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










Base de dados
Intervalo de ano de publicação
1.
Elife ; 102021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33527895

RESUMO

Understanding how injury to the central nervous system induces de novo neurogenesis in animals would help promote regeneration in humans. Regenerative neurogenesis could originate from glia and glial neuron-glia antigen-2 (NG2) may sense injury-induced neuronal signals, but these are unknown. Here, we used Drosophila to search for genes functionally related to the NG2 homologue kon-tiki (kon), and identified Islet Antigen-2 (Ia-2), required in neurons for insulin secretion. Both loss and over-expression of ia-2 induced neural stem cell gene expression, injury increased ia-2 expression and induced ectopic neural stem cells. Using genetic analysis and lineage tracing, we demonstrate that Ia-2 and Kon regulate Drosophila insulin-like peptide 6 (Dilp-6) to induce glial proliferation and neural stem cells from glia. Ectopic neural stem cells can divide, and limited de novo neurogenesis could be traced back to glial cells. Altogether, Ia-2 and Dilp-6 drive a neuron-glia relay that restores glia and reprogrammes glia into neural stem cells for regeneration.


Assuntos
Sistema Nervoso Central/lesões , Drosophila melanogaster/crescimento & desenvolvimento , Neurogênese , Regeneração , Animais , Autoanticorpos/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Larva/genética , Larva/metabolismo , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Somatomedinas/metabolismo
2.
Development ; 145(15)2018 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-30002129

RESUMO

During development, tissue growth is mediated by either cell proliferation or cell growth, coupled with polyploidy. Both strategies are employed by the cell types that make up the Drosophila blood-brain barrier. During larval growth, the perineurial glia proliferate, whereas the subperineurial glia expand enormously and become polyploid. Here, we show that the level of ploidy in the subperineurial glia is controlled by the N-terminal asparagine amidohydrolase homolog Öbek, and high Öbek levels are required to limit replication. In contrast, perineurial glia express moderate levels of Öbek, and increased Öbek expression blocks their proliferation. Interestingly, other dividing cells are not affected by alteration of Öbek expression. In glia, Öbek counteracts fibroblast growth factor and Hippo signaling to differentially affect cell growth and number. We propose a mechanism by which growth signals are integrated differentially in a glia-specific manner through different levels of Öbek protein to adjust cell proliferation versus endoreplication in the blood-brain barrier.


Assuntos
Asparaginase/genética , Barreira Hematoencefálica/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ploidias , Amidoidrolases/metabolismo , Animais , Asparaginase/metabolismo , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/embriologia , Núcleo Celular/metabolismo , Proliferação de Células , Sobrevivência Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Endorreduplicação , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Genes de Insetos , Modelos Biológicos , Neuroglia/citologia , Neuroglia/metabolismo , Transdução de Sinais
3.
Front Syst Neurosci ; 11: 68, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29021745

RESUMO

The biogenic amines octopamine (OA) and tyramine (TA) modulate insect motor behavior in an antagonistic manner. OA generally enhances locomotor behaviors such as Drosophila larval crawling and flight, whereas TA decreases locomotor activity. However, the mechanisms and cellular targets of TA modulation of locomotor activity are incompletely understood. This study combines immunocytochemistry, genetics and flight behavioral assays in the Drosophila model system to test the role of a candidate enzyme for TA catabolism, named Nazgul (Naz), in flight motor behavioral control. We hypothesize that the dehydrogenase/reductase Naz represents a critical step in TA catabolism. Immunocytochemistry reveals that Naz is localized to a subset of Repo positive glial cells with cell bodies along the motor neuropil borders and numerous positive Naz arborizations extending into the synaptic flight motor neuropil. RNAi knock down of Naz in Repo positive glial cells reduces Naz protein level below detection level by Western blotting. The resulting consequence is a reduction in flight durations, thus mimicking known motor behavioral phenotypes as resulting from increased TA levels. In accord with the interpretation that reduced TA degradation by Naz results in increased TA levels in the flight motor neuropil, the motor behavioral phenotype can be rescued by blocking TA receptors. Our findings indicate that TA modulates flight motor behavior by acting on central circuitry and that TA is normally taken up from the central motor neuropil by Repo-positive glial cells, desaminated and further degraded by Naz.

4.
Genome Res ; 27(7): 1273-1285, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28381612

RESUMO

Drosophila melanogaster is a widely used genetic model organism in developmental biology. While this model organism has been intensively studied at the RNA level, a comprehensive proteomic study covering the complete life cycle is still missing. Here, we apply label-free quantitative proteomics to explore proteome remodeling across Drosophila's life cycle, resulting in 7952 proteins, and provide a high temporal-resolved embryogenesis proteome of 5458 proteins. Our proteome data enabled us to monitor isoform-specific expression of 34 genes during development, to identify the pseudogene Cyp9f3Ψ as a protein-coding gene, and to obtain evidence of 268 small proteins. Moreover, the comparison with available transcriptomic data uncovered examples of poor correlation between mRNA and protein, underscoring the importance of proteomics to study developmental progression. Data integration of our embryogenesis proteome with tissue-specific data revealed spatial and temporal information for further functional studies of yet uncharacterized proteins. Overall, our high resolution proteomes provide a powerful resource and can be explored in detail in our interactive web interface.


Assuntos
Proteínas de Drosophila/biossíntese , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteoma/biossíntese , Animais , Drosophila melanogaster
5.
Artigo em Inglês | MEDLINE | ID: mdl-26224590

RESUMO

Throughout evolution, glia have key regulatory roles in neural development and function. Typically, they control the response to developmental and/or pathological signals, thereby affecting neural proliferation, remodeling, survival, and regeneration. Such complex biology depends on the plastic features of glial cells, but also on the presence of different classes of glial cells, hence the importance of understanding the cellular and the molecular mechanisms underlying their development. The fly community has made major breakthroughs by characterizing the bases of gliogenesis and here we describe the glial lineages as well as the glial promoting factor active in the embryo of Drosophila melanogaster. WIREs Dev Biol 2016, 5:67-84. doi: 10.1002/wdev.200 For further resources related to this article, please visit the WIREs website.


Assuntos
Drosophila melanogaster/citologia , Neuroglia/fisiologia , Animais , Evolução Biológica , Diferenciação Celular , Proteínas de Drosophila/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Larva/citologia , Células-Tronco Neurais/fisiologia , Regeneração
6.
Glia ; 63(8): 1291-302, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25779863

RESUMO

Development and general organization of the nervous system is comparable between insects and vertebrates. Our current knowledge on the formation of neurogenic anlagen and the generation of neural stem cells is deeply influenced by work done in invertebrate model organisms such as Drosophila and Caenorhabditis elegans. It is the aim of this review to summarize the most important steps in neurogenesis in the Drosophila embryo with a special emphasis on glial cell progenitors and the specification of glial cells. Induction of neurogenic regions during early embryogenesis and determination of neural stem cells are briefly described. Special attention is given to the formation of neural precursors called neuroblasts (NB) and their lineages. NBs divide in a stem cell mode to generate a cell clone of either neurons and/or glial cells. The latter require the activation of the transcription factor glial cells missing (gcm), thus providing a binary switch between neuronal and glial cell fates. Further aspects of glial cell specification and the resulting heterogeneity of the glial population in Drosophila are discussed.


Assuntos
Drosophila/embriologia , Drosophila/fisiologia , Células-Tronco Neurais/fisiologia , Neuroglia/fisiologia , Animais , Drosophila/citologia , Células-Tronco Neurais/citologia , Neuroglia/citologia
7.
Fly (Austin) ; 8(2): 86-90, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25483246

RESUMO

In the article "Predetermined embryonic glial cells form the distinct glial sheaths of the Drosophila peripheral nervous system" we combined our expertise to identify glial cells of the embryonic peripheral nervous system on a single cell resolution with the possibility to genetically label cells using Flybow. We show that all 12 embryonic peripheral glial cells (ePG) per abdominal hemisegment persist into larval (and even adult) stages and differentially contribute to the three distinct glial layers surrounding peripheral nerves. Repetitive labelings of the same cell further revealed that layer affiliation, morphological expansion, and control of proliferation are predetermined and subject to an intrinsic differentiation program. Interestingly, wrapping and subperineurial glia undergo enormous hypertrophy in response to larval growth and elongation of peripheral nerves, while perineurial glia respond to the same environmental changes with hyperplasia. Increase in cell number from embryo (12 cells per hemisegment) to third instar (up to 50 cells per hemisegment) is the result of proliferation of a single ePG that serves as transient progenitor and only contributes to the outermost perineurial glial layer.


Assuntos
Diferenciação Celular , Drosophila/embriologia , Neuroglia/citologia , Animais , Crescimento Celular , Proliferação de Células , Drosophila/citologia , Mitose , Sistema Nervoso Periférico/citologia
8.
Sci Signal ; 6(300): ra96, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24194583

RESUMO

During development, differentiation is often initiated by the activation of different receptor tyrosine kinases (RTKs), which results in the tightly regulated activation of cytoplasmic signaling cascades. In the differentiation of neurons and glia in the developing Drosophila eye, we found that the proper intensity of RTK signaling downstream of fibroblast growth factor receptor (FGFR) or epidermal growth factor receptor required two mutually antagonistic feedback loops. We identified a positive feedback loop mediated by the Ras association (RA) domain-containing protein Rau that sustained Ras activity and counteracted the negative feedback loop mediated by Sprouty. Rau has two RA domains that together showed a binding preference for GTP (guanosine 5'-triphosphate)-loaded (active) Ras. Rau homodimerized and was found in large-molecular weight complexes. Deletion of rau in flies decreased the differentiation of retinal wrapping glia and induced a rough eye phenotype, similar to that seen in alterations of Ras signaling. Further, the expression of sprouty was repressed and that of rau was increased by the COUP transcription factor Seven-up in the presence of weak, but not constitutive, activation of FGFR. Together, our findings reveal another regulatory mechanism that controls the intensity of RTK signaling in the developing neural network in the Drosophila eye.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Olho/embriologia , Retroalimentação Fisiológica/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Fatores de Transcrição COUP/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática/fisiologia , Olho/citologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Proteínas de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Neuroglia/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Esteroides/metabolismo
9.
Development ; 140(17): 3657-68, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23903191

RESUMO

One of the numerous functions of glial cells in Drosophila is the ensheathment of neurons to isolate them from the potassium-rich haemolymph, thereby establishing the blood-brain barrier. Peripheral nerves of flies are surrounded by three distinct glial cell types. Although all embryonic peripheral glia (ePG) have been identified on a single-cell level, their contribution to the three glial sheaths is not known. We used the Flybow system to label and identify each individual ePG in the living embryo and followed them into third instar larva. We demonstrate that all ePG persist until the end of larval development and some even to adulthood. We uncover the origin of all three glial sheaths and describe the larval differentiation of each peripheral glial cell in detail. Interestingly, just one ePG (ePG2) exhibits mitotic activity during larval stages, giving rise to up to 30 glial cells along a single peripheral nerve tract forming the outermost perineurial layer. The unique mitotic ability of ePG2 and the layer affiliation of additional cells were confirmed by in vivo ablation experiments and layer-specific block of cell cycle progression. The number of cells generated by this glial progenitor and hence the control of perineurial hyperplasia correlate with the length of the abdominal nerves. By contrast, the wrapping and subperineurial glia layers show enormous hypertrophy in response to larval growth. This characterisation of the embryonic origin and development of each glial sheath will facilitate functional studies, as they can now be addressed distinctively and genetically manipulated in the embryo.


Assuntos
Drosophila/embriologia , Neuroglia/fisiologia , Sistema Nervoso Periférico/embriologia , Animais , Diferenciação Celular/fisiologia , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/imunologia , Proteínas de Homeodomínio/imunologia , Imuno-Histoquímica , Microscopia Confocal , Neuroglia/citologia , Sistema Nervoso Periférico/crescimento & desenvolvimento
10.
EMBO Rep ; 13(9): 827-34, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22744314

RESUMO

Oligodendroglial Myelin Basic Protein (MBP) synthesis is essential for myelin formation in the central nervous system. During oligodendrocyte differentiation, MBP mRNA is kept in a translationally silenced state while intracellularly transported, until neuron-derived signals initiate localized MBP translation. Here we identify the small non-coding RNA 715 (sncRNA715) as an inhibitor of MBP translation. SncRNA715 localizes to cytoplasmic granular structures and associates with MBP mRNA transport granule components. We also detect increased levels of sncRNA715 in demyelinated chronic human multiple sclerosis lesions, which contain MBP mRNA but lack MBP protein.


Assuntos
Regulação da Expressão Gênica , Proteína Básica da Mielina/biossíntese , Pequeno RNA não Traduzido/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Grânulos Citoplasmáticos/metabolismo , Humanos , Camundongos , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/biossíntese , Ratos
11.
J Cell Sci ; 125(Pt 2): 461-72, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22302994

RESUMO

Here we report on the generation and in vivo analysis of a series of loss-of-function mutants for the Drosophila ArfGEF, Gartenzwerg. The Drosophila gene gartenzwerg (garz) encodes the orthologue of mammalian GBF1. garz is expressed ubiquitously in embryos with substantially higher abundance in cells forming diverse tubular structures such as salivary glands, trachea, proventriculus or hindgut. In the absence of functional Garz protein, the integrity of the Golgi complex is impaired. As a result, both vesicle transport of cargo proteins and directed apical membrane delivery are severely disrupted. Dysfunction of the Arf1-COPI machinery caused by a loss of Garz leads to perturbations in establishing a polarized epithelial architecture of tubular organs. Furthermore, insufficient apical transport of proteins and other membrane components causes incomplete luminal diameter expansion and deficiencies in extracellular matrix assembly. The fact that homologues of Garz are present in every annotated metazoan genome indicates that secretion processes mediated by the GBF-type ArfGEFs play a universal role in animal development.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila/embriologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Via Secretória , Animais , Linhagem Celular , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mutação , Glândulas Salivares/embriologia , Glândulas Salivares/ultraestrutura , Traqueia/embriologia , Traqueia/metabolismo , Traqueia/ultraestrutura
12.
Dev Biol ; 350(1): 89-100, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21094637

RESUMO

Transport of liquids or gases in biological tubes is fundamental for many physiological processes. Our knowledge on how tubular organs are formed during organogenesis and tissue remodeling has increased dramatically during the last decade. Studies on different animal systems have helped to unravel some of the molecular mechanisms underlying tubulogenesis. Tube architecture varies dramatically in different organs and different species, ranging from tubes formed by several cells constituting the cross section, tubes formed by single cells wrapping an internal luminal space or tubes that are formed within a cell. Some tubes display branching whereas others remain linear without intersections. The modes of shaping, growing and pre-patterning a tube are also different and it is still not known whether these diverse architectures and modes of differentiation are realized by sharing common signaling pathways or regulatory networks. However, several recent investigations provide evidence for the attractive hypothesis that the Drosophila cardiogenesis and heart tube formation shares many similarities with primary angiogenesis in vertebrates. Additionally, another important step to unravel the complex system of lumen formation has been the outcome of recent studies that junctional proteins, matrix components as well as proteins acting as attractant and repellent cues play a role in the formation of the Drosophila heart lumen. In this study we show the requirement for the repulsively active Unc5 transmembrane receptor to facilitate tubulogenesis in the dorsal vessel of Drosophila. Unc5 is localized in the luminal membrane compartment of cardiomyocytes and animals lacking Unc5 fail to form a heart lumen. Our findings support the idea that Unc5 is crucial for lumen formation and thereby represents a repulsive cue acting during Drosophila heart tube formation.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Coração/embriologia , Receptores de Superfície Celular/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Ligantes , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fatores de Crescimento Neural/metabolismo , Receptores de Netrina , Netrina-1 , Receptores de Superfície Celular/genética , Proteínas Supressoras de Tumor/metabolismo
13.
Development ; 137(8): 1251-62, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20223758

RESUMO

Development of the nervous system and establishment of complex neuronal networks require the concerted activity of different signalling events and guidance cues, which include Netrins and their receptors. In Drosophila, two Netrins are expressed during embryogenesis by cells of the ventral midline and serve as attractant or repellent cues for navigating axons. We asked whether glial cells, which are also motile, are guided by similar cues to axons, and analysed the influence of Netrins and their receptors on glial cell migration during embryonic development. We show that in Netrin mutants, two distinct populations of glial cells are affected: longitudinal glia (LG) fail to migrate medially in the early stages of neurogenesis, whereas distinct embryonic peripheral glia (ePG) do not properly migrate laterally into the periphery. We further show that early Netrin-dependent guidance of LG requires expression of the receptor Frazzled (Fra) already in the precursor cell. At these early stages, Netrins are not yet expressed by cells of the ventral midline and we provide evidence for a novel Netrin source within the neurogenic region that includes neuroblasts. Later in development, most ePG transiently express uncoordinated 5 (unc5) during their migratory phase. In unc5 mutants, however, two of these cells in particular exhibit defective migration and stall in, or close to, the central nervous system. Both phenotypes are reversible in cell-specific rescue experiments, indicating that Netrin-mediated signalling via Fra (in LG) or Unc5 (in ePG) is a cell-autonomous effect.


Assuntos
Drosophila/embriologia , Fatores de Crescimento Neural/fisiologia , Neuroglia/fisiologia , Animais , Movimento Celular , Polaridade Celular , Sinais (Psicologia) , Drosophila/genética , Drosophila/fisiologia , Éxons/genética , Imuno-Histoquímica , Mutação , Netrina-1 , Neuroglia/citologia , Fenótipo , Transdução de Sinais , Proteínas Supressoras de Tumor/fisiologia
14.
Mech Dev ; 125(5-6): 542-57, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18296030

RESUMO

In the Drosophila embryonic CNS several subtypes of glial cells develop, which arrange themselves at characteristic positions and presumably fulfil specific functions. The mechanisms leading to the specification and differentiation of glial subtypes are largely unknown. By DiI labelling in glia-specific Gal4 lines we have clarified the lineages of the lateral glia in the embryonic ventral nerve cord and linked each glial cell to a specific stem cell. For the lineage of the longitudinal glioblast we show that it consists of 9 cells, which acquire at least four different identities. A large collection of molecular markers (many of them representing transcription factors and potential Gcm target genes) reveals that individual glial cells express specific combinations of markers. However, cluster analysis uncovers similar combinatorial codes for cells within, and significant differences between the categories of surface-associated, cortex-associated, and longitudinal glia. Glial cells derived from the same stem cell may be homogeneous (though not identical; stem cells NB1-1, NB5-6, NB6-4, LGB) or heterogeneous (NB7-4, NB1-3) with regard to gene expression. In addition to providing a powerful tool to analyse the fate of individual glial cells in different genetic backgrounds, each of these marker genes represents a candidate factor involved in glial specification or differentiation. We demonstrate this by the analysis of a castor loss of function mutation, which affects the number and migration of specific glial cells.


Assuntos
Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Neuroglia/citologia , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Membrana Celular/metabolismo , Movimento Celular , Análise por Conglomerados , Marcadores Genéticos , Técnicas Genéticas , Hibridização In Situ , Sistema Nervoso/embriologia , Neuroglia/metabolismo , Sistema Nervoso Periférico/embriologia
15.
Mech Dev ; 125(3-4): 337-52, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18077143

RESUMO

Glial cells are crucial for the proper development and function of the nervous system. In the Drosophila embryo, the glial cells of the peripheral nervous system are generated both by central neuroblasts and sensory organ precursors. Most peripheral glial cells need to migrate along axonal projections of motor and sensory neurons to reach their final positions in the periphery. Here we studied the spatial and temporal pattern, the identity, the migration, and the origin of all peripheral glial cells in the truncal segments of wildtype embryos. The establishment of individual identities among these cells is reflected by the expression of a combinatorial code of molecular markers. This allows the identification of individual cells in various genetic backgrounds. Furthermore, mutant analysis of two of these marker genes, spalt major and castor, reveal their implication in peripheral glial development. Using confocal 4D microscopy to monitor and follow peripheral glia migration in living embryos, we show that the positioning of most of these cells is predetermined with minor variations, and that the order in which cells migrate into the periphery is almost fixed. By studying their lineages, we uncovered the origin of each of the peripheral glial cells and linked them to identified central and peripheral neural stem cells.


Assuntos
Movimento Celular , Drosophila/embriologia , Sistema Nervoso/embriologia , Neuroglia/fisiologia , Animais , Linhagem da Célula , Drosophila/citologia , Embrião não Mamífero/citologia , Sistema Nervoso/citologia , Neuroglia/citologia
16.
Dev Biol ; 301(1): 27-37, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17157832

RESUMO

A prominent feature of glial cells is their ability to migrate along axons to finally wrap and insulate them. In the embryonic Drosophila PNS, most glial cells are born in the CNS and have to migrate to reach their final destinations. To understand how migration of the peripheral glia is regulated, we have conducted a genetic screen looking for mutants that disrupt the normal glial pattern. Here we present an analysis of two of these mutants: Notch and numb. Complete loss of Notch function leads to an increase in the number of glial cells. Embryos hemizygous for the weak Notch(B-8X) allele display an irregular migration phenotype and mutant glial cells show an increased formation of filopodia-like structures. A similar phenotype occurs in embryos carrying the Notch(ts1) allele when shifted to the restrictive temperature during the glial cell migration phase, suggesting that Notch must be activated during glial migration. This is corroborated by the fact that cell-specific reduction of Notch activity in glial cells by directed numb expression also results in similar migration phenotypes. Since the glial migration phenotypes of Notch and numb mutants resemble each other, our data support a model where the precise temporal and quantitative regulation of Numb and Notch activity is not only required during fate decisions but also later during glial differentiation and migration.


Assuntos
Movimento Celular/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila/citologia , Hormônios Juvenis/fisiologia , Neuroglia/citologia , Receptores Notch/fisiologia , Animais , Drosophila/embriologia , Proteínas de Drosophila/genética , Imuno-Histoquímica , Hormônios Juvenis/genética , Mutagênese , Receptores Notch/genética , Transdução de Sinais
17.
Dev Biol ; 296(2): 545-60, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16762338

RESUMO

In the central nervous system of Drosophila, the induction of the glial cell fate is dependent on the transcription factor glial cells missing (gcm). Though a considerable number of other genes have been shown to be expressed in all or in subsets of glial cells, the course of glial cell differentiation and subtype specification is only poorly understood. This prompted us to design a whole genome microarray approach comparing gcm gain-of-function and, for the first time, gcm loss-of-function genetics to wildtype in time course experiments along embryogenesis. The microarray data were analyzed with special emphasis on the temporal profile of differential regulation. A comparison of both experiments enabled us to identify more than 300 potential gcm target genes. Validation by in situ hybridization revealed expression in glial cells, macrophages, and tendon cells (all three cell types depend on gcm) for 70 genes, of which more than 50 had been unknown to be under gcm control. Eighteen genes are exclusively expressed in glial cells, and their dependence on gcm was confirmed in situ. Initial considerations regarding the role of the newly discovered glial genes are discussed based on gene ontology and the temporal profile and subtype specificity of their expression. This collection of glial genes provides an important basis for the clarification of the genetic network controlling various aspects of glial development and function.


Assuntos
Drosophila/embriologia , Drosophila/genética , Perfilação da Expressão Gênica , Genes de Insetos , Neuroglia/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Drosophila/citologia , Proteínas de Drosophila , Perfilação da Expressão Gênica/normas , Regulação da Expressão Gênica no Desenvolvimento , Neuroglia/química , Neuroglia/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos/normas , Controle de Qualidade , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
18.
Dev Genes Evol ; 216(2): 105-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16328480

RESUMO

The analysis of mutants is an indispensable approach towards characterizing gene function. Combining several tools of Drosophila genetics, we designed a new strategy for a mutagenesis screen which is fast, easy-to-apply, and cheap. The combination of a cell-specific Gal4 line with an upstream activating sequence-green fluorescent protein (UAS-GFP) allows the in vivo detection of the cells or tissues of interest without the need for fixation and staining. To further simplify and accelerate the screening procedure, we generated recombinant flies that carry the Gal80 transgene in balancer chromosomes. Gal80 inactivates Gal4; and thus prevents GFP-expression during embryonic and postembryonic development in all individuals carrying the balancer chromosomes. This allows for an easy distinction in vivo between heterozygous and homozygous mutants, the latter being the only ones expressing GFP. Since most of the fly strains and balancer chromosomes can be substituted, this method is suitable for nearly any mutagenesis screen that does not have major restrictions.


Assuntos
Análise Mutacional de DNA/métodos , Drosophila/genética , Genes de Insetos/genética , Mutagênese , Animais , Animais Geneticamente Modificados , Sítios de Ligação , Linhagem Celular , Cromossomos/genética , Proteínas de Ligação a DNA , Drosophila/embriologia , Drosophila/crescimento & desenvolvimento , Embrião não Mamífero/anatomia & histologia , Embrião não Mamífero/química , Elementos Facilitadores Genéticos/genética , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Proteínas Repressoras/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo
19.
Development ; 131(18): 4521-32, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15342477

RESUMO

Locomotion relies on stable attachment of muscle fibres to their target sites, a process that allows for muscle contraction to generate movement. Here, we show that glide/gcm and glide2/gcm2, the fly glial cell determinants, are expressed in a subpopulation of embryonic tendon cells and required for their terminal differentiation. By using loss-of-function approaches, we show that in the absence of both genes, muscle attachment to tendon cells is altered, even though the molecular cascade induced by stripe, the tendon cell determinant, is normal. Moreover, we show that glide/gcm activates a new tendon cell gene independently of stripe. Finally, we show that segment polarity genes control the epidermal expression of glide/gcm and determine, within the segment, whether it induces glial or tendon cell-specific markers. Thus, under the control of positional cues, glide/gcm triggers a new molecular pathway involved in terminal tendon cell differentiation, which allows the establishment of functional muscle attachment sites and locomotion.


Assuntos
Diferenciação Celular , Drosophila/embriologia , Drosophila/metabolismo , Neuropeptídeos/metabolismo , Tendões/citologia , Tendões/metabolismo , Transativadores/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Drosophila/citologia , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células Epidérmicas , Epiderme/embriologia , Epiderme/metabolismo , Epistasia Genética , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Microscopia Eletrônica , Atividade Motora , Músculos/citologia , Músculos/embriologia , Músculos/metabolismo , Neuropeptídeos/genética , Especificidade de Órgãos , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Tendões/embriologia , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
20.
Gene ; 301(1-2): 53-60, 2002 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-12490323

RESUMO

We have cloned and sequenced cDNAs coding for the complete primary structure of HtH2, the second hemocyanin isoform of the marine gastropod Haliotis tuberculata. The deduced protein sequence comprises 3399 amino acids, corresponding to a molecular mass of 392 kDa. It shares only 66% of structural identity with the previously analysed first isoform HtH1, and according to a molecular clock, the two isoforms of Haliotis hemocyanin separated ca. 320 million years ago. By genomic polymerase chain reaction and 5' race, we have also sequenced the complete gene of HtH2 (18,598 bp), except of the 5' region in front of the secreted protein. It encompasses 15 exons and 14 introns and shows several microsatellite-rich regions. It mirrors the modular structure of the encoded hemocyanin subunit, with a linear arrangement of eight different functional units separated and bordered by seven phase 1 'linker introns'. In addition, within regions encoding three of the functional units, the HtH2 gene contains six 'internal introns'. Comparison to previously sequenced genes of Octopus dofleini hemocyanin and Haliotis hemocyanin isoform (HtH1) suggests Precambrian and Palaeocoic hot spot of intron gains, followed by 320 million years of absolute stasis.


Assuntos
Hemocianinas/genética , Íntrons/genética , Moluscos/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA/química , DNA/genética , DNA Complementar/química , DNA Complementar/genética , Evolução Molecular , Éxons , Genes/genética , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...