurônios/fisiologia , Neuropeptídeos/fisiologia , Fenótipo , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Fatores de Transcrição
20.
Mech Dev ; 64(1-2): 137-51, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9232604

RESUMO

The midline glial cells are required for correct formation of the axonal pattern in the embryonic ventral nerve cord of Drosophila. Initially, six midline cells form an equivalence group with the capacity to develop as glial cells. By the end of embryonic development three to four cells are singled out as midline glial cells. Midline glia development occurs in two steps, both of which depend on the activation of the Drosophila EGF-receptor homolog and subsequent ras1/raf-mediated signal transduction. Nuclear targets of this signalling cascade are the ETS domain transcription factors pointedP2 and yan. In the midline glia pointedP2 in turn activates the transcription of argos, which encodes a diffusible negative regulator of EGF-receptor signalling.


Assuntos
Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Proteínas de Drosophila , Drosophila/embriologia , Neuroglia/citologia , Proteínas Quinases , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Animais , Sistema Nervoso Central/metabolismo , Proteínas de Ligação a DNA/metabolismo , Drosophila/genética , Drosophila/metabolismo , Receptores ErbB/metabolismo , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Mutação , Proteínas do Tecido Nervoso , Neuroglia/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptores de Peptídeos de Invertebrados/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA